Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 4.428
1.
Front Endocrinol (Lausanne) ; 15: 1357594, 2024.
Article En | MEDLINE | ID: mdl-38699384

In mammals, gonadal somatic cell lineage differentiation determines the development of the bipotential gonad into either the ovary or testis. Sertoli cells, the only somatic cells in the spermatogenic tubules, support spermatogenesis during gonadal development. During embryonic Sertoli cell lineage differentiation, relevant genes, including WT1, GATA4, SRY, SOX9, AMH, PTGDS, SF1, and DMRT1, are expressed at specific times and in specific locations to ensure the correct differentiation of the embryo toward the male phenotype. The dysregulated development of Sertoli cells leads to gonadal malformations and male fertility disorders. Nevertheless, the molecular pathways underlying the embryonic origin of Sertoli cells remain elusive. By reviewing recent advances in research on embryonic Sertoli cell genesis and its key regulators, this review provides novel insights into sex determination in male mammals as well as the molecular mechanisms underlying the genealogical differentiation of Sertoli cells in the male reproductive ridge.


Cell Differentiation , Cell Lineage , Sertoli Cells , Sertoli Cells/cytology , Sertoli Cells/metabolism , Sertoli Cells/physiology , Male , Humans , Animals , Reproduction/physiology , Spermatogenesis/physiology , Sex Determination Processes/physiology
2.
Front Endocrinol (Lausanne) ; 15: 1349000, 2024.
Article En | MEDLINE | ID: mdl-38689732

Recent advancements in reproductive medicine have guided novel strategies for addressing male infertility, particularly in cases of non-obstructive azoospermia (NOA). Two prominent invasive interventions, namely testicular sperm extraction (TESE) and microdissection TESE (micro-TESE), have emerged as key techniques to retrieve gametes for assisted reproduction technologies (ART). Both heterogeneity and complexity of NOA pose a multifaceted challenge to clinicians, as the invasiveness of these procedures and their unpredictable success underscore the need for more precise guidance. Seminal plasma can be aptly regarded as a liquid biopsy of the male reproductive tract, encompassing secretions from the testes, epididymides, seminal vesicles, bulbourethral glands, and prostate. This fluid harbors a variety of cell-free nucleic acids, microvesicles, proteins, and metabolites intricately linked to gonadal activity. However, despite numerous investigations exploring potential biomarkers from seminal fluid, their widespread inclusion into the clinical practice remains limited. This could be partially due to the complex interplay of diverse clinical and genetic factors inherent to NOA that likely contributes to the absence of definitive biomarkers for residual spermatogenesis. It is conceivable that the integration of clinical data with biomarkers could increase the potential in predicting surgical procedure outcomes and their choice in NOA cases. This comprehensive review addresses the challenge of sperm retrieval in NOA through non-invasive biomarkers. Moreover, we delve into promising perspectives, elucidating innovative approaches grounded in multi-omics methodologies, including genomics, transcriptomics and proteomics. These cutting-edge techniques, combined with the clinical and genetics features of patients, could improve the use of biomarkers in personalized medical approaches, patient counseling, and the decision-making continuum. Finally, Artificial intelligence (AI) holds significant potential in the realm of combining biomarkers and clinical data, also in the context of identifying non-invasive biomarkers for sperm retrieval.


Azoospermia , Biomarkers , Sperm Retrieval , Humans , Male , Azoospermia/metabolism , Azoospermia/diagnosis , Biomarkers/metabolism , Biomarkers/analysis , Infertility, Male/metabolism , Infertility, Male/diagnosis , Infertility, Male/therapy , Semen/metabolism , Spermatogenesis/physiology
3.
Reprod Fertil Dev ; 362024 May.
Article En | MEDLINE | ID: mdl-38739740

Context A maternal high-fat diet is thought to pose a risk to spermatogenesis in the progeny. Aims We tested whether a maternal high-fat diet would affect Sertoli cell expression of transcription factors (insulin-like growth factor I (IGF-I); glial-cell line-derived neurotrophic factor (GDNF); Ets variant 5 (ETV5)) and cell proliferation and apoptotic proteins, in the testis of adult offspring. Methods Pregnant rats were fed ad libitum with a standard diet (Control) or a high-fat diet (HFat) throughout pregnancy and lactation. After weaning, male pups were fed the standard diet until postnatal day 160. Males were monitored daily from postnatal day 34 to determine onset of puberty. On postnatal day 160, their testes were processed for morphometry and immunohistochemistry. Key results The HFat diet increased seminiferous-tubule diameter (P P P P P P P P Conclusions A maternal high-fat diet alters the balance between spermatogonia proliferation and spermatid apoptosis. Implications A maternal high-fat diet seems to 'program' adult male fertility.


Apoptosis , Cell Proliferation , Diet, High-Fat , Lactation , Maternal Nutritional Physiological Phenomena , Prenatal Exposure Delayed Effects , Testis , Animals , Female , Male , Pregnancy , Apoptosis/physiology , Lactation/physiology , Testis/metabolism , Testis/pathology , Rats , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/metabolism , Maternal Nutritional Physiological Phenomena/physiology , Spermatogenesis/physiology , Sertoli Cells/metabolism , Sertoli Cells/pathology , Insulin-Like Growth Factor I/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Rats, Wistar
4.
Front Endocrinol (Lausanne) ; 15: 1327800, 2024.
Article En | MEDLINE | ID: mdl-38654926

Introduction: Azoospermia, characterized by an absence of sperm in the ejaculate, represents the most severe form of male infertility. While surgical sperm retrieval in obstructive azoospermia (OA) is successful in the majority of cases, patients with non-obstructive azoospermia (NOA) show retrieval rates of only about 50% and thus frequently have unnecessary surgery. Surgical intervention could be avoided if patients without preserved spermatogenesis are identified preoperatively. This prospective study aimed to discover biomarkers in seminal plasma that could be employed for a non-invasive differential diagnosis of OA/NOA in order to rationalize surgery recommendations and improve success rates. Methods: All patients signed written informed consent, underwent comprehensive andrological evaluation, received human genetics to exclude relevant pathologies, and patients with azoospermia underwent surgical sperm retrieval. Using label-free LC-MS/MS, we compared the proteomes of seminal plasma samples from fertile men (healthy controls (HC), n=8) and infertile men diagnosed with 1) OA (n=7), 2) NOA with successful sperm retrieval (mixed testicular atrophy (MTA), n=8), and 3) NOA without sperm retrieval (Sertoli cell-only phenotype (SCO), n=7). Relative abundance changes of two candidate markers of sperm retrieval, HSPA2 and LDHC, were confirmed by Western Blot. Results: We found the protein expression levels of 42 proteins to be significantly down-regulated (p ≤ 0.05) in seminal plasma from SCO NOA patients relative to HC whereas only one protein was down-regulated in seminal plasma from MTA patients. Analysis of tissue and cell expression suggested that the testis-specific proteins LDHC, PGK2, DPEP3, and germ-cell enriched heat-shock proteins HSPA2 and HSPA4L are promising biomarkers of spermatogenic function. Western blotting revealed a significantly lower abundance of LDHC and HSPA2 in the seminal plasma of men with NOA (SCO and MTA) compared to controls. Discussion: The results indicate that certain testis-specific proteins when measured in seminal plasma, could serve as indicators of the presence of sperm in the testis and predict the success of sperm retrieval. Used in conjunction with conventional clinical assessments, these proteomic biomarkers may assist in the non-invasive diagnosis of idiopathic male infertility.


Azoospermia , Biomarkers , Proteomics , Semen , Humans , Male , Azoospermia/metabolism , Azoospermia/diagnosis , Semen/metabolism , Semen/chemistry , Biomarkers/metabolism , Biomarkers/analysis , Biomarkers/blood , Adult , Proteomics/methods , Prospective Studies , Sperm Retrieval , Case-Control Studies , Spermatogenesis/physiology
5.
Cell Tissue Res ; 396(2): 157-175, 2024 May.
Article En | MEDLINE | ID: mdl-38564020

The blood-testis barrier (BTB) is formed adjacent to the seminiferous basement membrane. It is a distinct ultrastructure, partitioning testicular seminiferous epithelium into apical (adluminal) and basal compartments. It plays a vital role in developing and maturing spermatocytes into spermatozoa via reorganizing its structure. This enables the transportation of preleptotene spermatocytes across the BTB, from basal to adluminal compartments in the seminiferous tubules. Several bioactive peptides and biomolecules secreted by testicular cells regulate the BTB function and support spermatogenesis. These peptides activate various downstream signaling proteins and can also be the target themself, which could improve the diffusion of drugs across the BTB. The gap junction (GJ) and its coexisting junctions at the BTB maintain the immunological barrier integrity and can be the "gateway" during spermatocyte transition. These junctions are the possible route for toxicant entry, causing male reproductive dysfunction. Herein, we summarize the detailed mechanism of all the regulators playing an essential role in the maintenance of the BTB, which will help researchers to understand and find targets for drug delivery inside the testis.


Blood-Testis Barrier , Sertoli Cells , Male , Blood-Testis Barrier/metabolism , Sertoli Cells/metabolism , Sertoli Cells/cytology , Humans , Animals , Intercellular Junctions/metabolism , Spermatogenesis/physiology , Gap Junctions/metabolism
6.
Reprod Biol Endocrinol ; 22(1): 36, 2024 Apr 03.
Article En | MEDLINE | ID: mdl-38570783

Microtubule-associated protein 1a (Map1a) is a microtubule (MT) regulatory protein that binds to the MT protofilaments in mammalian cells to promote MT stabilization. Maps work with MT cleavage proteins and other MT catastrophe-inducing proteins to confer MT dynamics to support changes in the Sertoli cell shape to sustain spermatogenesis. However, no functional studies are found in the literature to probe its role in spermatogenesis. Using an RNAi approach, coupled with the use of toxicant-induced testis (in vivo)- and Sertoli cell (in vitro)-injury models, RNA-Seq analysis, transcriptome profiling, and relevant bioinformatics analysis, immunofluorescence analysis, and pertinent biochemical assays for cytoskeletal organization, we have delineated the functional role of Map1a in Sertoli cells and testes. Map1a was shown to support MT structural organization, and its knockdown (KD) also perturbed the structural organization of actin, vimentin, and septin cytoskeletons as these cytoskeletons are intimately related, working in concert to support spermatogenesis. More importantly, cadmium-induced Sertoli cell injury that perturbed the MT structural organization across the cell cytoplasm was associated with disruptive changes in the distribution of Map1a and a surge in p-p38-MAPK (phosphorylated p38-mitogen-activated protein kinase) expression but not total p38-MAPK. These findings thus support the notion that p-p38-MAPK activation is involved in cadmium-induced Sertoli cell injury. This conclusion was supported by studies using doramapimod, a specific p38-MAPK phosphorylation (activation) inhibitor, which was capable of restoring the cadmium-induced disruptive structural organization of MTs across the Sertoli cell cytoplasm. In summary: this study provides mechanistic insights regarding restoration of toxicant-induced Sertoli cell and testis injury and male infertility.


Actins , Sertoli Cells , Rats , Animals , Male , Actins/metabolism , Sertoli Cells/metabolism , Cadmium , Rats, Sprague-Dawley , Blood-Testis Barrier/metabolism , Microtubules/metabolism , Testis/metabolism , Spermatogenesis/physiology , Mammals
7.
Gen Comp Endocrinol ; 353: 114529, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38643847

Germ cells are highly conserved in the gonads, nurtured to either develop into a gamete or self-renew into a stem cell reserve. Preserving the germ cell pool and protecting the reproductive organs is essential for maintaining an individual's fertility. Several factors, including a sedentary lifestyle, pollutants, hormonal disruption, drugs, and a disease condition, have been shown to impair normal reproductive function. Irisin has recently been identified as an adipomyokine involved in modulating physiological functions based on the body's metabolic status. It is being studied for its role in various functions, including fertility. Findings show the localization of irisin in various parts of the reproductive axis, with the highest levels observed during puberty and pregnancy. This raises questions about its role and function in reproduction. Studies support irisin's role in protecting against disease-induced reproductive abnormalities and infertility. Therefore, the current review focuses on how irisin influences spermatogenesis and ovarian follicular development and plays a significant role in indirectly preserving the germ cell pool by protecting the gonads against oxidative stress and inflammation.


Fibronectins , Reproduction , Humans , Fibronectins/metabolism , Animals , Female , Reproduction/physiology , Male , Spermatogenesis/physiology
8.
Spectrochim Acta A Mol Biomol Spectrosc ; 315: 124232, 2024 Jul 05.
Article En | MEDLINE | ID: mdl-38593538

The present study aims to identify spermatogenesis in testicular seminiferous tubules (ST) and testicular tissue of adult normal and busulfan-treated mice utilizing PCA and Raman spectroscopy. Raman measurements were conducted on single tubules and testes samples from adult and immature mice, comparing them with those from busulfan-treated adult mice, with validation through histological examination. The analysis revealed a higher signal variability (30 %-40 % at the peaks), prompting scrutiny of individual Raman spectra as a means of spermatogenesis measurement. However, principal component analysis (PCA) demonstrated significant cluster separation between the ST of mature and immature mice. Similar investigations were performed to compare ST from normal mature mice and those from busulfan-treated (BS-treated) mature mice, revealing substantial separation along PC1 and PC2 for all comparison sets. Additionally, comparing testicular samples from mature and immature mice revealed distinct separation in PCA. The study concludes that the combined approach of PCA and Raman spectroscopy proves to be a noninvasive and potentially valuable method for identifying spermatogenesis in seminiferous tubules and testicular samples.


Busulfan , Principal Component Analysis , Seminiferous Tubules , Spectrum Analysis, Raman , Spermatogenesis , Testis , Animals , Spectrum Analysis, Raman/methods , Male , Spermatogenesis/drug effects , Spermatogenesis/physiology , Seminiferous Tubules/drug effects , Testis/drug effects , Mice
9.
Toxicol Pathol ; 52(1): 4-12, 2024 Jan.
Article En | MEDLINE | ID: mdl-38465599

The indirect assessment of adverse effects on fertility in cynomolgus monkeys requires that tissue sections of the testis be microscopically evaluated with awareness of the stage of spermatogenesis that a particular cross-section of a seminiferous tubule is in. This difficult and subjective task could very much benefit from automation. Using digital whole slide images (WSIs) from tissue sections of testis, we have developed a deep learning model that can annotate the stage of each tubule with high sensitivity, precision, and accuracy. The model was validated on six WSI using a six-stage spermatogenic classification system. Whole slide images contained an average number of 4938 seminiferous tubule cross-sections. On average, 78% of these tubules were staged with 29% in stage I-IV, 12% in stage V-VI, 4% in stage VII, 19% in stage VIII-IX, 18% in stage X-XI, and 17% in stage XII. The deep learning model supports pathologists in conducting a stage-aware evaluation of the testis. It also allows derivation of a stage-frequency map. The diagnostic value of this stage-frequency map is still unclear, as further data on its variability and relevance need to be generated for testes with spermatogenic disturbances.


Deep Learning , Macaca fascicularis , Spermatogenesis , Testis , Animals , Male , Macaca fascicularis/anatomy & histology , Testis/anatomy & histology , Testis/pathology , Spermatogenesis/physiology , Image Processing, Computer-Assisted/methods , Seminiferous Tubules/anatomy & histology
10.
Environ Pollut ; 346: 123625, 2024 Apr 01.
Article En | MEDLINE | ID: mdl-38401636

The blood-testis barrier (BTB) plays a vital role in mammalian spermatogenesis by separating the seminiferous epithelium into an adluminal and a basal compartment. Cadmium (Cd) is a toxic heavy metal that is widely present in the environment. We observed that Cd can induce BTB disruption, leading to apoptosis of testicular cells. However, the molecular mechanisms contributing to BTB injury induced by Cd have not yet been fully clarified. Vimentin (Vim) is an important desmosome-like junction protein that mediates robust adhesion in the BTB. In this study, we investigated how Vim responds to Cd. We found that Cd treatment led to a significant decrease in Vim expression, accompanied by a marked increase in LC3-II expression and a higer number of autophagosomes. Interestingly, we also observed that Cd-induced autophagy was associated with decreased Vim activity and enhanced apoptosis of testicular cells. To further investigate the role of autophagy in Vim regulation under Cd exposure, we treated cells with an autophagy inhibitor called 3-MA. We found that 3-MA treatment enhanced Vim expression and improved the disruption of the BTB under Cd exposure. Additionally, the inhibition of Vim confirmed the role of autophagy in modulating Vim expression. These results reveal a previously unknown regulatory mechanism of Cd involving the interplay between a heavy metal and a protein.


Blood-Testis Barrier , Cadmium , Male , Animals , Cadmium/toxicity , Cadmium/metabolism , Vimentin/metabolism , Blood-Testis Barrier/metabolism , Testis/metabolism , Spermatogenesis/physiology , Autophagy , Mammals
11.
Eur J Pharmacol ; 967: 176405, 2024 Mar 15.
Article En | MEDLINE | ID: mdl-38341078

The function and regulatory mechanisms of 5-methylcytidine (m5C) in oligoasthenospermia remain unclear. In this study, we made a mouse model of oligoasthenospermia through the administration of busulfan (BUS). For the first time, we demonstrated that m5C levels decreased in oligoasthenospermia. The m5C levels were upregulated through the treatments of 5-methylcytidine. The testicular morphology and sperm concentrations were improved via upregulating m5C. The cytoskeletal regenerations of testis and sperm were accompanying with m5C treatments. m5C treatments improved T levels and reduced FSH and LH levels. The levels of ROS and MDA were significantly reduced through m5C treatments. RNA sequencing analysis showed m5C treatments increased the expression of genes involved in spermatid differentiation/development and cilium movement. Immunofluorescent staining demonstrated the regeneration of cilium and quantitative PCR (qPCR) confirmed the high expression of genes involved in spermatogenesis. Collectively, our findings suggest that the upregulation of m5C in oligoasthenospermia facilitates testicular morphology recovery and male infertility via multiple pathways, including cytoskeletal regeneration, hormonal levels, attenuating oxidative stress, spermatid differentiation/development and cilium movement. m5C may be a potential therapeutic agent for oligoasthenospermia.


Busulfan , Cytidine/analogs & derivatives , Semen , Male , Mice , Animals , Busulfan/pharmacology , Spermatogenesis/physiology , Testis
12.
Biol Reprod ; 110(5): 1000-1011, 2024 May 09.
Article En | MEDLINE | ID: mdl-38408206

The germ cell-less gene is crucial for gonad development in various organisms. Early interventions in its expression suggested a regulatory role at the mitotic stages of spermatogenesis, and its early knockout resulted in complete sterility in Drosophila. Genomic and transcriptomic data available for the catadromous giant prawn Macrobrachium rosenbergii enabled the identification of a germ cell-less homolog for this species, which we termed MroGCL (mRNA accession number OQ533056). An open reading frame containing 494 amino acids and a typical evolutionarily conserved BTB/POZ domain suggests possible protein-protein interaction functions in keeping with the Drosophila germ cell-less protein. Genomic mapping of MroGCL showed a full length of 120 896 bases. Analysis of the temporal expression of MroGCL showed constant expression in early prawn embryonic and larval stages, but a significant increase 10 days after metamorphosis when crucial sexual differentiation processes occur in prawns. In adult animals, high expression was detected in the gonads compared to the somatic tissues. RNAi-based knock-down experiments showed that both the silenced and control groups reached advanced spermatogenic stages, but that there was a significant decrease in the yield of spermatozoa in about half of the silenced animals. This finding supports our hypothesis that MroGCL is crucial for mitosis during early stage spermatogenesis. In conclusion, this study contributes to the understanding of crustacean gonad development and provides a stepping stone in the development of environmentally valuable sterile crustacean populations.


Palaemonidae , Spermatogenesis , Animals , Palaemonidae/genetics , Palaemonidae/physiology , Spermatogenesis/physiology , Spermatogenesis/genetics , Male , Amino Acid Sequence , Gene Expression Regulation, Developmental , Arthropod Proteins/genetics , Arthropod Proteins/metabolism
13.
Bioorg Chem ; 144: 107174, 2024 Mar.
Article En | MEDLINE | ID: mdl-38320369

Ursonic acid (UNA) is a natural pentacyclic triterpene found in some medicinal plants and foods. The reproductive protective effect of UNA was evaluated in a mouse model of oligozoospermia induced by busulfan (BUS) at 30 mg/kg b.w.. The mice were initially divided into groups with UNA concentrations of 10, 30, 50, 100 mg/kg. Subsequently, based on sperm parameters, the optimal concentration of 50 mg/kg was identified. As a control, an additional group was supplemented with ursolic acid at a concentration of 50 mg/kg. The results indicated that BUS caused the loss of spermatogenic cells in testis, the decrease of sperm in epididymis, the disorder of testicular cytoskeleton, the decrease of serum sex hormones such as testosterone which induced an increase in feedback of androgen receptor and other testosterone-related proteins, the increase of malondialdehyde and reactive oxygen species levels and the increase of ferroptosis in testis while UNA successfully reversed these injuries. High-throughput sequencing revealed that UNA administration significantly upregulated the expression of genes associated with spermatogenesis, such as Tnp1, Tnp2, Prm1, among others. These proteins are crucial in the histone to protamine transition during sperm chromatin remodeling. Network pharmacology analysis reveals a close association between UNA and proteins related to the transformation of histones to protamine. Molecular docking studies reveal that UNA can interact with the ferroptosis-inhibiting gene SLC7A11, thereby modulating ferroptosis. Taken together, UNA alleviated BUS-induced oligozoospermia by regulating hormone secretion, mitigating oxidative stress and promoting recovery of spermatogenesis by inhibiting the ferroptosis.


Ferroptosis , Oligospermia , Triterpenes , Humans , Male , Mice , Animals , Oligospermia/chemically induced , Oligospermia/drug therapy , Molecular Docking Simulation , Semen/metabolism , Spermatogenesis/physiology , Testosterone/pharmacology , Histones/pharmacology , Protamines/genetics , Protamines/metabolism , Protamines/pharmacology
14.
Biol Reprod ; 110(5): 877-894, 2024 May 09.
Article En | MEDLINE | ID: mdl-38236177

The centrosome is critical for maintaining the sperm head-tail connection and the formation of flagellar microtubules. In this study, we found that in mouse testes, CCDC159 (coiled-coil domain-containing protein 159) is specifically localized to the head-tail coupling apparatus (HTCA) of spermatids, a structure that ensures sperm head-tail tight conjunction. CCDC159 contains a C-terminal coiled-coil domain that functions as the centrosomal localization signal. Gene knockout (KO) of Ccdc159 in mice resulted in acephalic spermatozoa, abnormal flagella, and male infertility. To explore the mechanism behind CCDC159 regulating spermatogenesis, we identified CCDC159-binding proteins using a yeast two-hybrid screen and speculated that CCDC159 participates in HTCA assembly by regulating protein phosphatase PP1 activity. Further RNA-sequencing analyses of Ccdc159 KO testes revealed numerous genes involved in male gamete generation that were downregulated. Together, our results show that CCDC159 in spermatids is a novel centrosomal protein anchoring the sperm head to the tail. Considering the limitation of KO mouse model in clarifying the biological function of CCDC159 in spermatogenesis, a gene-rescue experiment will be performed in the future.


Mice, Knockout , Sperm Head , Sperm Tail , Spermatids , Spermatogenesis , Animals , Male , Mice , Spermatids/metabolism , Sperm Tail/metabolism , Spermatogenesis/physiology , Sperm Head/metabolism , Infertility, Male/genetics , Infertility, Male/metabolism , Testis/metabolism , Centrosome/metabolism
15.
Cell Prolif ; 57(2): e13551, 2024 Feb.
Article En | MEDLINE | ID: mdl-37743695

Busulfan is an antineoplastic, which is always accompanied with the abnormal of spermatogonia self-renewal and differentiation. It has been demonstrated that the omega-3 polyunsaturated fatty acids (PUFAs) benefits mature spermatozoa. However, whether omega-3 can protect endogenous spermatogonia and the detailed mechanisms are still unclear. Evaluate of spermatogenesis function (in vivo) were examined by histopathological analysis, immunofluorescence staining, and western blotting. The levels of lipid metabolites in testicular tissue were determined via liquid chromatography. We investigated the effect of lipid metabolites on Sertoli cells provided paracrine factors to regulate spermatogonia proliferation and differentiation using co-culture system. In our study, we showed that omega-3 PUFAs significantly improved the process of sperm production and elevated the quantity of both undifferentiated Lin28+ spermatogonia and differentiated c-kit+ spermatogonia in a mouse model where spermatogenic function was disrupted by busulfan. Mass spectrometry revealed an increase in the levels of several omega-3 metabolites in the testes of mice fed with omega-3 PUFAs. The eicosapentaenoic acid metabolite 12-hydroxyeicosapentaenoic acid (12-HEPE) up-regulated bone morphogenic protein 4 (BMP4) expression through GPR120-ERK1/2 pathway activation in Sertoli cells and restored spermatogonia proliferation and differentiation. Our study provides evidence that omega-3 PUFAs metabolite 12-HEPE effectively protects spermatogonia and reveals that GPR120 might be a tractable pharmacological target for fertility in men received chemotherapy or severe spermatogenesis dysfunction.


Busulfan , Semen , Humans , Male , Mice , Animals , Busulfan/pharmacology , Busulfan/metabolism , Spermatogenesis/physiology , Spermatogonia , Spermatozoa , Testis/metabolism
16.
Andrology ; 12(4): 899-917, 2024 May.
Article En | MEDLINE | ID: mdl-37772683

BACKGROUND: Acetaminophen and ibuprofen are widely administered to babies due to their presumed safety as over-the-counter drugs. However, no reports exist on the effects of cyclooxygenase inhibitors on undifferentiated spermatogonia and spermatogonial stem cells. Infancy represents a critical period for spermatogonial stem cell formation and disrupting spermatogonial stem cells or their precursors may be associated with infertility and testicular cancer formation. OBJECTIVES: The goal of this study was to examine the molecular and functional impact of cyclooxygenase inhibition and silencing on early steps of undifferentiated spermatogonia (u spg) and spermatogonial stem cell development, to assess the potential reproductive risk of pharmaceutical cyclooxygenase inhibitors. METHODS: The effects of cyclooxygenase inhibition were assessed using the mouse C18-4 undifferentiated juvenile spermatogonial cell line model, previously shown to include cells with spermatogonial stem cell features, by measuring prostaglandins, cell proliferation, and differentiation, using cyclooxygenase 1- and cyclooxygenase 2-selective inhibitors NS398, celecoxib, and FR122047, acetaminophen, and ibuprofen. Cyclooxygenase 1 gene silencing was achieved using a stable short-hairpin RNA approach and clone selection, then assessing gene and protein expression in RNA sequencing, quantitative real-time polymerase chain reaction, and immunofluorescence studies. RESULTS: Cyclooxygenase 2 inhibitors NS398 and celecoxib, as well as acetaminophen, but not ibuprofen, dose-dependently decreased retinoic acid-induced expression of the spg differentiation gene Stra8, while NS398 decreased the spg differentiation marker Kit, suggesting that cyclooxygenase 2 is positively associated with spg differentiation. In contrast, short-hairpin RNA-based cyclooxygenase 1 silencing in C18-4 cells altered cellular morphology and upregulated Stra8 and Kit, implying that cyclooxygenase 1 prevented spg differentiation. Furthermore, RNA sequencing analysis of cyclooxygenase 1 knockdown cells indicated the activation of several signaling pathways including the TGFb, Wnt, and Notch pathways, compared to control C18-4 cells. Notch pathway genes were upregulated by selective cyclooxygenase inhibitors, acetaminophen and ibuprofen. CONCLUSION: We report that cyclooxygenase 1 and 2 differentially regulate undifferentiated spermatogonia/spermatogonial stem cell differentiation. Cyclooxygenases regulate Notch3 expression, with the Notch pathway targeted by PGD2. These data suggest an interaction between the eicosanoid and Notch signaling pathways that may be critical for the development of spermatogonial stem cells and subsequent spermatogenesis, cautioning about using cyclooxygenase inhibitors in infants.


Nitrobenzenes , Spermatogonia , Sulfonamides , Testicular Neoplasms , Humans , Male , Animals , Mice , Spermatogonia/metabolism , Testicular Neoplasms/metabolism , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cyclooxygenase 1/pharmacology , Cyclooxygenase 2/metabolism , Celecoxib/pharmacology , Celecoxib/metabolism , Ibuprofen/pharmacology , Acetaminophen , Spermatogenesis/physiology , Cell Differentiation/physiology , Cyclooxygenase Inhibitors/pharmacology , RNA/metabolism , Testis/metabolism
17.
Biol Reprod ; 110(1): 78-89, 2024 Jan 13.
Article En | MEDLINE | ID: mdl-37776549

The kinesin light chain 3 protein (KLC3) is the only member of the kinesin light chain protein family that was identified in post-meiotic mouse male germ cells. It plays a role in the formation of the sperm midpiece through its association with both spermatid mitochondria and outer dense fibers (ODF). Previous studies showed a significant correlation between its expression level and sperm motility and quantitative semen parameters in humans, while the overexpression of a KLC3-mutant protein unable to bind ODF also affected the same traits in mice. To further assess the role of KLC3 in fertility, we used CRISPR/Cas9 genome editing in mice and investigated the phenotypes induced by the invalidation of the gene or of a functional domain of the protein. Both approaches gave similar results, i.e. no detectable change in male or female fertility. Testis histology, litter size and sperm count were not altered. Apart from the line-dependent alterations of Klc3 mRNA levels, testicular transcriptome analysis did not reveal any other changes in the genes tested. Western analysis supported the absence of KLC3 in the gonads of males homozygous for the inactivating mutation and a strong decrease in expression in males homozygous for the allele lacking one out of the five tetratricopeptide repeats. Overall, these observations raise questions about the supposedly critical role of this kinesin in reproduction, at least in mice where its gene mutation or inactivation did not translate into fertility impairment.


Kinesins , Sperm Motility , Animals , Female , Humans , Male , Mice , Fertility/genetics , Kinesins/genetics , Kinesins/metabolism , Mice, Knockout , Mutation , Proteins/metabolism , Semen , Sperm Motility/genetics , Spermatogenesis/physiology , Spermatozoa/metabolism , Testis/metabolism
18.
Vet Res Commun ; 48(1): 391-401, 2024 Feb.
Article En | MEDLINE | ID: mdl-37733150

Bovine reproduction, including male fertility traits like semen quality, are influenced by a variety of different factors like breed, nutrition, environment, and feeding management. Diet in a crucial determinant, and in this regard although corn silage is generally considered to be a favorable roughage for fattening meat type breeds, it tends to have a negative impact on semen quality. In the current study, alfalfa hay was substituted by corn silage as a roughage source in the diet of bulls to investigate its effects on the fertility of breeding bulls. A feeding trail spanning 140 days was conducted, with semen collection occurring twice a week commencing 60 days after the start of trial. Semen quality parameters, serum antioxidant indexes, sex hormone content in semen, rumen microflora, and sperm transcriptome were characterized. Feeding corn silage enhanced host antioxidant capacity, significantly decreased spermatozoal motility and increased sperm deformity rate in bulls. Furthermore, follicle-stimulating hormone (FSH) and luteinizing hormone (LH) content in semen were significantly decreased (P < 0.05), and the inhibin B (INHB) content was significantly increased (P < 0.01). Feeding corn silage led to significant changes in the diversity of rumen microbiota of cattle at the phylum and genus levels, some of which were significantly correlated with semen quality. Subsequent RNA sequencing indicated that DHH and PITHD1, two genes related to sperm and reproductive development, were differentially expressed, and enrichment analysis also identified several pathways and biological functions relevant to sperm development and reproduction. These results indicate that feeding corn silage modulates semen quality via different pathways. Firstly, corn silage metabolites likely affect the secretion of INHB through the testicular capillaries, which affects semen quality by regulating genes involved in spermatogenesis. Secondly, low lignin content in silage corn appears to reduce abundance of rumen flora that are positively correlated with semen quality. Overall, results indicate that feeding bulls corn silage as the primary source of forage could negatively impact semen quality and may not be appropriate as the primary roughage of forage for breeding bulls.


Semen Analysis , Silage , Animals , Male , Cattle , Semen Analysis/veterinary , Zea mays , Antioxidants , Plant Breeding , Seeds , Diet/veterinary , Spermatogenesis/physiology , Rumen , Dietary Fiber/metabolism
19.
J Exp Zool A Ecol Integr Physiol ; 341(1): 86-98, 2024 Jan.
Article En | MEDLINE | ID: mdl-37902254

Adropin is a versatile peptide which was discovered as a novel metabolic hormone that is involved in the regulation of lipid and glucose homeostasis. However, its possible role in the testicular function is not yet understood. The aim of our study was to explore the distribution pattern of adropin and GPR19 in various cell types and its possible role in testicular functions of adult mice. Immunohistochemical study revealed the intense immunoreactivity of adropin in the Leydig cells, while GPR19 showed intense immunoreactivity in the pachytene spermatocytes and mild immunoreactivity in Leydig cells and primary as well as secondary spermatocytes in mouse testis. Enho mRNA was also found to be expressed in the mouse testis. These findings suggested that adropin-GPR19 signaling may act in autocrine/paracrine manner to modulate testicular functions. Furthermore, to find out the direct role of adropin in the testicular function, in vitro study was performed in which testicular slices were cultured with adropin alone (10 and 100 ng/mL) and in combination with insulin (5 µg/mL). Adropin alone inhibited testicular testosterone synthesis by inhibiting the expression of P450-SCC, 3ß-HSD, and 17ß-HSD while along with insulin stimulated the testicular testosterone synthesis by increasing the expression of GPR19, IR, StAR, P450-SCC, 3ß-HSD, and 17ß-HSD. Adropin alone or in combination with insulin promoted germ cell survival and proliferation by upregulating the expression of PCNA, Bcl2, and pERK1/2. Thus, it can be concluded that adropin-GPR19 signaling promotes insulin stimulated steroidogenesis and germ cell survival as well as proliferation in the mice testes in an autocrine/paracrine manner.


Testis , Testosterone , Animals , Male , Mice , Insulin/metabolism , Leydig Cells/metabolism , Spermatogenesis/physiology , Testis/metabolism
20.
FASEB J ; 38(1): e23376, 2024 01.
Article En | MEDLINE | ID: mdl-38112167

Male germ cell development is dependent on the orchestrated regulation of gene networks. TATA-box binding protein associated factors (TAFs) facilitate interactions of TATA-binding protein with the TATA element, which is known to coordinate gene transcription during organogenesis. TAF7 like (Taf7l) is situated on the X chromosome and has been implicated in testis development. We examined the biology of TAF7L in testis development using the rat. Taf7l was prominently expressed in preleptotene to leptotene spermatocytes. To study the impact of TAF7L on the testis we generated a global loss-of-function rat model using CRISPR/Cas9 genome editing. Exon 3 of the Taf7l gene was targeted. A founder was generated possessing a 110 bp deletion within the Taf7l locus, which resulted in a frameshift and the premature appearance of a stop codon. The mutation was effectively transmitted through the germline. Deficits in TAF7L did not adversely affect pregnancy or postnatal survival. However, the Taf7l disruption resulted in male infertility due to compromised testis development and failed sperm production. Mutant germ cells suffer meiotic arrest at late zygotene/early pachynema stages, with defects in sex body formation. This testis phenotype was more pronounced than previously described for the subfertile Taf7l null mouse. We conclude that TAF7L is essential for male germ cell development in the rat.


Semen , Spermatogenesis , TATA-Binding Protein Associated Factors , Transcription Factor TFIID , Animals , Female , Male , Pregnancy , Rats , Cell Differentiation , Meiosis , Semen/metabolism , Spermatocytes/metabolism , Spermatogenesis/physiology , Spermatozoa/metabolism , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , Testis/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism
...