Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 416
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Pathog ; 16(4): e1008509, 2020 04.
Article in English | MEDLINE | ID: mdl-32302362

ABSTRACT

Zika virus (ZIKV) is a unique flavivirus with high tropism to the testes. ZIKV can persist in human semen for months and can cause testicular damage in male mice. However, the mechanisms through which ZIKV enters the testes remain unclear. In this study, we revealed that matrix metalloproteinase 9 (MMP9) was upregulated by ZIKV infection in cell culture and in A129 mice. Furthermore, using an in vitro Sertoli cell barrier model and MMP9-/- mice, we found that ZIKV infection directly affected the permeability of the blood-testis barrier (BTB), and knockout or inhibition of MMP9 reduced the effects of ZIKV on the Sertoli cell BTB, highlighting its role in ZIKV-induced disruption of the BTB. Interestingly, the protein levels of MMP9 were elevated by ZIKV nonstructural protein 1 (NS1) in primary mouse Sertoli cells (mSCs) and other cell lines. Moreover, the interaction between NS1 and MMP9 induced the K63-linked polyubiquitination of MMP9, which enhanced the stability of MMP9. The upregulated MMP9 level led to the degradation of essential proteins involved in the maintenance of the BTB, such as tight junction proteins (TJPs) and type Ⅳ collagens. Collectively, we concluded that ZIKV infection promoted the expression of MMP9 which was further stabilized by NS1 induced K63-linked polyubiquitination to affect the TJPs/ type Ⅳ collagen network, thereby disrupting the BTB and facilitating ZIKV entry into the testes.


Subject(s)
Blood-Testis Barrier/metabolism , Blood-Testis Barrier/virology , Matrix Metalloproteinase 9/metabolism , Testis/virology , Zika Virus Infection/metabolism , Zika Virus/physiology , A549 Cells , Animals , Blood-Testis Barrier/enzymology , Collagen Type IV/metabolism , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , Semen/metabolism , Semen/virology , Sertoli Cells/enzymology , Sertoli Cells/metabolism , Sertoli Cells/virology , Spermatogenesis , Testis/blood supply , Testis/metabolism , Tight Junction Proteins/metabolism , Up-Regulation , Viral Nonstructural Proteins/metabolism , Virus Internalization , Zika Virus Infection/enzymology , Zika Virus Infection/virology
2.
Biochem Biophys Res Commun ; 525(4): 1011-1017, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32178872

ABSTRACT

In seminiferous epithelium, tight junctions (TJs) between adjacent Sertoli cells constitute the blood-testis barrier and must change synchronically for germ cells to translocate from the basal to the adluminal compartment during the spermatogenic cycle. Rho GTPase activation through stimulation with specific L-selectin ligands has been proposed to modulate tight junctional dynamics. However, little is known regarding the role of Ca+2 dynamics in Sertoli cell and how Ca+2 relays L-selectin signals to modulate Rho GTPase activity in Sertoli cells, thus prompting us to investigate the Ca+2 flux induced by L-selectin ligand in ASC-17D cells. Using fluorescent real-time image, we first demonstrated the increase of intracellular Ca+2 level following L-selectin ligand stimulation. This Ca+2 increase was inhibited in ASC-17D cells pretreated with nifedipine, the L-type voltage-operated Ca+2 channel (VOCC) blocker, but not mibefradil, the T-type VOCC blocker. We then demonstrated the up-regulation of Rho and Rac1 in ASC-17D cells following the administration of L-selectin ligand, and the pre-treatment with nifedipine, but not mibefradil, prior to L-selectin ligand-binding abolished the activation of both Rho and Rac1. Together, we conclude that the activation of L-selectin induces Ca+2 influx through the L-type VOCC, which up-regulates Rho and Rac1 proteins, in ASC-17D cells.


Subject(s)
Calcium/metabolism , L-Selectin/metabolism , Sertoli Cells/metabolism , Spermatozoa/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels , Cell Line , Ligands , Male , Mibefradil/pharmacology , Nifedipine/pharmacology , Optical Imaging , Rats , Sertoli Cells/drug effects , Sertoli Cells/enzymology , Signal Transduction/drug effects , Signal Transduction/physiology , Spermatogenesis/drug effects , Spermatogenesis/genetics , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/genetics
3.
Andrologia ; 52(6): e13654, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32436229

ABSTRACT

The pandemic caused by Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to several hypotheses of functional alteration of different organs. The direct influence of this virus on the male urogenital organs is still to be evaluated. However some hypotheses can already be made, especially in the andrological field, for the biological similarity of the SARS-CoV and SARS-CoV2. As well as SARS-CoV, SARS CoV-2 uses the 'Angiotensin Converting Enzyme-2' (ACE2) as a receptor to enter human cells. It was found that ACE2, Angiotensin (1-7) and its MAS receptors are present, over in the lung, also in the testicles, in particular in Leydig and Sertoli cells. A first hypothesis is that the virus could enter the testicle and lead to alterations in testicular functionality. A second hypothesis is that the binding of the virus to the ACE2 receptor, could cause an excess of ACE2 and give rise to a typical inflammatory response. The inflammatory cells could interfere with the function of Leydig and Sertoli cells. Both hypotheses should be evaluated and confirmed, in order to possibly monitor fertility in patients COVID-19+.


Subject(s)
Betacoronavirus , Coronavirus Infections/complications , Infertility, Male/virology , Pneumonia, Viral/complications , Angiotensin-Converting Enzyme 2 , Betacoronavirus/metabolism , COVID-19 , Coronavirus Infections/physiopathology , Humans , Inflammation/virology , Leydig Cells/enzymology , Leydig Cells/virology , Male , Pandemics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/physiopathology , SARS-CoV-2 , Sertoli Cells/enzymology , Sertoli Cells/virology , Testis/enzymology , Testis/virology
4.
Reproduction ; 157(3): R85-R94, 2019 03.
Article in English | MEDLINE | ID: mdl-30608903

ABSTRACT

SRC family kinases (SFKs) are known regulators of multiple cellular events, including cell movement, differentiation, proliferation, survival and apoptosis. SFKs are expressed virtually by all mammalian cells. They are non-receptor protein kinases that phosphorylate a variety of cellular proteins on tyrosine, leading to the activation of protein targets in response to environmental stimuli. Among SFKs, SRC, YES and FYN are the ubiquitously expressed and best studied members. In fact, SRC, the prototypical SFK, was the first tyrosine kinase identified in mammalian cells. Studies have shown that SFKs are regulators of cell junctions, and function in endocytosis and membrane trafficking to regulate junction restructuring events. Herein, we briefly summarize the recent findings in the field regarding the role of SFKs in the testis in regulating spermatogenesis, particularly in Sertoli-Sertoli and Sertoli-germ cell adhesion. While it is almost 50 years since the identification of the oncogene v-Src encoded by Rous sarcoma transforming virus, the understanding of SFK involvement during spermatogenesis in the testis remains far behind that in other epithelia and tissues. The goal of this review is to bridge this gap.


Subject(s)
Cell Adhesion , Cell Differentiation , Germ Cells/cytology , Sertoli Cells/cytology , Spermatogenesis , src-Family Kinases/metabolism , Animals , Germ Cells/enzymology , Humans , Male , Sertoli Cells/enzymology
5.
Mol Reprod Dev ; 86(8): 984-998, 2019 08.
Article in English | MEDLINE | ID: mdl-31134714

ABSTRACT

Sertoli cells are a type of nurse cell in the seminiferous epithelium that are crucial for sustaining spermatogenesis by extending nutritional and energy support to the developing germ cells. Dysfunction of Sertoli cells could cause disordered spermatogenesis and reduced fertility in males. In this study, we focused on the expression and function of palmitoyl protein thioesterase 1 (PPT1), a lysosomal depalmitoylating enzyme, in Sertoli cells. Here, we show that PPT1 expression in Sertoli cells is responsive to cholesterol treatment and that specific knockout of Ppt1 in Sertoli cells causes male subfertility associated with poor sperm quality and a high ratio of sperm deformity. Specifically, Ppt1 deficiency leads to poor cell variably accompanied with abnormal lysosome accumulation and increased cholesterol levels in Sertoli cells. Further, Ppt1 deficiency results in poor adhesion of developing germ cells to Sertoli cells in the seminiferous epithelium, which is likely to be responsible for the reduced male fertility as a consequence of declines in sperm count and motility as well as a high incidence of sperm head deformity. In summary, PPT1 affects sperm quality and male fertility through regulating lysosomal function and cholesterol metabolism in Sertoli cells.


Subject(s)
Cholesterol/metabolism , Fertility , Gene Expression Regulation, Enzymologic , Sertoli Cells/enzymology , Spermatozoa/enzymology , Thiolester Hydrolases/biosynthesis , Animals , Male , Mice , Seminiferous Tubules/cytology , Seminiferous Tubules/enzymology , Sertoli Cells/cytology , Sperm Count , Spermatozoa/cytology
6.
FASEB J ; 32(6): 3229-3241, 2018 06.
Article in English | MEDLINE | ID: mdl-29401623

ABSTRACT

The pituitary gonadotrophins and testosterone are the main hormonal regulators of spermatogenesis, but estradiol is also known to play a role in the process. The hormonal responses in the testis are partially mediated by somatic Sertoli cells that provide nutritional and physical support for differentiating male germ cells. Hydroxysteroid (17ß) dehydrogenase 1 (HSD17B1) is a steroidogenic enzyme that especially catalyzes the conversion of low potent 17keto-steroids to highly potent 17ß-hydroxysteroids. In this study, we show that Hsd17b1 is highly expressed in Sertoli cells of fetal and newborn mice, and HSD17B1 knockout males present with disrupted spermatogenesis with major defects, particularly in the head shape of elongating spermatids. The cell-cell junctions between Sertoli cells and germ cells were disrupted in the HSD17B1 knockout mice. This resulted in complications in the orientation of elongating spermatids in the seminiferous epithelium, reduced sperm production, and morphologically abnormal spermatozoa. We also showed that the Sertoli cell-expressed HSD17B1 participates in testicular steroid synthesis, evidenced by a compensatory up-regulation of HSD17B3 in Leydig cells. These results revealed a novel role for HSD17B1 in the control of spermatogenesis and male fertility, and that Sertoli cells significantly contribute to steroid synthesis in the testis.-Hakkarainen, J., Zhang, F.-P., Jokela, H., Mayerhofer, A., Behr, R., Cisneros-Montalvo, S., Nurmio, M., Toppari, J., Ohlsson, C., Kotaja, N., Sipilä, P., Poutanen, M. Hydroxysteroid (17ß) dehydrogenase 1 expressed by Sertoli cells contributes to steroid synthesis and is required for male fertility.


Subject(s)
17-Hydroxysteroid Dehydrogenases/biosynthesis , Fertility/physiology , Gene Expression Regulation, Enzymologic/physiology , Sertoli Cells/enzymology , Spermatogenesis/physiology , Steroids/biosynthesis , 17-Hydroxysteroid Dehydrogenases/genetics , Animals , Male , Mice , Mice, Knockout , Seminiferous Epithelium/cytology , Seminiferous Epithelium/enzymology , Sertoli Cells/cytology , Spermatids/cytology , Spermatids/enzymology
7.
Semin Cell Dev Biol ; 30: 65-74, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24727349

ABSTRACT

Non-receptor protein tyrosine kinases are cytoplasmic kinases that activate proteins by phosphorylating tyrosine residues, which in turn affect multiple functions in eukaryotic cells. Herein, we focus on the role of non-receptor protein tyrosine kinases, most notably, FAK, c-Yes and c-Src, in the transport of spermatids across the seminiferous epithelium during spermatogenesis. Since spermatids, which are formed from spermatocytes via meiosis, are immotile haploid cells, they must be transported by Sertoli cells across the seminiferous epithelium during the epithelial cycle of spermatogenesis. Without the timely transport of spermatids across the epithelium, the release of sperms at spermiation fails to occur, leading to infertility. Thus, the molecular event pertinent to spermatid transport is crucial to spermatogenesis. We provide a critical discussion based on recent findings in this review. We also provide a hypothetical model on spermatid transport, and the role of non-receptor protein tyrosine kinases in this event. We also highlight areas of research that deserve attention by investigators in the field.


Subject(s)
Protein-Tyrosine Kinases/physiology , Sperm Transport , Spermatids/enzymology , Spermatogenesis , Animals , Blood-Testis Barrier/cytology , Blood-Testis Barrier/physiology , Humans , Male , Phosphorylation , Protein Processing, Post-Translational , Seminiferous Epithelium/cytology , Sertoli Cells/enzymology , Signal Transduction , Spermatids/physiology
8.
J Cell Biochem ; 117(11): 2597-607, 2016 11.
Article in English | MEDLINE | ID: mdl-27017955

ABSTRACT

Sertoli cell metabolism actively maintains the nutritional needs of germ cells. It has been described that after glucose incorporation in Sertoli cells, less than 1% is converted to glycogen suggesting low levels of glycogen synthase activity. Phosphorylation of muscle glycogen synthase (MGS) at serine 640 (pS640MGS) decreases its activity, and this form of the enzyme was discovered as a non-ribosomal protein that modulates the translation of a subset of transcripts in HeLa cells. The aim of our study was to functionally characterize MGS in cultured Sertoli cells, as well as to explore this new feature related to RNA molecules. We detected MGS in the cytoplasm of Sertoli cells as well as in the nuclei. The activity rates of the enzyme were extremely low indicating that MGS is expressed but almost inactive. Protein targeting to glycogen (PTG) overexpression was performed to activate MGS by dephosphorylation. PTG induced glycogen synthesis massively, confirming that this enzyme is present but inactive. This finding correlates with high levels of pS640MGS, which were assayed by phosphatase treatment. To explore a putative new function for MGS in Sertoli cells, we performed RNA immunoprecipitation coupled to microarray studies. The results revealed that MGS co-immunoprecipitated with the several mRNAs and also rRNAs. These findings indicate that MGS is expressed Sertoli cells but in an inactive form, and also support a possibly novel feature of this metabolic enzyme associated with RNA-related molecules. J. Cell. Biochem. 117: 2597-2607, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Glycogen Synthase/metabolism , Glycogen/biosynthesis , Muscle, Skeletal/enzymology , RNA/metabolism , Sertoli Cells/enzymology , Animals , Blotting, Western , Cells, Cultured , Fluorescent Antibody Technique , Glucose/metabolism , Immunoprecipitation , Male , Mice , Mice, Inbred C57BL
9.
Histochem Cell Biol ; 146(6): 737-748, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27688058

ABSTRACT

Within the human testis, large amounts of sulfated steroid hormones are produced. As shown in breast tissue and placenta, these might not only be excretion intermediates, but re-activated in target cells by steroid sulfatase (STS). This process is called sulfatase pathway and may play a pivotal role in para- and/or intracrine regulation by creating a local supply for steroid hormones. This requires a facilitated transport via uptake carriers and efflux transporters as these hydrophilic molecules cannot pass the cell membrane. Moreover, blood-testis barrier formation in the testis requires a transport through Sertoli cells (SCs) to reach germ cells (GCs). Sertoli cells are therefore expected to play a key role as gate-keepers for sulfatase pathway in human seminiferous epithelium. We analyzed the mRNA and protein expression of uptake carriers and efflux transporters like organic anion-transporting polypeptides (OATP2B1, OATP3A1) and multidrug resistance-related proteins (MRP1, MRP4) in testicular tissue and cultured Sertoli cells (FS1, HSEC). Additionally, expression pattern of STS as well as sulfonating enzymes (SULTs) were assessed. OATP2B1, OATP3A1 and STS were detected in SCs as well as GCs, whereas MRP1 is only expressed in SCs, and SULT1E1 only in Leydig cells, respectively. By transcellular transport of [H3]DHEAS in HSEC, we showed a functional transport of sulfated steroids in vitro. Our data indicate that steroid synthesis via sulfatase pathway in Sertoli cells in vivo and in vitro is possible and may contribute to paracrine and intracrine regulation employing the local supply of sulfated and free steroid hormones inside seminiferous tubules.


Subject(s)
Sertoli Cells/enzymology , Sulfatases/metabolism , Testis/enzymology , Cells, Cultured , Humans , Male , Sertoli Cells/cytology , Sertoli Cells/metabolism , Steroids/biosynthesis , Testis/metabolism
10.
Cell Biol Int ; 39(6): 770-4, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25639863

ABSTRACT

4-Methylcatechol (4-MC) is one of the metabolites of quercetin, which is a potential drug for neuroprotection and tumorigenesis inhibition. This study was performed to investigate the cytotoxic effect of 4-MC in mouse TM4 Sertoli cells. TM4 Sertoli cell viability was significantly inhibited by 4-MC in a time- and dose-dependent manner. The number of apoptotic and dead cells was significantly increased after 4-MC treatment. Caspase 3 activity increased by prolonged exposure of TM4 Sertoli cells to 200 µM 4-MC. The 4-MC significantly upregulated the mRNA level of Bax gene and considerably downregulated the Bcl-2 gene expression in a concentration-dependent manner. Results showed that 4-MC could induce TM4 Sertoli cell apoptosis, and the cytotoxic effect of 4-MC on TM4 Sertoli cells may be associated with upregulated Bax gene expression, which induced caspase cascade activation.


Subject(s)
Catechols/pharmacology , Sertoli Cells/cytology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Caspase 3/metabolism , Cell Count , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Gene Expression Regulation/drug effects , Male , Mice , Real-Time Polymerase Chain Reaction , Sertoli Cells/drug effects , Sertoli Cells/enzymology
11.
Stem Cells ; 31(11): 2517-27, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23897718

ABSTRACT

Coordination of stem cell fate is regulated by extrinsic niche signals and stem cell intrinsic factors. In mammalian testes, spermatogonial stem cells maintain constant production of abundant spermatozoa by alternating between self-renewal and differentiation at regular intervals according to a periodical program known as the seminiferous epithelial cycle. Although retinoic acid (RA) signaling has been suggested to direct the cyclical differentiation of spermatogonial stem cells, it remains largely unclear how their cycle-dependent self-renewal/proliferation is regulated. Here, we show that MEK/ERK signaling contributes to the cyclical activity of spermatogonial stem cells. We found that ERK1/2 is periodically activated in Sertoli cells during the stem cell self-renewal/proliferation phase, and that MEK/ERK signaling is required for the stage-related expression of the critical niche factor GDNF. In addition, ERK1/2 is activated in GFRα1-positive spermatogonial stem cells under the control of GDNF and prevent them from being differentiated. These results suggest that MEK/ERK signaling directly and indirectly maintains spermatogonial stem cells by mediating a signal that promotes their periodical self-renewal/proliferation. Conversely, RA signaling directly and indirectly induces differentiation of spermatogonial stem cells. We propose that temporally regulated activations of RA signaling and a signal regulating MEK/ERK antagonistically coordinates the cycle-related activity of spermatogonial stem cells.


Subject(s)
MAP Kinase Signaling System/physiology , Spermatogonia/cytology , Spermatogonia/enzymology , Stem Cells/cytology , Stem Cells/enzymology , Animals , Cell Differentiation/physiology , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sertoli Cells/cytology , Sertoli Cells/enzymology
12.
J Appl Toxicol ; 34(6): 628-36, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23677851

ABSTRACT

Nonylphenol (NP), a representative endocrine disruptor, interferes with reproductive function in aquatic organisms and animals. Although many previous studies have focused on apoptotic cell death by NP, the fundamental mechanism of NP on apoptosis remains poorly understood. Here, we investigated the molecular mechanism on NP-induced apoptotic cell death in mouse TM4 Sertoli cells. To evaluate NP treatment on cell viability, formazan and lactate dehydrogenase (LDH) assays were performed. Results indicate that NP reduced cell viability and increased the release of LDH in dose- and time-dependent manners. The reduction of cell viability by NP treatment appeared to involve necrosis as well as apoptosis based on nuclear fragmentation, an increase in the sub G1 population, and the detection of poly(ADP ribose) polymerase and caspase-3 cleavage. Additionally, the anti-apoptotic protein Bcl-2 diminished, whereas the pro-apoptotic protein Bax increased in a time-dependent manner. Note that NP-induced apoptotic cell death was enhanced by the generation of reactive oxygen species (ROS) and activation of extracellular signal-regulated kinase (ERK) signaling. Pretreatment with N-acetylcysteine, an antioxidant, attenuated NP-induced apoptotic cell death. Moreover, NP caused a transient activation of the MAPK pathway. In particular, NP-induced cell death was significantly suppressed by U0126, a specific inhibitor of ERK. Taken together, our results suggest that NP induces apoptosis in mouse TM4 Sertoli cells via ROS generation and ERK activation.


Subject(s)
Apoptosis/drug effects , Endocrine Disruptors/toxicity , Extracellular Signal-Regulated MAP Kinases/metabolism , Oxidative Stress/drug effects , Phenols/toxicity , Reactive Oxygen Species/metabolism , Sertoli Cells/drug effects , Signal Transduction/drug effects , Animals , Antioxidants/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Male , Mice , Protein Kinase Inhibitors/pharmacology , Sertoli Cells/enzymology , Sertoli Cells/pathology , Time Factors
13.
Andrologia ; 46(1): 9-16, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23134192

ABSTRACT

Icariin (ICA), a major constituent of flavonoids from the Chinese medical herb Epimedium brevicornum Maxim, is found to be protective for male reproductive ability, with the underlying mechanism largely unknown. Our study here investigated the effects of ICA on Sertoli cells, which act as nurse cells for the germ cells developing. Icariin was found to stimulate Sertoli cell proliferation in a dose-dependent manner. Further study revealed that Icariin induced an obvious phosphorylation of ERK in Sertoli cells. Inhibition of activation of ERK by the ERK inhibitor U0126 nearly blocked the Icariin-induced proliferation of Sertoli cells. Taken together, our results suggest that Icariin promotes the proliferation of Sertoli cells in vitro by activating the ERK1/2 signal pathway, which might at least partially, explain the protective role of Icariin on male reproductive ability.


Subject(s)
Cell Proliferation/drug effects , Flavonoids/pharmacology , MAP Kinase Signaling System , Sertoli Cells/drug effects , Animals , Cells, Cultured , Male , Rats , Rats, Sprague-Dawley , Sertoli Cells/cytology , Sertoli Cells/enzymology
14.
Biochim Biophys Acta ; 1820(2): 84-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22146232

ABSTRACT

BACKGROUND: Sertoli cells metabolize glucose producing lactate for developing germ cells. As insulin regulates glucose uptake and its disturbance/insensitivity is associated with diabetes mellitus, we aimed to determine the effect of insulin deprivation in human Sertoli cell (hSC) metabolism and metabolism-associated gene expression. METHODS: hSC-enriched primary cultures were maintained in the absence/presence of insulin and metabolite variations were determined by (1)H-NMR. mRNA expression levels of glucose transporters (GLUT1, GLUT3), lactate dehydrogenase (LDHA) and monocarboxylate transporter (MCT4) were determined by RT-PCR. RESULTS: Insulin deprivation resulted in decreased lactate production and in decrease of glucose consumption that was completely reverted after 6h. Cells of both groups consumed similar amounts of glucose. In insulin-deprived cells, transcript levels of genes associated to lactate metabolism (LDHA and MCT4) were decreased. Transcript levels of genes involved in glucose uptake exhibited a divergent variation: GLUT3 levels were decreased while GLUT1 levels increased. Insulin-deprived hSCs presented: 1) altered glucose consumption and lactate secretion; 2) altered expression of metabolism-associated genes involved in lactate production and export; 3) an adaptation of glucose uptake by modulating the expression of GLUT1 and GLUT3. GENERAL SIGNIFICANCE: This is the first report regarding the effect of insulin-deprivation on hSC metabolism.


Subject(s)
Gene Expression Regulation , Insulin/deficiency , Sertoli Cells/metabolism , Alanine/biosynthesis , Cells, Cultured , Gene Expression Regulation/drug effects , Glucose/metabolism , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Humans , Insulin/pharmacology , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Lactic Acid/biosynthesis , Male , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Pyruvates/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sertoli Cells/enzymology
15.
Biochem Biophys Res Commun ; 441(1): 120-5, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24129193

ABSTRACT

The function of protease during male meiosis has not been well studied. We previously cloned and characterized four testis-specific serine proteases in the mouse testis. One of the proteases, Prss41/Tessp-1, was expressed in the germ and Sertoli cell. This time, to examine the involvement of Prss41/Tessp-1 in spermatogenesis, we conducted the organ culture of testis fragments in the presence of the anti-Prss41/Tessp-1 antibody. Because in the Sertoli cell, the Prss41/Tessp-1 protein was mostly associated with the membrane of intracellular organelles by glycosylphosphatidylinositol, the antibody was expected to affect Prss41/Tessp-1 at the plasma membrane of spermatogonia. By adding the antibody, the number of germ cells was decreased in some seminiferous tubules. The marker genes expression strongly suggested that meiosis was arrested at spermatogonia, and the number of apoptotic germ cells increased by terminal deoxynucleotidyl transferase dUTP nick end labeling assay. These data indicated that Prss41/Tessp-1 was necessary for the progression of meiosis at the stage of spermatogonia during in vitro spermatogenesis. Together with our previous study, the current results suggest that the Prss/Tessp proteases are important for the progression of meiosis at each stage.


Subject(s)
Meiosis , Serine Endopeptidases/metabolism , Spermatogenesis , Testis/cytology , Testis/enzymology , Animals , Antibodies/metabolism , Apoptosis , Male , Mice , Mice, Inbred C57BL , Models, Biological , Organ Culture Techniques , Organ Specificity , Protein Transport , Sertoli Cells/cytology , Sertoli Cells/enzymology , Spermatogonia/cytology , Spermatogonia/enzymology , Subcellular Fractions/enzymology
16.
Biol Reprod ; 88(3): 59, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23325809

ABSTRACT

The blood-testis barrier (BTB) is a large junctional complex composed of tight junctions, adherens junctions, and gap junctions between adjacent Sertoli cells in the seminiferous tubules of the testis. Maintenance of the BTB as well as the controlled disruption and reformation of the barrier is essential for spermatogenesis and male fertility. Tyrosine phosphorylation of BTB proteins is known to regulate the integrity of adherens and tight junctions found at the BTB. SHP2 is a nonreceptor protein tyrosine phosphatase (PTP) and a key regulator of growth factor-mediated tyrosine kinase signaling pathways. We found that SHP2 is localized to Sertoli-Sertoli cell junctions in rat testis. The overexpression of a constitutive active SHP2 mutant, SHP2 Q79R, up-regulated the BTB disruptor ERK1/2 via Src kinase in primary rat Sertoli cells in culture. Furthermore, focal adhesion kinase (FAK), which also supports BTB integrity, was found to interact with SHP2 and constitutive activation of SHP2 decreased FAK tyrosine phosphorylation. Expression of the SHP2 Q79R mutant in primary cultured Sertoli cells also resulted in the loss of tight junction and adherens junction integrity that corresponded with the disruption of the actin cytoskeleton and mislocalization of adherens junction and tight junction proteins N-cadherin, ß-catenin, and ZO-1 away from the plasma membrane. These results suggest that SHP2 is a key regulator of BTB integrity and Sertoli cell support of spermatogenesis and fertility.


Subject(s)
Blood-Testis Barrier/metabolism , Intercellular Junctions/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Sertoli Cells/enzymology , Animals , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Hepatocyte Growth Factor/metabolism , Male , Mitogen-Activated Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley
17.
Cell Tissue Res ; 354(3): 861-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24057877

ABSTRACT

Sertoli cells (SCs) glucose metabolism is crucial for spermatogenesis since developing germ cells consume lactate produced by SCs as their main energy source. Recently, androgens and estrogens have been implicated in SCs energy metabolism modulation, although the molecular mechanisms remained undisclosed. Here, we report the effect of sex steroid hormones on key points of cultured rat SCs glycolytic pathway. We used primary cultures of immature rat SCs treated with 17ß-estradiol (E2) or 5α-dihydrotestosterone (DHT). The transcript levels of glucose transporters (GLUTs), phosphofructokinase 1 (PFK-1) and lactate dehydrogenase C (LDH C) were analyzed after 25 and 50 h of culture by qPCR. Protein levels of GLUTs, PFK-1, LDH and monocarboxylate transporter 4 (MCT4) after 25 and 50 h were determined by western blot and LDH activity was also assessed. Our results show that both E2 and DHT downregulated the transcript levels of PFK-1, GLUT1 and GLUT3 after 50 h. However, only DHT-treated cells presented a downregulation of LDH C transcript levels. Interestingly, the protein levels of these enzymes and transporters remained unaltered except in DHT-treated cells that presented a significant decrease on GLUT1 protein levels evidencing a possible site for the regulation of SCs glucose metabolism by androgens. Taken together, our results provide evidence that sex steroid hormones action in SCs energy metabolism is mediated through modulation in glycolysis-related transporters and enzymes, particularly at the transcriptional level. DHT decreased GLUT1 protein levels and increased LDH activity after 25 h, evidencing key points for this hormone action in the regulation of SCs metabolism.


Subject(s)
Dihydrotestosterone/pharmacology , Estradiol/pharmacology , Glucose Transport Proteins, Facilitative/metabolism , L-Lactate Dehydrogenase/metabolism , Phosphofructokinase-1/metabolism , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Animals , Energy Metabolism , Glucose/metabolism , Glucose Transport Proteins, Facilitative/biosynthesis , Glucose Transport Proteins, Facilitative/genetics , Glycolysis/drug effects , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/metabolism , L-Lactate Dehydrogenase/biosynthesis , L-Lactate Dehydrogenase/genetics , Male , Monocarboxylic Acid Transporters/biosynthesis , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Muscle Proteins/metabolism , Phosphofructokinase-1/biosynthesis , Phosphofructokinase-1/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Sertoli Cells/enzymology , Transcription, Genetic/drug effects
18.
Reproduction ; 145(3): 297-310, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23580951

ABSTRACT

The severe degenerative phenomena that characterises spermatogenesis in mating blue sharks involves spatially separated germ cell and Sertoli cell apoptosis. Unlike that observed in multilayered type B spermatogonial and spermatocyte cysts caspase-3-depend entapoptosis of single and multinucleate type B spermatogonia in one to three spermatogonial layered cysts resulted in their complete fragmentation, delayed phagocytic removal and displacement of the apoptotic bodies towards the perilumenar Sertoli nuclei. Changes were observed in the immunostaining patterns of proliferating cell nuclear antigen (PCNA), including subtle changes in cytoplasmic and overall intense immunostaining, labelled single and multinucleate cell (MNC) apoptotic spermatogonial masses in premeiotic cysts indifferent stages of the protracted death process. Initial massive MNC formation at the mitosis­meiosis transition eventually left its imprint in the spermatogenic sequence in the form of vacuolated areas in the affected and subsequent stages. Some of the latter attempted further developmental advance but eventually degenerated. The observed higher PCNA index of spermatogonia in vacuolated testes compared to testes with the MNC type of degeneration indicated that the former testicular morphology represented, in essence, the recovery phase from the pronounced MNC death earlier. Events culminating in the eventual apoptotic demise of the Sertoli cells themselves included the abortion of further development (presumably due to a suboptimal Sertoli: germ cell ratio) of those germ cells left over from the wave of MNC death that swept the cysts. Eventually the Sertoli-cell-only cysts became apoptotic as they were engulfed by the infiltrating lymphomyeloid cells from the epigonal organ associated with the mature pole of the testis.


Subject(s)
Apoptosis , Caspase 3/analysis , Fish Proteins/analysis , Sharks/metabolism , Spermatogenesis , Testis/enzymology , Testis/pathology , Animals , Male , Meiosis , Mitosis , Phagocytosis , Proliferating Cell Nuclear Antigen/analysis , Sertoli Cells/enzymology , Sertoli Cells/pathology , Spermatogonia/enzymology , Spermatogonia/pathology
19.
Reproduction ; 146(5): 471-80, 2013.
Article in English | MEDLINE | ID: mdl-23962454

ABSTRACT

Male germ cell differentiation entails the synthesis and remodeling of membrane polar lipids and the formation of triacylglycerols (TAGs). This requires fatty acid-binding proteins (FABPs) for intracellular fatty acid traffic, a diacylglycerol acyltransferase (DGAT) to catalyze the final step of TAG biosynthesis, and a TAG storage mode. We examined the expression of genes encoding five members of the FABP family and two DGAT proteins, as well as the lipid droplet protein perilipin 2 (PLIN2), during mouse testis development and in specific cells from seminiferous epithelium. Fabp5 expression was distinctive of Sertoli cells and consequently was higher in prepubertal than in adult testis. The expression of Fabp3 increased in testis during postnatal development, associated with the functional differentiation of interstitial cells, but was low in germ cells. Fabp9, together with Fabp12, was prominently expressed in the latter. Their transcripts increased from spermatocytes to spermatids and, interestingly, were highest in spermatid-derived residual bodies (RB). Both Sertoli and germ cells, which produce neutral lipids and store them in lipid droplets, expressed Plin2. Yet, while Dgat1 was detected in Sertoli cells, Dgat2 accumulated in germ cells with a similar pattern of expression as Fabp9. These results correlated with polyunsaturated fatty acid-rich TAG levels also increasing with mouse germ cell differentiation highest in RB, connecting DGAT2 with the biosynthesis of such TAGs. The age- and germ cell type-associated increases in Fabp9, Dgat2, and Plin2 levels are thus functionally related in the last stages of germ cell differentiation.


Subject(s)
Diacylglycerol O-Acyltransferase/metabolism , Fatty Acid-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Lipid Metabolism , Membrane Proteins/metabolism , Sexual Maturation , Testis/cytology , Animals , Animals, Newborn , Cells, Cultured , Diacylglycerol O-Acyltransferase/biosynthesis , Diacylglycerol O-Acyltransferase/genetics , Fatty Acid-Binding Proteins/biosynthesis , Fatty Acid-Binding Proteins/genetics , Leydig Cells/cytology , Leydig Cells/enzymology , Leydig Cells/metabolism , Lysosomes/enzymology , Lysosomes/metabolism , Male , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred Strains , Perilipin-2 , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Seminiferous Epithelium/cytology , Seminiferous Epithelium/growth & development , Seminiferous Epithelium/metabolism , Sertoli Cells/cytology , Sertoli Cells/enzymology , Sertoli Cells/metabolism , Specific Pathogen-Free Organisms , Spermatids/cytology , Spermatids/enzymology , Spermatids/metabolism , Spermatogenesis , Testis/growth & development , Testis/metabolism , Up-Regulation
20.
Urol Int ; 90(4): 449-54, 2013.
Article in English | MEDLINE | ID: mdl-23306140

ABSTRACT

Sperm-specific glyceraldehyde 3-phosphate dehydrogenase (GAPDS) originates from spermatogenic cells of the human testis. A strong staining of GAPDS was detected in epididymal epithelium, especially in principal cells and basal cells of the epithelium. GAPDS also bound to the fibrous sheet of the sperm tail and inhibited the motility and penetration ability of sperms. The rat model showed that at postnatal day 28 the spermatogenic cells began to express GAPDS protein. By day 60 its expression decreased in spermatogenic cells while it increased in Sertoli cells. After sexual maturation (120 days) GAPDS protein was expressed in both Sertoli cells and elongated sperms. The expression of GAPDS gradually increased with age in the epididymis.


Subject(s)
Epididymis/enzymology , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Spermatozoa/enzymology , Testis/enzymology , Age Factors , Animals , Cricetinae , Female , Humans , Immunohistochemistry , Male , Mesocricetus , Rats , Sertoli Cells/enzymology , Sperm Motility , Sperm Tail/enzymology , Sperm-Ovum Interactions
SELECTION OF CITATIONS
SEARCH DETAIL