Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 203
Filter
Add more filters










Publication year range
1.
Adv Exp Med Biol ; 1288: 69-93, 2021.
Article in English | MEDLINE | ID: mdl-34453732

ABSTRACT

Cancer/testis (CT) antigens are proteins aberrantly overexpressed in various tumorigenic cells, but they can also be normally expressed in the mammalian germline. Most CT antigens are highly immunogenic and known to be involved in cancer cell proliferation and tumor metastasis. A recent genome-wide analysis systematically identified CT antigen expression in 19 cancer types, significantly expanding the repertoire of CT antigens by 5-fold, from over 200 to approximately 1000. However, their function and regulation in tumorigenesis remain poorly understood. The shared functional characteristics between germ cells and cancer cells, if methodically defined, offer a unique gateway to understanding the regulation of CT antigens in cancers by studying gametogenesis. Nonetheless, such studies also provide insightful information on the role of CT antigens in spermatogenesis. Herein, we analyzed publicly available next generation sequencing datasets generated from normal adult testes in rodents, primordial germ cells and cancer samples across a series of published studies and databases. Based on these analyses, we report that a subset of CT antigens belonged to the core fitness gene family. Furthermore, super-enhancers both in normal testes and various cancers controlled specific CT antigens. We found that DNA methylation of CT antigens, such as TEX101 and TAF7L, was inversely correlated with their expression in both normal primordial germ cells and various cancers, which was mediated at least partly by DNA methyltransferase1 (DNMT1). By analyzing data from a testis knockout model, we showed that TAF7L could further influence the expression of additional CT antigens, which also held true in tumors. These findings not only confirmed the previous notion that CT antigens regulate cancer dynamics, but also showed that understanding the regulation of CT antigens during gametogenesis can offer new insights for cancer research.


Subject(s)
Antigens, Neoplasm , Testis , Animals , Antigens, Neoplasm/genetics , Carcinogenesis/genetics , Germ Cells , Male , Rodentia
2.
Adv Exp Med Biol ; 1288: 241-254, 2021.
Article in English | MEDLINE | ID: mdl-34453740

ABSTRACT

Spermatogenesis is comprised of a series of cellular events that lead to the generation of haploid sperm. These events include self-renewal of spermatogonial stem cells (SSC), proliferation of spermatogonia by mitosis, differentiation of spermatogonia and spermatocytes, generation of haploid spermatids via meiosis I/II, and spermiogenesis. Spermiogenesis consists of a series of morphological events in which spermatids are being transported across the apical compartment of the seminiferous epithelium while maturing into spermatozoa, which include condensation of the genetic materials, biogenesis of acrosome, packaging of the mitocondria into the mid-piece, and elongation of the sperm tail. However, the biology of spermiation remains poorly understood. In this review, we provide in-depth analysis based on the use of bioinformatics tools and an animal model that mimics spermiation through treatment of adult rats with adjudin, a non-hormonal male contraceptive known to induce extensive germ cell exfoliation across the seminiferous epithelium, but nost notably elongating/elongated spermatids. These analyses have shed insightful information regaridng the biology of spermiation.


Subject(s)
Spermatids , Spermatogenesis , Animals , Hydrazines , Indazoles , Male , Rats , Seminiferous Epithelium , Spermatogonia
3.
Trends Pharmacol Sci ; 41(10): 690-700, 2020 10.
Article in English | MEDLINE | ID: mdl-32792159

ABSTRACT

Testicular cells produce several biologically active peptides that exert their downstream effects by activating distinct signaling proteins. These biomolecules are now known to support spermatogenesis and effectively enhance paracellular and transcellular diffusion of drugs (e.g., adjudin) across the blood-testis barrier (BTB). We briefly discuss the biomolecules that maintain the BTB: these provide new insights into how the BTB can be modulated to allow therapeutic drugs, including male contraceptives, to be transported across the BTB and more generally across blood-tissue barriers. Information gleaned by studying the BTB, as well as other blood-tissue barriers, augments our understanding of blood-tissue barriers and provides new insights into how drugs can be delivered to organs that are effectively protected by tissue barriers.


Subject(s)
Contraceptive Agents, Male , Pharmaceutical Preparations , Blood-Testis Barrier , Contraceptive Agents, Male/pharmacology , Drug Delivery Systems , Humans , Male , Spermatogenesis
4.
J Cell Physiol ; 235(9): 6127-6138, 2020 09.
Article in English | MEDLINE | ID: mdl-31975378

ABSTRACT

The blood-testis barrier (BTB) separates the seminiferous epithelium into the apical and basal compartments. The BTB has to operate timely and accurately to ensure the correct migration of germ cells, meanwhile maintaining the immunological barrier. Testin was first characterized from primary Sertoli cells, it is a secretory protein and a sensitive biomarker to monitor junctions between Sertoli and germ cells. Till now, the functions of testin on BTB dynamics and the involving mechanisms are unknown. Herein, testin acts as a regulatory protein on BTB integrity. In vitro testin knockdown by RNAi caused significant damage to the Sertoli cell barrier with no apparent changes in the protein levels of several major tight junction (TJ), adhesion junction, and gap junction proteins. Also, testin RNAi caused the diffusion of two TJ structural proteins, occludin and ZO-1, diffusing away from the Sertoli cell surface into the cytoplasm. Association and colocalization between ZO-1 and occludin were decreased after testin RNAi, examined by Co-IP and coimmunofluorescent staining, respectively. Furthermore, testin RNAi induced a dramatic disruption on the arrangement of actin filament bundles and a reduced F-actin/G-actin ratio. The actin regulatory protein ARP3 appeared at the Sertoli cell interface after testin RNAi without its protein level change, whereas overexpressing testin in Sertoli cells showed no effect on TJ barrier integrity. The above findings suggest that besides as a monitor for Sertoli-germ cell junction integrity, testin is also an essential molecule to maintain Sertoli-Sertoli junctions.


Subject(s)
Actin-Related Protein 3/genetics , Blood-Testis Barrier/metabolism , Proteins/genetics , Zonula Occludens-1 Protein/genetics , Actin Cytoskeleton/genetics , Adherens Junctions/genetics , Animals , Male , Mice , Occludin/genetics , Proteins/metabolism , Rats , Rats, Sprague-Dawley , Seminiferous Epithelium/growth & development , Seminiferous Epithelium/metabolism , Sertoli Cells/metabolism , Spermatogenesis/genetics , Tight Junctions/genetics
5.
Am J Physiol Endocrinol Metab ; 317(1): E121-E138, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31112404

ABSTRACT

Studies have shown that the mTORC1/rpS6 signaling cascade regulates Sertoli cell blood-testis barrier (BTB) dynamics. For instance, specific inhibition of mTORC1 by treating Sertoli cells with rapamycin promotes the Sertoli cell barrier, making it "tighter." However, activation of mTORC1 by overexpressing a full-length rpS6 cDNA clone (i.e., rpS6-WT, wild type) in Sertoli cells promotes BTB remodeling, making the barrier "leaky." Also, there is an increase in rpS6 and p-rpS6 (phosphorylated and activated rpS6) expression at the BTB in testes at stages VIII-IX of the epithelial cycle, and it coincides with BTB remodeling to support the transport of preleptotene spermatocytes across the barrier, illustrating that rpS6 is a BTB-modifying signaling protein. Herein, we used a constitutively active, quadruple phosphomimetic mutant of rpS6, namely p-rpS6-MT of p-rpS6-S235E/S236E/S240E/S244E, wherein Ser (S) was converted to Glu (E) at amino acid residues 235, 236, 240, and 244 from the NH2 terminus by site-directed mutagenesis, for its overexpression in rat testes in vivo using the Polyplus in vivo jet-PEI transfection reagent with high transfection efficiency. Overexpression of this p-rpS6-MT was capable of inducing BTB remodeling, making the barrier "leaky." This thus promoted the entry of the nonhormonal male contraceptive adjudin into the adluminal compartment in the seminiferous epithelium to induce germ cell exfoliation. Combined overexpression of p-rpS6-MT with a male contraceptive (e.g., adjudin) potentiated the drug bioavailability by modifying the BTB. This approach thus lowers intrinsic drug toxicity due to a reduced drug dose, further characterizing the biology of BTB transport function.


Subject(s)
Blood-Testis Barrier/metabolism , Contraceptive Agents, Male/pharmacology , Hydrazines/pharmacology , Indazoles/pharmacology , Mechanistic Target of Rapamycin Complex 1/metabolism , Ribosomal Protein S6/metabolism , Animals , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mutagenesis, Site-Directed , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6/genetics , Seminiferous Epithelium/metabolism , Sertoli Cells/metabolism , Signal Transduction/drug effects , Spermatocytes/metabolism , Spermatogenesis/drug effects
6.
Contraception ; 99(6): 350-356, 2019 06.
Article in English | MEDLINE | ID: mdl-30763581

ABSTRACT

OBJECTIVE: The bioavailability of the non-hormonal male contraceptive adjudin is low in rats due to the blood-testis barrier (BTB). This study was designed to examine if F5-peptide, an endogenously produced reversible BTB modifier, could enhance the bioavailability of adjudin to affect spermatogenesis and provide a contraceptive effect in rats while reducing systemic toxicity. STUDY DESIGN: We overexpressed F5-peptide in adult male rats (n=10 rats; with 3 or 4 rats for each of the three different experiments noted in the three regimens) by intratesticular injection of a mammalian expression vector pCI-neo (pCI-neo/F5-peptide) vs. empty vector alone (pCI-neo/Ctrl) to be followed by treatment with adjudin by oral gavage at a dose of 10 or 20 mg/kg. The status of spermatogenesis was assessed by histological analysis and dual-labeled immunofluorescence analysis on Day 16. To assess fertility, we allowed treated males (n=3-4 rats) to mate with mature female rats (n=3-4) individually, and assessed the number of pups on Days 23, 36 and 82 to assess fertility and reversibility. RESULTS: All 4 treated rats overexpressed with F5-peptide and low-dose adjudin were infertile by Day 36, and half of these rats were fertile by Day 82, illustrating reversibility. However, overexpression of F5-peptide alone (or low-dose adjudin alone) had no effects on fertility in n=3 rats. These findings were consistent with the histology data that illustrated the BTB modifier F5-peptide promoted the action of adjudin to induce germ cell exfoliation, mediated by changes in cytoskeletal organization of F-actin and microtubules across the epithelium, thereby reducing the systemic toxicity of adjudin. CONCLUSION: In this proof-of-concept study, it was shown that overexpression of the F5-peptide prior to administration of adjudin to rats at a low (and ineffective dose by itself) was found to induce reversible male infertility. IMPLICATIONS: Overexpression of F5-peptide, an endogenously produced biomolecule in the testis known to induce BTB remodeling, enhanced the contraceptive effect of adjudin in rats, supporting proof of concept studies of BTB disrupters in men.


Subject(s)
Blood-Testis Barrier/metabolism , Hydrazines/pharmacology , Indazoles/pharmacology , Microtubules/metabolism , Peptide Fragments/metabolism , Sertoli Cells/metabolism , Spermatogenesis , Animals , Blood-Testis Barrier/pathology , Female , Laminin/genetics , Laminin/metabolism , Male , Microtubules/pathology , Peptide Fragments/genetics , Proof of Concept Study , Rats , Rats, Sprague-Dawley , Sertoli Cells/pathology , Transfection
7.
Trends Mol Med ; 24(9): 781-793, 2018 09.
Article in English | MEDLINE | ID: mdl-30056046

ABSTRACT

Studies have proven that per- and polyfluoroalkyl substances are harmful to humans, most notably perfluorooctanesulfonate (PFOS). PFOS induces rapid disorganization of actin- and microtubule (MT)-based cytoskeletons in primary cultures of rodent and human Sertoli cells, perturbing Sertoli cell gap junction communication, thereby prohibiting Sertoli cells from maintaining cellular homeostasis in the seminiferous epithelium to support spermatogenesis. PFOS perturbs several signaling proteins/pathways, such as FAK and mTORC1/rpS6/Akt1/2. The use of either an activator of Akt1/2 or overexpression of a phosphomimetic (and constitutively active) mutant of FAK or connexin 43 has demonstrated that such treatment blocks PFOS-induced Sertoli cell injury by preventing actin- and MT-based cytoskeletal disorganization. These findings thus illustrate an approach to manage PFOS-induced reproductive dysfunction.


Subject(s)
Alkanesulfonic Acids/toxicity , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , Sertoli Cells/drug effects , Sertoli Cells/pathology , Animals , Gap Junctions/drug effects , Gap Junctions/metabolism , Gap Junctions/pathology , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Sertoli Cells/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
8.
Methods Mol Biol ; 1748: 245-252, 2018.
Article in English | MEDLINE | ID: mdl-29453576

ABSTRACT

The blood-testis barrier is a unique ultrastructure in the mammalian testis, located near the basement membrane of the seminiferous tubule that segregates the seminiferous epithelium into the basal and the adluminal (apical) compartment. Besides restricting paracellular and transcellular passage of biomolecules (e.g., paracrine factors, hormones), water, electrolytes, and other substances including toxicants and/or drugs to enter the adluminal compartment of the epithelium, the BTB is an important ultrastructure that supports spermatogenesis. As such, a sensitive and reliable assay to monitor its integrity in vivo is helpful for studying testis biology. This assay is based on the ability of an intact BTB to exclude the diffusion of a small molecule such as sulfo-NHS-LC-biotin (C20H29N4NaO9S2, Mr. 556.59, a water-soluble and membrane-impermeable biotinylation reagent) from the basal to the apical compartment of the seminiferous epithelium. Herein, we summarize the detailed procedures on performing the assay and to obtain semiquantitative data to assess the extent of BTB damage when compared to positive controls, such as treatment of rats with cadmium chloride (CdCl2) which is known to compromise the BTB integrity.


Subject(s)
Biotin/metabolism , Blood-Testis Barrier/physiology , Seminiferous Epithelium/metabolism , Sertoli Cells/metabolism , Tight Junctions/metabolism , Animals , Blood-Testis Barrier/drug effects , Cells, Cultured , Male , Rats , Rats, Sprague-Dawley , Seminiferous Epithelium/cytology , Sertoli Cells/cytology
9.
Methods Mol Biol ; 1748: 229-243, 2018.
Article in English | MEDLINE | ID: mdl-29453575

ABSTRACT

The blood-testis barrier (BTB) is an important ultrastructure in the testis that supports meiosis and postmeiotic spermatid development since a delay in the establishment of a functional Sertoli cell barrier during postnatal development in rats or mice by 17-20 day postpartum (dpp) would lead to a delay of the first wave of meiosis. Furthermore, irreversible disruption of the BTB by toxicants also induces infertility in rodents. Herein, we summarize recent findings that BTB dynamics (i.e., disassembly, reassembly, and stabilization) are supported by the concerted efforts of the actin- and microtubule (MT)-based cytoskeletons. We focus on the role of two actin nucleation protein complexes, namely, the Arp2/3 (actin-related protein 2/3) complex and formin 1 (or the formin 1/spire 1 complex) known to induce actin nucleation, respectively, by conferring plasticity to actin cytoskeleton. We also focus on the MT plus (+)-end tracking protein (+TIP) EB1 (end-binding protein 1) which is known to confer MT stabilization. Furthermore, we discuss in particular how the interactions of these proteins modulate BTB dynamics during spermatogenesis. These findings also yield a novel hypothetical concept regarding the molecular mechanism that modulates BTB function.


Subject(s)
Actins/metabolism , Blood-Testis Barrier/physiology , Cytoskeleton/metabolism , Microtubules/metabolism , Sertoli Cells/metabolism , Spermatogenesis , Animals , Cells, Cultured , Humans , Male , Mice , Rats , Sertoli Cells/cytology
10.
Semin Cell Dev Biol ; 81: 71-77, 2018 09.
Article in English | MEDLINE | ID: mdl-28923514

ABSTRACT

In adult mammalian testes, spermatids, most notably step 17-19 spermatids in stage IV-VIII tubules, are aligned with their heads pointing toward the basement membrane and their tails toward the tubule lumen. On the other hand, these polarized spermatids also align across the plane of seminiferous epithelium, mimicking planar cell polarity (PCP) found in other hair cells in cochlea (inner ear). This orderly alignment of developing spermatids during spermiogenesis is important to support spermatogenesis, such that the maximal number of developing spermatids can be packed and supported by a fixed population of differentiated Sertoli cells in the limited space of the seminiferous epithelium in adult testes. In this review, we provide emerging evidence to demonstrate spermatid PCP in the seminiferous epithelium to support spermatogenesis. We also review findings in the field regarding the biology of spermatid cellular polarity (e.g., head-tail polarity and apico-basal polarity) and its inter-relationship to spermatid PCP. Furthermore, we also provide a hypothetical concept on the importance of PCP proteins in endocytic vesicle-mediated protein trafficking events to support spermatogenesis through protein endocytosis and recycling.


Subject(s)
Cell Polarity/physiology , Signal Transduction/physiology , Spermatids/physiology , Spermatogenesis/physiology , Animals , Humans , Male , Sertoli Cells/cytology , Sertoli Cells/metabolism , Spermatids/cytology , Testis/cytology , Testis/metabolism
11.
Semin Cell Dev Biol ; 81: 21-32, 2018 09.
Article in English | MEDLINE | ID: mdl-28965865

ABSTRACT

Cell polarity in the adult mammalian testis refers to the polarized alignment of developing spermatids during spermiogenesis and the polarized organization of organelles (e.g., phagosomes, endocytic vesicles, Sertoli cell nuclei, Golgi apparatus) in Sertoli cells and germ cells to support spermatogenesis. Without these distinctive features of cell polarity in the seminiferous epithelium, it is not possible to support the daily production of millions of sperm in the limited space provided by the seminiferous tubules in either rodent or human males through the adulthood. In short, cell polarity provides a novel mean to align spermatids and the supporting organelles (e.g., phagosomes, Golgi apparatus, endocytic vesicles) in a highly organized fashion spatially in the seminiferous epithelium during the epithelial cycle of spermatogenesis. This is analogous to different assembling units in a manufacturing plant such that as developing spermatids move along the "assembly line" conferred by Sertoli cells, different structural/functional components can be added to (or removed from) the developing spermatids during spermiogenesis, so that functional spermatozoa are produced at the end of the assembly line. Herein, we briefly review findings regarding the regulation of cell polarity in the testis with specific emphasis on developing spermatids, supported by an intriguing network of regulatory proteins along a local functional axis. Emerging evidence has suggested that cell cytoskeletons provide the tracks which in turn confer the unique assembly lines in the seminiferous epithelium. We also provide some thought-provoking concepts based on which functional experiments can be designed in future studies.


Subject(s)
Cell Polarity , Cytoskeleton/metabolism , Sertoli Cells/metabolism , Testis/metabolism , Animals , Humans , Male , Microtubules/metabolism , Sertoli Cells/cytology , Spermatids/cytology , Spermatogenesis , Testis/cytology
12.
Sci Rep ; 7(1): 15810, 2017 Nov 17.
Article in English | MEDLINE | ID: mdl-29150642

ABSTRACT

PFOS induces Sertoli cell injury using testicular cells isolated from rodent testes, but it remains unknown if PFOS has similar effects in humans. Herein, we maintained human Sertoli cells in a mitotically active state in vitro, thus enabling transfection experiments that altered gene expression to explore the molecular mechanism(s) underlying toxicant-induced cell injury. Human Sertoli cells obtained from men at ages 15, 23, 36 and 40 were cultured in vitro. These differentiated Sertoli cells remained mitotically active when cultured in the presence of 10% FBS (fetal bovine serum), with a replication time of ~1-3 weeks. At ~80% confluency, they were used for studies including toxicant exposure, immunoblotting, immunofluorescence analysis, tight junction (TJ)-permeability assessment, and overexpression of BTB (blood-testis barrier) regulatory genes such as FAK and its phosphomimetic mutants. PFOS was found to induce Sertoli cell injury through disruptive effects on actin microfilaments and microtubule (MT) organization across the cell cytosol. As a consequence, these cytoskeletal networks failed to support cell adhesion at the BTB. Overexpression of a FAK phosphomimetic and constitutively active mutant p-FAK-Y407E in these cells was capable of rescuing the PFOS-induced injury through corrective cellular organization of cytoskeletal elements. SUMMARY: PFOS induces human Sertoli cell injury which can be rescued by overexpressing p-FAK-Y407E mutant.


Subject(s)
Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Mutation/genetics , Sertoli Cells/pathology , Actins/metabolism , Adolescent , Adult , Cells, Cultured , Epithelium/drug effects , Epithelium/metabolism , Humans , Male , Microtubules/drug effects , Microtubules/metabolism , Permeability , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Transport/drug effects , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism , Young Adult
13.
Sci Rep ; 7(1): 6950, 2017 07 26.
Article in English | MEDLINE | ID: mdl-28747671

ABSTRACT

A correction to this article has been published and is linked from the HTML version of this paper. The error has been fixed in the paper.

14.
FASEB J ; 31(8): 3587-3607, 2017 08.
Article in English | MEDLINE | ID: mdl-28487282

ABSTRACT

Spermatogenesis takes place in the epithelium of the seminiferous tubules of the testes, producing millions of spermatozoa per day in an adult male in rodents and humans. Thus, multiple cellular events that are regulated by an array of signaling molecules and pathways are tightly coordinated to support spermatogenesis. Here, we report findings of a local regulatory axis between the basement membrane (BM), the blood-testis barrier (BTB), and the apical ectoplasmic specialization (apical ES; a testis-specific, actin-rich adherens junction at the Sertoli cell-spermatid interface) to coordinate cellular events across the seminiferous epithelium during the epithelial cycle. In short, a biologically active fragment, noncollagenous 1 (NC1) domain that is derived from collagen chains in the BM, was found to modulate cell junction dynamics at the BTB and apical ES. NC1 domain from the collagen α3(IV) chain was cloned into a mammalian expression vector, pCI-neo, with and without a collagen signal peptide. We also prepared a specific Ab against the purified recombinant NC1 domain peptide. These reagents were used to examine whether overexpression of NC1 domain with high transfection efficacy would perturb spermatogenesis, in particular, spermatid adhesion (i.e., inducing apical ES degeneration) and BTB function (i.e., basal ES and tight junction disruption, making the barrier leaky), in the testis in vivo We report our findings that NC1 domain derived from collagen α3(IV) chain-a major structural component of the BM-was capable of inducing BTB remodeling, making the BTB leaky in studies in vivo Furthermore, NC1 domain peptide was transported across the epithelium via a microtubule-dependent mechanism and is capable of inducing apical ES degeneration, which leads to germ cell exfoliation from the seminiferous epithelium. Of more importance, we show that NC1 domain peptide exerted its regulatory effect by disorganizing actin microfilaments and microtubules in Sertoli cells so that they failed to support cell adhesion and transport of germ cells and organelles (e.g., residual bodies, phagosomes) across the seminiferous epithelium. This local regulatory axis between the BM, BTB, and the apical ES thus coordinates cellular events that take place across the seminiferous epithelium during the epithelial cycle of spermatogenesis.-Chen, H., Mruk, D. D., Lee, W. M., Cheng, C. Y. Regulation of spermatogenesis by a local functional axis in the testis: role of the basement membrane-derived noncollagenous 1 domain peptide.


Subject(s)
Basement Membrane/metabolism , Collagen Type IV/metabolism , Peptide Fragments/metabolism , Sertoli Cells/drug effects , Spermatogenesis/physiology , Actins/metabolism , Animals , Male , Paclitaxel , Protein Domains , Rats , Rats, Sprague-Dawley , Recombinant Proteins , Seminiferous Epithelium/metabolism , Tight Junctions
15.
Reprod Fertil Dev ; 29(5): 998-1011, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28442050

ABSTRACT

Several compounds affect male fertility by disrupting the adhesion of germ cells to Sertoli cells, which results in the release of undeveloped germ cells into the seminiferous tubule lumen that are incapable of fertilising the ovum. Indazole carboxylic acids are one class of compounds exhibiting such effects and they have been investigated as non-hormonal contraceptives for potential human use. The aims of this study were to investigate the effects of lonidamine-ethyl ester, an indazole carboxylic acid, on spermatogenesis and cell junctions, in particular, desmosomes. We found two doses of lonidamine-ethyl ester at 50mg kg-1 to disrupt Sertoli-germ cell adhesion. By light and fluorescent microscopy, pronounced changes were observed in the distribution of actin microfilaments and intermediate filaments, as well as in the localisation of plakoglobin, a protein with structural and signalling roles at the desmosome and adherens junction at the blood-testis barrier. Furthermore, immunoblotting and immunoprecipitation experiments using testis lysates revealed a significant upregulation (P<0.01) of plakoglobin and Tyr-phosphorylated plakoglobin. Co-immunoprecipitation experiments showed an increase in the interaction between plakoglobin and fyn proto-oncogene, an Src family non-receptor tyrosine kinase, after treatment, as well as an increase in the interaction between plakoglobin and α-catenin. Taken collectively, these data indicate that a disruption of Sertoli cell and spermatocyte-spermatid adhesion in the seminiferous epithelium by lonidamine-ethyl ester results in the phosphorylation of plakoglobin, thereby promoting its interaction with α-catenin at the blood-testis barrier.


Subject(s)
Antispermatogenic Agents/pharmacology , Blood-Testis Barrier/drug effects , Cytoskeleton/drug effects , Indazoles/pharmacology , Sertoli Cells/drug effects , alpha Catenin/metabolism , gamma Catenin/metabolism , Animals , Blood-Testis Barrier/metabolism , Cytoskeleton/metabolism , Male , Phosphorylation/drug effects , Proto-Oncogene Mas , Rats , Rats, Sprague-Dawley , Seminiferous Epithelium/drug effects , Seminiferous Epithelium/metabolism , Sertoli Cells/metabolism , Testis/drug effects , Testis/metabolism
16.
Sci Rep ; 7(1): 1110, 2017 04 24.
Article in English | MEDLINE | ID: mdl-28439067

ABSTRACT

PFOS (perfluorooctanesulfonate, or perfluorooctane sulfonic acid) is an anthropogenic fluorosurfactant widely used in consumer products. While its use in Europe, Canada and the U.S. has been banned due to its human toxicity, it continues to be used in China and other developing countries as a global pollutant. Herein, using an in vitro model of Sertoli cell blood-testis barrier (BTB), PFOS was found to induce Sertoli cell injury by perturbing actin cytoskeleton through changes in the spatial expression of actin regulatory proteins. Specifically, PFOS caused mis-localization of Arp3 (actin-related protein 3, a branched actin polymerization protein) and palladin (an actin bundling protein). These disruptive changes thus led to a dis-organization of F-actin across Sertoli cell cytosol, causing truncation of actin microfilament, thereby failing to support the Sertoli cell morphology and adhesion protein complexes (e.g., occludin-ZO-1, CAR-ZO-1, and N-cadherin-ß-catenin), through a down-regulation of p-Akt1-S473 and p-Akt2-S474. The use of SC79, an Akt1/2 activator [corrected], was found to block the PFOS-induced Sertoli cell injury by rescuing the PFOS-induced F-actin dis-organization. These findings thus illustrate PFOS exerts its disruptive effects on Sertoli cell function downstream through Akt1/2. As such, PFOS-induced male reproductive dysfunction can possibly be managed through an intervention on Akt1/2 expression.


Subject(s)
Actins/drug effects , Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Microtubules/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Sertoli Cells/drug effects , Sertoli Cells/pathology , Actin-Related Protein 3/analysis , Animals , Animals, Newborn , Cells, Cultured , Cytoskeletal Proteins/analysis , Male , Phosphoproteins/analysis , Rats, Sprague-Dawley
17.
Biochim Biophys Acta Mol Cell Res ; 1864(3): 527-545, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27974247

ABSTRACT

Throughout spermatogenesis, two important processes occur at late stage VIII of the seminiferous epithelial cycle in the rat testis: preleptotene spermatocytes commence entry into the adluminal compartment and step 19 spermatids release from the seminiferous epithelium. Presently, it is not clear how these processes, which involve extensive restructuring of unique Sertoli-Sertoli and Sertoli-germ cell junctions, are mediated. We aimed to determine whether annexin A2 (ANXA2), a Ca2+-dependent and phospholipid-binding protein, participates in cell junction dynamics. To address this, in vitro and in vivo RNA interference studies were performed on prepubertal Sertoli cells and adult rat testes. The endpoints of Anxa2 knockdown were determined by immunoblotting, morphological analyses, fluorescent immunostaining, and barrier integrity assays. In the testis, ANXA2 localized to the Sertoli cell stalk, with specific staining at the blood-testis barrier and the concave (ventral) surface of elongated spermatids. ANXA2 also bound actin when testis lysates were used for immunoprecipitation. Anxa2 knockdown was found to disrupt the Sertoli cell/blood-testis barrier in vitro and in vivo. The disruption in barrier function was substantiated by changes in the localization of claudin-11, zona occludens-1, N-cadherin, and ß-catenin. Furthermore, Anxa2 knockdown resulted in spermiation defects caused by a dysfunction of tubulobulbar complexes, testis-specific actin-rich ultrastructures that internalize remnant cell junction components prior to spermiation. Additionally, there were changes in the localization of several tubulobulbar complex component proteins, including actin-related protein 3, cortactin, and dynamin I/II. Our results indicate that ANXA2 is critical for the integrity of the blood-testis barrier and the timely release of spermatids.


Subject(s)
Annexin A2/genetics , Blood-Testis Barrier/metabolism , Gene Expression Regulation, Developmental , Sertoli Cells/metabolism , Spermatids/metabolism , Spermatogenesis/genetics , Actin-Related Protein 3/genetics , Actin-Related Protein 3/metabolism , Animals , Annexin A2/antagonists & inhibitors , Annexin A2/metabolism , Blood-Testis Barrier/growth & development , Cadherins/genetics , Cadherins/metabolism , Claudins/genetics , Claudins/metabolism , Cortactin/genetics , Cortactin/metabolism , Dynamin I/genetics , Dynamin I/metabolism , Dynamin II/genetics , Dynamin II/metabolism , Intercellular Junctions/genetics , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Seminiferous Epithelium/cytology , Seminiferous Epithelium/growth & development , Seminiferous Epithelium/metabolism , Sertoli Cells/cytology , Signal Transduction , Spermatids/growth & development , Spermatids/ultrastructure , Spermatocytes/growth & development , Spermatocytes/metabolism , Spermatocytes/ultrastructure , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism , beta Catenin/genetics , beta Catenin/metabolism
18.
FASEB J ; 31(2): 584-597, 2017 02.
Article in English | MEDLINE | ID: mdl-27815338

ABSTRACT

Laminin α2 is one of the constituent components of the basement membrane (BM) in adult rat testes. Earlier studies that used a mouse genetic model have shown that a deletion of laminin α2 impedes male fertility by disrupting ectoplasmic specialization (ES; a testis-specific, actin-rich anchoring junction) function along the length of Sertoli cell in the testis. This includes ES at the Sertoli cell-elongating/elongated spermatid interface, which is known as apical ES and possibly the Sertoli-Sertoli cell interface, known as basal ES, at the blood-testis barrier (BTB). Studies have also illustrated that there is a local regulatory axis that functionally links cellular events of spermiation that occur near the luminal edge of tubule lumen at the apical ES and the basal ES/BTB remodeling near the BM at opposite ends of the seminiferous epithelium during the epithelial cycle, known as the apical ES-BTB-BM axis. However, the precise role of BM in this axis remains unknown. Here, we show that laminin α2 in the BM serves as the crucial regulator in this axis as laminin α2, likely its 80-kDa fragment from the C terminus, was found to be transported across the seminiferous epithelium at stages VIII-IX of the epithelial cycle, from the BM to the luminal edge of the tubule, possibly being used to modulate apical ES restructuring at these stages. Of more importance, a knockdown of laminin α2 in Sertoli cells was shown to induce the Sertoli cell tight junction permeability barrier disruption via changes in localization of adhesion proteins at the tight junction and basal ES at the Sertoli cell BTB. These changes were found to be mediated by a disruption of F-actin organization that was induced by changes in the spatiotemporal expression of actin binding/regulatory proteins. Furthermore, laminin α2 knockdown also perturbed microtubule (MT) organization by considerable down-regulation of MT polymerization via changes in the spatiotemporal expression of EB1 (end-binding protein 1), a +TIP (MT plus-end tracking protein). In short, laminin α2 in the BM seems to play a crucial role in the BTB-BM axis by modulating BTB dynamics during spermatogenesis.-Gao, Y., Mruk, D., Chen, H., Lui, W.-Y., Lee, W. M., Cheng, C. Y. Regulation of the blood-testis barrier by a local axis in the testis: role of laminin α2 in the basement membrane.


Subject(s)
Basement Membrane/metabolism , Blood-Testis Barrier/physiology , Gene Expression Regulation/physiology , Laminin/metabolism , Testis/physiology , Animals , Gene Knockdown Techniques , Laminin/genetics , Male , Paclitaxel/pharmacology , Rats , Rats, Sprague-Dawley , Spermatogenesis/physiology , Testis/drug effects , Tubulin Modulators/pharmacology
19.
Oncotarget ; 7(39): 64203-64220, 2016 Sep 27.
Article in English | MEDLINE | ID: mdl-27611949

ABSTRACT

During the release of sperm at spermiation, a biologically active F5-peptide, which can disrupt the Sertoli cell tight junction (TJ) permeability barrier, is produced at the site of the degenerating apical ES (ectoplasmic specialization). This peptide coordinates the events of spermiation and blood-testis barrier (BTB) remodeling at stage VIII of the epithelial cycle, creating a local apical ES-BTB axis to coordinate cellular events across the epithelium. The mechanism(s) by which F5-peptide perturbs BTB restructuring, and its involvement in apical ES dynamics remain unknown. F5-peptide, besides perturbing BTB integrity, was shown to induce germ cell release from the epithelium following its efficient in vivo overexpression in the testis. Overexpression of F5-peptide caused disorganization of actin- and microtubule (MT)-based cytoskeletons, mediated by altering the spatiotemporal expression of actin binding/regulatory proteins in the seminiferous epithelium. F5-peptide perturbed the ability of actin microfilaments and/or MTs from converting between their bundled and unbundled/defragmented configuration, thereby perturbing adhesion between spermatids and Sertoli cells. Since apical ES and basal ES/BTB are interconnected through the underlying cytoskeletal networks, this thus provides an efficient and novel mechanism to coordinate different cellular events across the epithelium during spermatogenesis through changes in the organization of actin microfilaments and MTs. These findings also illustrate the potential of F5-peptide being a male contraceptive peptide for men.


Subject(s)
Actin Cytoskeleton/metabolism , Laminin/metabolism , Microtubules/metabolism , Peptide Fragments/metabolism , Sertoli Cells/metabolism , Spermatids/metabolism , Spermatogenesis , Actin Cytoskeleton/pathology , Age Factors , Animals , Animals, Newborn , Blood-Testis Barrier/metabolism , Blood-Testis Barrier/pathology , Cell Adhesion , Cells, Cultured , Laminin/genetics , Male , Microtubules/pathology , Peptide Fragments/genetics , Rats, Sprague-Dawley , Sertoli Cells/pathology , Signal Transduction , Spermatids/pathology , Tight Junctions/metabolism , Tight Junctions/pathology , Transfection
20.
Sci Rep ; 6: 29667, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27436542

ABSTRACT

Perfluorooctanesulfonate (PFOS) is an environmental toxicant used in developing countries, including China, as a stain repellent for clothing, carpets and draperies, but it has been banned in the U.S. and Canada since the late 2000s. PFOS perturbed the Sertoli cell tight junction (TJ)-permeability barrier, causing disruption of actin microfilaments in cell cytosol, perturbing the localization of cell junction proteins (e.g., occluden-ZO-1, N-cadherin-ß-catenin). These changes destabilized Sertoli cell blood-testis barrier (BTB) integrity. These findings suggest that human exposure to PFOS might induce BTB dysfunction and infertility. Interestingly, PFOS-induced Sertoli cell injury associated with a down-regulation of the gap junction (GJ) protein connexin43 (Cx43). We next investigated if overexpression of Cx43 in Sertoli cells could rescue the PFOS-induced cell injury. Indeed, overexpression of Cx43 in Sertoli cells with an established TJ-barrier blocked the disruption in PFOS-induced GJ-intercellular communication, resulting in the re-organization of actin microfilaments, which rendered them similar to those in control cells. Furthermore, cell adhesion proteins that utilized F-actin for attachment became properly distributed at the cell-cell interface, resealing the disrupted TJ-barrier. In summary, Cx43 is a good target that might be used to manage PFOS-induced reproductive dysfunction.


Subject(s)
Alkanesulfonic Acids/adverse effects , Connexin 43/genetics , Connexin 43/metabolism , Environmental Pollutants/adverse effects , Fluorocarbons/adverse effects , Sertoli Cells/cytology , Animals , Blood-Testis Barrier/drug effects , Cell Adhesion/drug effects , Cell Communication/drug effects , Cytosol/metabolism , Male , Rats , Rats, Sprague-Dawley , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Tight Junctions/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...