Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.988
Filter
1.
Theranostics ; 14(6): 2622-2636, 2024.
Article in English | MEDLINE | ID: mdl-38646657

ABSTRACT

Rationale: In recent years, nicotinamide adenine dinucleotide (NAD+) precursors (Npre) have been widely employed to ameliorate female reproductive problems in both humans and animal models. However, whether and how Npre plays a role in the male reproductive disorder has not been fully clarified. Methods: In the present study, a busulfan-induced non-obstructive azoospermic mouse model was used, and Npre was administered for five weeks following the drug injection, with the objective of reinstating spermatogenesis and fertility. Initially, we assessed the NAD+ level, germ cell types, semen parameters and sperm fertilization capability. Subsequently, testis tissues were examined through RNA sequencing analysis, ELISA, H&E, immunofluorescence, quantitative real-time PCR, and Western blotting techniques. Results: The results indicated that Npre restored normal level of NAD+ in blood and significantly alleviated the deleterious effects of busulfan (BU) on spermatogenesis, thereby partially reestablishing fertilization capacity. Transcriptome analysis, along with recovery of testicular Fe2+, GSH, NADPH, and MDA levels, impaired by BU, and the fact that Fer-1, an inhibitor of ferroptosis, restored spermatogenesis and semen parameters close to CTRL values, supported such possibility. Interestingly, the reduction in SIRT2 protein level by the specific inhibitor AGK2 attenuated the beneficial effects of Npre on spermatogenesis and ferroptosis by affecting PGC-1α and ACLY protein levels, thus suggesting how these compounds might confer spermatogenesis protection. Conclusion: Collectively, these findings indicate that NAD+ protects spermatogenesis against ferroptosis, probably through SIRT2 dependent mechanisms. This underscores the considerable potential of Npre supplementation as a feasible strategy for preserving or restoring spermatogenesis in specific conditions of male infertility and as adjuvant therapy to preserve male fertility in cancer patients receiving sterilizing treatments.


Subject(s)
Busulfan , Ferroptosis , NAD , Sirtuin 2 , Spermatogenesis , Animals , Busulfan/pharmacology , Male , Spermatogenesis/drug effects , Mice , NAD/metabolism , Ferroptosis/drug effects , Sirtuin 2/metabolism , Sirtuin 2/genetics , Disease Models, Animal , Testis/metabolism , Testis/drug effects , Azoospermia/drug therapy , Azoospermia/metabolism , Azoospermia/chemically induced
2.
J Hazard Mater ; 470: 134165, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38574660

ABSTRACT

It has been reported that N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine-quinone (6PPD-Q), a derivative of the tire antioxidant, N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine (6PPD), exhibits acute toxicity towards organisms. However, the possible reproductive toxicity of 6PPD-Q in mammals has rarely been reported. In this study, the effects of 6PPD-Q on the reproductive toxicity of C57Bl/6 male mice were assessed after exposure to 6PPD-Q for 40 days at 4 mg/kg body weight (bw). Exposure to 6PPD-Q not only led to a decrease in testosterone levels but also adversely affected semen quality and in vitro fertilization (IVF) outcomes, thereby indicating impaired male fertility resulting from 6PPD-Q exposure. Additionally, transcriptomic and metabolomic analyses revealed that 6PPD-Q elicited differential expression of genes and metabolites primarily enriched in spermatogenesis, apoptosis, arginine biosynthesis, and sphingolipid metabolism in the testes of mice. In conclusion, our study reveals the toxicity of 6PPD-Q on the reproductive capacity concerning baseline endocrine disorders, sperm quality, germ cell apoptosis, and the sphingolipid signaling pathway in mice. These findings contribute to an enhanced understanding of the health hazards posed by 6PPD-Q to mammals, thereby facilitating the development of more robust safety regulations governing the utilization and disposal of rubber products.


Subject(s)
Mice, Inbred C57BL , Spermatozoa , Testosterone , Animals , Male , Spermatozoa/drug effects , Testosterone/blood , Testis/drug effects , Testis/metabolism , Testis/pathology , Phenylenediamines/toxicity , Rubber/toxicity , Apoptosis/drug effects , Spermatogenesis/drug effects , Mice , Reproduction/drug effects , Semen Analysis
3.
Ecotoxicol Environ Saf ; 274: 116193, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38460407

ABSTRACT

Chlorocholine chloride (CCC) is a plant growth regulator used worldwide that is detectable in cereals, fruits and animal products. The health effects of CCC exposure have raised public concern. Our previous research showed that CCC exposure decreased testosterone synthesis in pubertal rats. However, little is known about whether and how pubertal CCC exposure impacts spermatogenesis. In this study, we used BALB/c mice and spermatogonia-derived GC-1 cells to examine CCC-induced spermatogenic dysfunction. In vivo, pubertal CCC exposure led to decreased testicular weight, decreased testicular germ cells and poor sperm quality. This effect worsened after cessation of CCC exposure for the next 30 days. RNA-seq and western blot analysis revealed that CCC induced aryl hydrocarbon receptor (AhR) signaling, endoplasmic reticulum stress (ERS) and ferritinophagy. Increased iron content and lipid peroxidation levels were also observed in CCC-treated testes. In vitro, it was identified that iron overload mediated by enhanced ferritinophagy occurred in CCC-treated GC-1 cells, which might be attributed to the PERK pathway in ERS. Further, for the first time, our study elucidated the involvement of AhR in CCC-induced iron overload, which aggravated testicular oxidative damage via lipid peroxidation. Considering the adverse impact of CCC exposure on rodents, supportive evidence from GC-1 cells, and the critical importance of spermatogenesis on male development, the effects of CCC on the male reproduction warrant increased attention.


Subject(s)
Acetates , Chlormequat , Iron Overload , Phenols , Spermatogenesis , Animals , Male , Mice , Rats , Chlormequat/metabolism , Chlormequat/toxicity , Iron Overload/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Seeds , Spermatogenesis/drug effects , Testis , eIF-2 Kinase/drug effects , eIF-2 Kinase/metabolism
4.
J Hazard Mater ; 470: 134126, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38554509

ABSTRACT

Cadmium (Cd) is a well-known testis toxicant. The blood-testis barrier (BTB) is a crucial component of the testis. Cd can disrupt the integrity of the BTB and reproductive function. However, the mechanism of Cd-induced disruption of BTB and testicular damage has not been fully elucidated. Here, our study investigates the effects of Cd on BTB integrity and testicular dysfunction. 80 (aged 1 day) Hy-Line white variety chickens were randomly designed into 4 groups and treated for 90 days, as follows: control group (essential diet), 35 Cd, 70 Cd and 140 Cd groups (35, 70 and 140 mg/kg Cd). The results found that Cd exposure diminished volume of the testes and induced histopathological lesions in the testes. Exposure to Cd induced an inflammatory response, disrupted the structure and function of the FAK/occludin/ZO-1 protein complex and disrupted the tight junction and adherens junction in the BTB. In addition, Cd exposure reduced the expression of steroid-related proteins and inhibited testosterone synthesis. Taken together, these data elucidate that Cd disrupts the integrity of the BTB and further inhibits spermatogenesis by dissociating the FAK/occludin/ZO-1 complex, which provides a basis for further investigation into the mechanisms of Cd-induced impairment of male reproductive function and pharmacological protection.


Subject(s)
Blood-Testis Barrier , Cadmium , Chickens , Testis , Testosterone , Zonula Occludens-1 Protein , Animals , Male , Blood-Testis Barrier/drug effects , Cadmium/toxicity , Testis/drug effects , Testis/metabolism , Testis/pathology , Zonula Occludens-1 Protein/metabolism , Testosterone/blood , Occludin/metabolism , Focal Adhesion Kinase 1/metabolism , Spermatogenesis/drug effects
5.
Ultrastruct Pathol ; 48(3): 192-212, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38420954

ABSTRACT

This study was done to estimate the testicular histological alterations induced by Busulfan (BUS) and compare the possible protective effects of melatonin (MT) and platelet rich plasma (PRP) in a rat model. Sixty-four male rats were dispersed into: control group, BUS group, melatonin group, and PRP group. Blood samples were processed for biochemical analysis. Tissue specimens were managed for light and electron microscopic studies. Immunohistochemical expression of vimentin and proliferating cell nuclear antigen (PCNA) was performed. Busulfan induced severe testicular damage in all studied methodologies. It showed a statistically significant decrease in serum testosterone and elevation of MDA when compared to the control group. Abnormal testicular cytostructures suggesting defective spermatogenesis were observed: distorted seminiferous tubules, deformed spermatogenic cells, low germinal epithelium height, few mature spermatozoa, and also deformed barrier. Vimentin and PCNA expressions were reduced. Ultrastructurally, Sertoli cells and the blood testis barrier were deformed, spermatogenic cells were affected, and mature spermatozoa were few and showed abnormal structure. Both melatonin and PRP induced improvement in all the previous parameters and restoration of spermatogenesis as confirmed by improvement of Johnsen's score from 2.6 ± .74 to 7.6 ± .92. In conclusion, melatonin and PRP have equal potential to ameliorate the testicular toxicity of BUS. Melatonin can provide a better noninvasive way to combat BUS induced testicular injury.


Subject(s)
Busulfan , Melatonin , Platelet-Rich Plasma , Testis , Animals , Male , Busulfan/toxicity , Busulfan/pharmacology , Melatonin/pharmacology , Testis/drug effects , Testis/pathology , Testis/ultrastructure , Rats , Immunohistochemistry , Spermatogenesis/drug effects , Rats, Wistar , Antioxidants/pharmacology , Testicular Diseases/chemically induced , Testicular Diseases/pathology , Testicular Diseases/prevention & control
6.
Int. j. morphol ; 41(5): 1492-1500, oct. 2023. ilus, tab
Article in English | LILACS | ID: biblio-1521050

ABSTRACT

SUMMARY: The aim of the present work was to study the closer effect of clomiphene citrate on the ultrastructure of the testis of adult albino rats to provide a basis for optimizing this drug in the treatment of male infertility. The testes were removed from both groups under anesthesia and then prepared for examination by light using hematoxylin and eosin stains and a transmission electron microscope. Semithin sections were cut into 1 µm thick sections, stained with toluidine blue, and examined by light microscopy for a survey. The desired areas were placed in the center, and other areas were trimmed. Primary spermatocytes showed marked nuclear changes (pyknosis), and their nuclear membranes were ill-defined and disrupted. The cytoplasm showed widespread degeneration of mitochondria and lysosomes and focal degeneration of the rough endoplasmic reticulum compared with the control group. The spermatids were pale, and the two phases of spermatogenesis were distinctly identifiable in the control group but were confused in the treated group. Some spermatids had interrupted nuclear membranes, also containing degenerated mitochondria, focal fragmentation of rough endoplasmic reticulum, and free ribosomes. Spermatozoa in the treated group appeared deformed compared to the control, where they had deformed head caps. Leydig cells of the treated group have an irregularly shaped nucleus, with focal chromatin aggregation and peripheral chromatin condensation on the inner surface of the nuclear membrane. The observations of the present work indicate a possible causal relationship between testicular affection and ingestion of clomiphene citrate, which can be avoided by close medical observations using ultrasonography, semen analysis, or testicular biopsy to detect early malignant changes. Furthermore, the drug should not be used for more than three to six cycles and should be stopped for at least three cycles before reuse. When clomiphene citrate is ineffective in the treatment of male infertility, human menopausal gonadotropin (hMG) administration is typically selected. However, high-dose hMG therapy is associated with a variety of adverse effects. In this work, we report the success of a modified clomiphene citrate regimen in increasing sperm count without any hazards to the testicular tissue.


El objetivo del trabajo fue estudiar el efecto del citrato de clomifeno sobre la estructura de los testículos de la rata albina adulta, con la finalidad de determinar la mejor manera de utilizar este fármaco en el tratamiento de la infertilidad masculina. Los testículos se extrajeron bajo anestesia y para su análisis a través de microscopio de luz se tiñeron con HE. Además, las muestras fueron preparadas para su examen con microscopía electrónica de transmisión. Por otra parte, se cortaron secciones semifinas de 1 µm de espesor, se tiñeron con azul de toluidina y se examinaron mediante microscopía óptica. Los espermatocitos primarios mostraron cambios nucleares marcados (picnosis) y sus membranas nucleares estaban mal definidas y alteradas. En el grupo experimental las células presentaban el citoplasma con degeneración generalizada de las mitocondrias y de los lisosomas y una degeneración focal del retículo endoplásmico rugoso en comparación con el grupo control. Las espermátidas estaban pálidas y las dos fases de la espermatogénesis eran claramente identificables en el grupo control, pero se confundían en el grupo tratado. Algunas espermátidas tenían membranas nucleares interrumpidas, y también contenían mitocondrias degeneradas, fragmentación focal del retículo endoplásmico rugoso y ribosomas libres. Los espermatozoides del grupo tratado se presentaban deformados en comparación con el control. Las células de Leydig del grupo tratado presentaban un núcleo de forma irregular, con agregación focal de cromatina y condensación de cromatina periférica en la superficie interna de la membrana nuclear. Las observaciones del presente trabajo indican una posible relación causal entre la afección testicular y la ingestión de citrato de clomifeno, que puede evitarse mediante observaciones médicas minuciosas a través de ecografía, análisis de semen o biopsia testicular para detectar cambios malignos tempranos. Además, el medicamento no debiera ser usado durante más de tres a seis ciclos y debe suspenderse durante al menos tres ciclos antes de volver a usarlo. Cuando el citrato de clomifeno es ineficaz en el tratamiento de la infertilidad masculina, normalmente se selecciona la administración de gonadotropina menopáusica humana (hMG). Sin embargo, la terapia con hMG en dosis altas se asocia con una variedad de efectos adversos. En este trabajo, informamos el éxito de un régimen modificado con citrato de clomifeno para aumentar el recuento de espermatozoides sin riesgo para el tejido testicular.


Subject(s)
Animals , Male , Rats , Testis/drug effects , Clomiphene/pharmacology , Spermatogenesis/drug effects , Testis/ultrastructure , Microscopy, Electron
7.
Biomed Pharmacother ; 165: 115130, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37413898

ABSTRACT

As a common metabolic disorder, hyperglycemia (HG) affects and disrupts the physiology of various systems in the body. Transplantation of mesenchymal stem cells (MSCs) has been used to control the complications of disease. Most of the therapeutic properties of MSCs are attributed to their secretome. This study aimed to investigate the effects of conditioned media extracted from sole or caffeine pre-treated bone-marrow-derived MSCs on hyperglycemia-induced detrimental impact on some aspects of reproduction. The HG was induced by intraperitoneally injection of streptozotocin (65 mg/kg) and nicotinamide (110 mg/kg). Twenty-four male Wistar rats (190 ± 20 g) were divided into control, HG, and the hyperglycemic groups receiving conditioned media of proliferated MSCs solely (CM) or MSCs pre-treated with caffeine (CCM). During the 49-day treatment, body weight and blood glucose were measured weekly. Finally, HbA1c, spermatogenesis development, sperm count, morphology, viability, motility, chromatin condensation, and DNA integrity were examined. Also, testicular total antioxidant capacity (TAC), malondialdehyde, sperm fertilization potential, and pre-implantation embryo development were evaluated. A one-way ANOVA and Tukey's post-hoc tests were used to analyze the quantitative data. The p < 0.05 was considered statistically significant. The CM and with a higher efficiency, the CCM remarkably (p < 0.05) improved body weight and HG-suppressed spermatogenesis, enhanced sperm parameters, chromatin condensation, DNA integrity, and TAC, reduced HbA1c, sperm abnormalities, and malondialdehyde, and significantly improved pre-implantation embryo development versus HG group. The conditioned media of MSCs solely (CM) and more effectively after pre-treatment of MSCs with caffeine (CCM) could improve spermatogenesis development, sperm quality, pre-implantation embryo development, and testicular global antioxidant potential during hyperglycemia.


Subject(s)
Caffeine , Culture Media, Conditioned , Fertilization , Hyperglycemia , Mesenchymal Stem Cells , Spermatogenesis , Caffeine/pharmacology , Mesenchymal Stem Cells/drug effects , Culture Media, Conditioned/pharmacology , Hyperglycemia/physiopathology , Fertilization/drug effects , Male , Animals , Rats , Disease Models, Animal , Body Weight/drug effects , Blood Glucose/drug effects , Glycated Hemoglobin , Spermatogenesis/drug effects , Sperm Count , Testis/drug effects , Testis/metabolism , Spermatozoa/abnormalities , Spermatozoa/drug effects
8.
J Steroid Biochem Mol Biol ; 224: 106178, 2022 11.
Article in English | MEDLINE | ID: mdl-36108814

ABSTRACT

The expression of apelin system has been shown in the adult testis of rat and mice. It has also been emphasized that regulation of testicular activity in early stages is important to sustain normal testicular activity in adulthood. Since the expression of apelin receptor (APJ) has been shown in the adult testis, moreover, developmental expression of APJ and its role has not been explored yet. Thus, we have examined the testicular expression of APJ during postnatal stages with special reference to proliferation, apoptosis and hormone secretion in early postnatal stage. Postnatal analysis showed that circulating apelin was lowest at PND1 and maximum at PND42. Among testosterone, estrogen and androstenedione, only circulating testosterone showed a gradual increase from PND1 to PND42. Testicular expression of APJ was also developmenatly regulated from PND1 to PND42, revealing a positive correlation with circulating apelin, testosterone, and androstenedione. Immunohistochemical study showed that APJ was mainly confined to Leydig cells of early postnatal stages, whereas, seminiferous tubules at PND42 showed immunostaining in the round spermatids. APJ inhibition from PND14-PND20 by ML221 suppressed the testicular proliferation, increased apoptosis and increased estrogen secretion. However, expression of AR was down-regulated by ML221 treatment. Furthermore, ML221 decreased the abundance of p-Akt. In vitro study also showed that APJ antagonist, ML221 decreased AR expression. These results suggests that apelin signaling during early developmental stages might be required to stimulate the germ cell proliferation, and inhibition of apoptosis. Both in vivo and in vitro study have shown that expression of AR was regulated by apelin signaling. Since the first wave spermatogenesis involves proliferation and apoptosis, therefore, further study would be required to unravel the exact mechanism of apelin mediated regulation of testicular activity during early postnatal stages. In conclusion, the present results are an indicative of apelin mediated signaling during early postnatal stage for regulation of germ cell proliferation, apoptosis and AR expression.


Subject(s)
Apelin Receptors , Apelin , Sexual Development , Spermatogenesis , Testis , Animals , Male , Mice , Androstenedione/blood , Apelin/blood , Apelin/metabolism , Apelin Receptors/metabolism , Carrier Proteins , Estrogens , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Testis/drug effects , Testis/metabolism , Testosterone/blood , Testosterone/metabolism , Sexual Development/drug effects , Sexual Development/genetics , Spermatogenesis/drug effects , Spermatogenesis/genetics
9.
Toxicol In Vitro ; 83: 105397, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35643342

ABSTRACT

In response to the EU cosmetics directive regulation and REACH legislation which encourage cell culture methods in order to reduce or replace the use of animals in toxicology studies, we settled the culture of prepubertal domestic cat seminiferous tubules in our validated BioAlter® model, usually used with prepubertal rat, called here BioAlter®-rat, by opposition to BioAlter®-cat settled here. We carried out a comparative study on the effects of 3 testicular toxicants, 1,3-dinitrobenzene at 60 µM, 2-methoxyacetic acid at 2.5 mM and carbendazim at 50 nM or 500 nM in both BioAlter®-cat and BioAlter®-rat over a 3-week culture period. Sertoli cell or each germ cell populations as well as the levels of Sertoli cell or germ cell specific mRNAs were studied. The harmful effects of the 3 toxicants on pre-meiotic, meiotic and post-meiotic cell numbers and on Sertoli or germ cell specific mRNAs were clearly observed in the two species, even if there might be some small differences in the intensity of the effects on some of the studied parameters. Hence, BioAlter®-cat might be a solution to the requirements of the EU cosmetics directive and REACH legislation for male reproductive toxicology studies.


Subject(s)
Seminiferous Tubules , Spermatogenesis , Acetates/toxicity , Animals , Benzimidazoles/toxicity , Carbamates/toxicity , Cats , Dinitrobenzenes/toxicity , Male , Rats , Seminiferous Tubules/drug effects , Sertoli Cells/drug effects , Spermatogenesis/drug effects , Testis/drug effects
10.
Reprod Biol Endocrinol ; 20(1): 43, 2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35236366

ABSTRACT

The heavy metal cadmium is proposed to be one of the environmental endocrine disruptors of spermatogenesis. Cadmium-induced inhibition of spermatogenesis is associated with a hormone secretion disorder. Letrozole is an aromatase inhibitor that increases peripheral androgen levels and stimulates spermatogenesis. However, the potential protective effects of letrozole on cadmium-induced reproductive toxicity remain to be elucidated. In this study, male mice were administered CdCl2 (4 mg/kg BW) orally by gavage alone or in combination with letrozole (0.25 mg/kg BW) for 30 days. Cd exposure caused a significant decreases in body weight, sperm count, motility, vitality, and plasma testosterone levels. Histopathological changes revealed extensive vacuolization and decreased spermatozoa in the lumen. However, in the Cd + letrozole group, letrozole treatment compensated for deficits in sperm parameters (count, motility, and vitality) induced by Cd. Letrozole treatment significantly increased serum testosterone levels, which were reduced by Cd. Histopathological studies revealed a systematic array of all germ cells, a preserved basement membrane and relatively less vacuolization. For a mechanistic examination, RNA-seq was used to profile alterations in gene expression in response to letrozole. Compared with that in the Cd-treated group, RNA-Seq analysis showed that 214 genes were differentially expressed in the presence of letrozole. Gene ontology (GO) enrichment analysis and KEGG signaling pathway analysis showed that steroid biosynthetic processes were the processes most affected by letrozole treatment. Furthermore, we found that the expression of the testosterone synthesis-related genes LHCGR (luteinizing hormone/choriogonadotropin receptor) and Hsd3b6 (3 beta- and steroid delta-isomerase 6) was significantly downregulated in Cd-treated testes, but these genes maintained similar expression levels in letrozole-treated testes as those in the control group. However, the transcription levels of inflammatory cytokines, such as IL-1ß and IL-6, and oxidative stress-related genes (Nrf2, Nqo1, and Ho-1) showed no changes. The present study suggests that the potential protective effect of letrozole on Cd-induced reproductive toxicity might be mediated by the upregulation of LHCGR and Hsd3b6, which would beneficially increase testosterone synthesis to achieve optimum protection of sperm quality and spermatogenesis.


Subject(s)
Cadmium , Letrozole , Spermatogenesis , Testosterone , Animals , Male , Mice , Cadmium/toxicity , Cytoprotection/drug effects , Cytoprotection/genetics , Letrozole/pharmacology , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Mice, Inbred ICR , Protective Agents/pharmacology , Receptors, LH/drug effects , Receptors, LH/genetics , Receptors, LH/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Spermatogenesis/drug effects , Spermatogenesis/genetics , Spermatozoa/drug effects , Spermatozoa/metabolism , Steroid Isomerases/drug effects , Steroid Isomerases/genetics , Steroid Isomerases/metabolism , Testis/drug effects , Testis/metabolism , Testosterone/biosynthesis
11.
Comput Math Methods Med ; 2022: 7174399, 2022.
Article in English | MEDLINE | ID: mdl-35242210

ABSTRACT

The testicles and sperm are extremely susceptible to inflammation and oxidative stress. Although Zhibai Dihuang Pill (ZDP) has been reported to treat various infertilities including male infertility induced by Ureaplasma urealyticum (UU) infection, its mechanism is still poorly understood. This study is aimed at clarifying the underlying mechanism of ZDP to protect against UU-infected male infertility. We found that UU-infected infertile rats exhibited weight loss, reduced food intake, and decreased sperm count and vitality. The administration of ZDP improved the general state and sperm motility of rats. In addition, UU infection led to spermatogenesis disorders, impaired secretory function and blood-testis barrier (BTB) of Sertoli cells, and elevated inflammation and oxidative stress. As expected, ZDP suppressed inflammation and oxidative stress to alleviate spermatogenesis disorders. Our research showed that ZDP could improve spermatogenesis disorders and testicular function primarily through the mitogen-activated protein kinase (MAPK) signaling pathway. ZDP exerts its anti-inflammatory and antioxidant effects via the MAPK signaling pathway, thus playing an important role in ameliorating spermatogenesis failure and testicular dysfunction.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Infertility, Male/drug therapy , Testicular Diseases/drug therapy , Ureaplasma Infections/drug therapy , Ureaplasma urealyticum , Animals , Computational Biology , Disease Models, Animal , Humans , Infertility, Male/etiology , Infertility, Male/metabolism , Inflammation Mediators/metabolism , MAP Kinase Signaling System/drug effects , Male , Oxidative Stress/drug effects , Phytotherapy , Rats , Rats, Sprague-Dawley , Spermatogenesis/drug effects , Testicular Diseases/etiology , Testicular Diseases/metabolism , Testis/drug effects , Testis/metabolism , Testis/pathology , Ureaplasma Infections/complications , Ureaplasma Infections/metabolism
12.
Reprod Biol Endocrinol ; 20(1): 45, 2022 Mar 07.
Article in English | MEDLINE | ID: mdl-35255928

ABSTRACT

Diabetes mellitus (DM), a high incidence metabolic disease, is related to the impairment of male spermatogenic function. Spermidine (SPM), one of the biogenic amines, was identified from human seminal plasma and believed to have multiple pharmacological functions. However, there exists little evidence that reported SPM's effects on moderating diabetic male spermatogenic function. Thus, the objective of this study was to investigate the SPM's protective effects on testicular spermatogenic function in streptozotocin (STZ)-induced type 1 diabetic mice. Therefore, 40 mature male C57BL/6 J mice were divided into four main groups: the control group (n = 10), the diabetic group (n = 10), the 2.5 mg/kg SPM-treated diabetic group (n = 10) and the 5 mg/kg SPM-treated diabetic group (n = 10), which was given intraperitoneally for 8 weeks. The type 1 diabetic mice model was established by a single intraperitoneal injection of STZ 120 mg/kg. The results showed that, compare to the control group, the body and testis weight, as well the number of sperm were decreased, while the rate of sperm malformation was significantly increased in STZ-induced diabetic mice. Then the testicular morphology was observed, which showed that seminiferous tubule of testis were arranged in mess, the area and diameter of which was decreased, along with downregulated anti-apoptotic factor (Bcl-2) expression, and upregulated pro-apoptotic factor (Bax) expression in the testes. Furthermore, testicular genetic expression levels of Sertoli cells (SCs) markers (WT1, GATA4 and Vimentin) detected that the pathological changes aggravated observably, such as the severity of tubule degeneration increased. Compared to the saline-treated DM mice, SPM treatment markedly improved testicular function, with an increment in the body and testis weight as well as sperm count. Pro-apoptotic factor (Bax) was down-regulated expression with the up-regulated expression of Bcl-2 and suppression of apoptosis in the testes. What's more, expression of WT1, GATA4, Vimentin and the expressions of glycolytic rate-limiting enzyme genes (HK2, PKM2, LDHA) in diabetic testes were also upregulated by SPM supplement. The evidence derived from this study indicated that the SMP's positive effect on moderating spermatogenic disorder in T1DM mice's testis. This positive effect is delivered via promoting spermatogenic cell proliferation and participating in the glycolytic pathway's activation.


Subject(s)
Diabetes Mellitus, Experimental , Glycolysis/drug effects , Infertility, Male , Spermatogenesis/drug effects , Spermidine/pharmacology , Animals , Diabetes Complications/drug therapy , Diabetes Complications/metabolism , Diabetes Complications/pathology , Diabetes Complications/physiopathology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Infertility, Male/drug therapy , Infertility, Male/etiology , Infertility, Male/metabolism , Male , Mice , Mice, Inbred C57BL , Semen Analysis , Spermatogenesis/physiology , Spermidine/therapeutic use , Streptozocin , Testis/drug effects , Testis/metabolism
13.
Pharm Biol ; 60(1): 404-416, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35175170

ABSTRACT

CONTEXT: Cordycepin (COR), from Cordyceps militaris L., (Cordycipitaceae), is a valuable agent with immense health benefits. OBJECTIVE: The protective effects of COR in ageing-associated oxidative and apoptosis events in vivo and hydrogen peroxide (H2O2)-exposed spermatogenesis gene alterations in TM3 Leydig cells was investigated. MATERIALS AND METHODS: Male Sprague-Dawley rats were divided into young control (YC), aged control (AC) and COR treated (COR-20) aged groups. COR-20 group received daily doses of COR (20 mg/kg) for 6 months. Cell viability and hormone levels were analysed by MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] and enzyme immunoassay kits with COR treated at 1, 5, and 10 µg/mL. Oxidative enzymes, spermatogenic, and apoptotic expression in testis tissues were evaluated by Western blotting and real-time RT-PCR. RESULTS: COR treatment (1, 5, and 10 µg/mL) significantly (p < 0.05 ∼ p < 0.001) inhibited the H2O2-induced decrease in the percentage of viable cells (from 63.27% to 71.25%, 85.67% and 93.97%, respectively), and reduced the malondialdehyde (MDA) content (from 4.28 to 3.98, 3.14 and 1.78 nM MDA/mg protein, respectively). Further, the decreased antioxidant enzymes (glutathione-S-transferase mu5, glutathione peroxidase 4 and peroxiredoxin 3), spermatogenesis-related factors (nectin-2 and inhibin-α) and testosterone levels in H2O2-exposed TM3 cells were significantly (p < 0.05 ∼ p < 0.001) ameliorated by COR. In aged rats, COR (20 mg/kg) restored the altered enzymatic and non-enzymatic antioxidative status and attenuated the apoptotic p53 and Bax/Bcl-2 expression significantly (p < 0.05). CONCLUSION: COR might be developed as a potential agent against ageing-associated and oxidative stress-induced male infertility.


Subject(s)
Deoxyadenosines/pharmacology , Leydig Cells/drug effects , Spermatogenesis/drug effects , Testis/drug effects , Aging , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Cordyceps/chemistry , Deoxyadenosines/isolation & purification , Hydrogen Peroxide , Leydig Cells/metabolism , Male , Mice , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
14.
Chem Biol Interact ; 353: 109797, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34998821

ABSTRACT

Although several studies have reported testicular impairments caused by cadmium (Cd) or obesity alone, the combined effect of Cd and obesity on the testes and its underlying mechanism remains unclear. We examined the combined effect of whole-life exposure to low-dose Cd started at preconception and post-weaning high-fat diet (HFD) on the testes of offspring mice. At weaning, male offspring parented with and without exposure to low-dose Cd were continued on the same drinking water regimen as their parents and fed with either a normal diet (ND) or HFD for 10 or 24 weeks. Whole-life exposure to Cd resulted in its accumulation in testes, and HFD induced obesity and lipid metabolism disorder. Exposure to Cd or HFD alone significantly decreased Johnsen scores, disrupted testicular structure, and increased germ cell apoptosis at both 10 and 24 weeks. However, co-exposure to Cd and HFD did not induce the toxic effects that were induced by either alone, as revealed by preserved testicular structure and spermatogenesis, lack of significant apoptosis, and increased cell proliferation. Mechanistically, the combined effects of low-dose Cd and HFD consumption were associated with the activation of the JAK/STAT pathway. These findings suggest that co-exposure to low-dose Cd and HFD did not cause Cd- or HFD-induced testicular injury, probably because of the activation of the JAK/STAT pathway to prevent germ cell apoptosis.


Subject(s)
Cadmium/toxicity , Diet, High-Fat , Spermatogenesis/drug effects , Testis/physiology , Animals , Animals, Newborn , Apoptosis/drug effects , Cadmium/analysis , Female , Germ Cells/cytology , Germ Cells/metabolism , Janus Kinases/metabolism , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Models, Animal , Oxidative Stress/drug effects , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Testis/anatomy & histology , Testis/chemistry
15.
Toxicol Lett ; 360: 1-10, 2022 May 01.
Article in English | MEDLINE | ID: mdl-35063618

ABSTRACT

3-Monochloropane-1,2-diol (3-MCPD), a common food contaminant, has been confirmed to impair male fertility, but the mechanism has not been fully clarified. This study systematically explored the spermatogenesis impairment induced by 3-MCPD in vivo and in vitro with a focus on Sertoli cells (SCs) and spermatogonial stem cells (SSCs). After adult male Sprague-Dawley rats were administered 36 and 72 mg/kg b.w./day 3-MCPD daily for 4 weeks, the total sperm concentration dramatically decreased by 28.9 % and 57.7 %, respectively, and obvious testicular seminiferous tubule atrophy was observed. 3-MPCD exposure decreased serum testosterone levels but not intratesticular testosterone levels and upregulated the expression of steroidogenesis enzymes in both rat testes and primary Leydig cells. 3-MCPD did not reduce the number and self-renewal marker PLZF+ of SSCs; however, it downregulated the key meiotic genes Stra8 and Rec8 in the rat testis but not in primary germ cells. Although SC counts were not affected, 3-MCPD downregulated androgen receptor (AR) in rat testes and primary SCs. In addition, 3-MCPD downregulated p-CREB (transcription factor of AR), paracrine meiosis regulators Nrg1 and Nrg3 and retinoic acid synthetase Aldh1a1 in primary SCs. In summary, 3-MCPD caused impairment of spermatogenesis by inhibiting secretion of meiosis regulators and disturbing testosterone signalling in SCs.


Subject(s)
Sertoli Cells , alpha-Chlorohydrin , Animals , Male , Rats , Rats, Sprague-Dawley , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Spermatogenesis/drug effects , Testis/drug effects , Testis/metabolism , Testosterone/analysis , Testosterone/metabolism , alpha-Chlorohydrin/toxicity
16.
J Ethnopharmacol ; 289: 115025, 2022 May 10.
Article in English | MEDLINE | ID: mdl-35074455

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Roots of Argyreia nervosa (Burm.f.) Bojer is used traditionally as an aphrodisiac and mentioned in the indigenous system of medicine as spermatogenic. The roots of the plant are also used as bitter, tonic, and alternative. AIM OF THE STUDY: To study the effect of n-butanol fraction (BTF) and ethyl acetate fraction (ETF) of methanol extract prepared from the roots of Argyreia nervosa and scopoletin isolated from ETF on testosterone biosynthesis in testis and spermatogenesis using rats. MATERIALS AND METHODS: The effect of BTF, ETF, and scopoletin on the testosterone biosynthesis was evaluated by determining the alteration in expression of mRNA corresponding to steroidogenic enzymes and concentration of testosterone using TM-3 cell line. The ability of BTF and ETF in altering the level of testicular cholesterol and testosterone along with mRNA expression corresponding to 3ß-Hydroxy-Δ5-steroid dehydrogenase (3ß-HSD) and Acute Steroid Regulatory Protein (StAR) was evaluated using rats as experimental animals. The sperm concentration in the seminal fluid was determined, and histological studies of testicular tissues were also carried out. RESULTS: Test solutions containing BTF, ETF, and scopoletin showed a dose-dependent and statistically significant increase in the testosterone content when incubated with TM-3 cells. The test solutions also increased the fold expression of mRNA corresponding to StAR and 3ß-HSD enzymes from TM-3 cells. BTF and ETF elevated testicular testosterone levels by 3.57 and 3.84-fold as compared to control animals, while the fractions showed 9.04 and 10.41-fold alteration in expression of mRNA corresponding to StAR, respectively. BTF and ETF altered the expression of mRNA corresponding to 3ß-HSD by 13.43 and 15.04-fold in testicular tissues; moreover, they elevated the activity of 3ß-HSD by 7.11 and 7.73 fold, respectively. The animals treated with BTF and ETF showed increased sperm concentration. Histological observations showed that the lumen of seminiferous tubules was densely populated with spermatozoa and Leydig cells were intensely stained. Extract prepared from fruits of Tribulus terrestris Linn and testosterone served as positive controls. CONCLUSION: BTF, ETF, and scopoletin could promote testosterone biosynthesis by elevating mRNA expression corresponding to StAR, 3ß-HSD, and by increasing 3ß-HSD activity in the testicular tissues. Elevated testosterone concentration in testis promoted spermatogenesis. The studies provided the probable mechanism through which the roots of A. nervosa act as spermatogenic.


Subject(s)
Convolvulaceae/chemistry , Plant Extracts/pharmacology , Spermatogenesis/drug effects , Testosterone/biosynthesis , 3-Hydroxysteroid Dehydrogenases/metabolism , Animals , Cell Line , Dose-Response Relationship, Drug , Leydig Cells/drug effects , Leydig Cells/metabolism , Male , Mice , Plant Extracts/administration & dosage , Plant Roots , RNA, Messenger/metabolism , Rats , Rats, Wistar , Testis/drug effects , Testis/metabolism
17.
Toxicology ; 468: 153102, 2022 02 28.
Article in English | MEDLINE | ID: mdl-35074511

ABSTRACT

Cisplatin exposure represents a significant fertility problem for childhood cancer. In this study we examined the possible therapeutic role of Zinc oxide nanoparticles (ZnO-NPs) on Cisplatin (Cis) induced impairment in the spermatogenesis initiation during puberty. Seventy-two male rats aged 30 days were distributed into four equal groups: Control group; ZnO-NPs group (intraperitoneal i.p. injected with 5 mg/kg ZnO-NPs once a week for eight weeks); Cis group (i.p. injected with a single dose of 5 mg/kg); ZnO-NPs + Cis group (ZnO-NPs injection 2 hrs before Cis). Each group was subdivided into three groups and was sacrificed 7, 30 and, 60 days after cisplatin induction, which considered prepubertal at 37-day-old, productive at 60-day-old, and completely adult at 90-day-old. Biochemical, molecular, immunohistochemical, and ultrastructural examinations were studied on the testicular tissues and sperm samples. Group treated with Cis showed a decrease in the antioxidant activity and an increase in the reactive oxygen species (ROS), which in turn caused disruption in blood-testis barrier (BTB) proteins in the three different rat ages, and sperm DNA damage in the adult rats compared to control group (p < 0.05). Moreover, alterations in the structural and the ultrastructural morphology of the testis were observed compared with the control at 37, 60 and 90 days old rats. ZnO-NPs administration to Cis group manifested a significant decrease in the ROS that restored the BTB proteins, enhanced the architecture of the testis in the three different rat ages, and increased sperm DNA integrity in the adult rats. Zinc oxide nanoparticles could restore the male reproductive capacity in the adult rats after induction of Cis in the prepubertal period by promoting spermatogenesis.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Spermatogenesis/drug effects , Testis/drug effects , Zinc Oxide/pharmacology , Animals , Body Weight , Comet Assay , Immunohistochemistry , Male , Microscopy, Electron, Transmission , Nanoparticles , Organ Size , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Testis/chemistry , Testis/physiology , Testis/ultrastructure , Vimentin/pharmacology
18.
J Pharm Pharmacol ; 74(2): 268-281, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34928371

ABSTRACT

OBJECTIVES: Curcumin is a promising nutraceutical with reported diverse therapeutic properties, but of limited oral bioavailability. The current manuscript investigates the role of encapsulation of curcumin in nanoemulsion form in counteracting the adverse effect of chronic ingestion of a high-fat high-fructose diet (HFHF) by juvenile male rats regarding testicular abnormalities and declined spermatogenesis. METHODS: Curcumin nanoemulsion was administered orally to Wistar rats at a dose of 5 or 10 mg/kg and compared with curcumin powder, followed by a pharmacological and histological assessment. KEY FINDINGS: Results demonstrated that curcumin nanoemulsion was superior to curcumin powder, particularly in enhancing the percentage progressive motility of spermatozoa, normalization of essential and non-essential amino acids in semen, normalization of serum leptin and testosterone levels, as well as normalization of oxidative and nitrosative parameters. It was also proven to reduce testicular DNA fragmentation, while elevating testicular cellular energy. In addition, curcumin nanoemulsion administered at a dose of 10 mg/kg induced the highest level of spermatogenesis, delineated by histological examination of the seminiferous tubules. CONCLUSIONS: It can be concluded that curcumin nanoemulsion administered at a dose of 10 mg/kg successfully ameliorates the adverse effects of a HFHF on spermatogenesis.


Subject(s)
Curcumin/pharmacology , Nanoparticles , Spermatogenesis/drug effects , Spermatozoa/drug effects , Administration, Oral , Animals , Curcumin/administration & dosage , DNA Fragmentation/drug effects , Diet, High-Fat/adverse effects , Dose-Response Relationship, Drug , Emulsions , Fructose/adverse effects , Male , Rats , Rats, Wistar
19.
Food Funct ; 13(1): 425-436, 2022 Jan 04.
Article in English | MEDLINE | ID: mdl-34913451

ABSTRACT

Our previous study confirmed the beneficial effects of chestnut polysaccharides (CPs) on the spermatogenesis process, but the exact mechanism is not clear. Several studies have demonstrated the importance of balanced gut microbiota in maintaining normal reproductive function. In this study, we investigated the biological functions of CPs from the perspective of gut microbiota function, expecting to find out the specific mechanism of CPs in restoring impaired spermatogenesis. Compared with the control group, the mice treated with busulfan showed a reduced number of germ cells, structural changes in the small intestine and composition alteration in the gut microbiota at several levels, including the phylum and genus. In contrast, the number of germ cells in seminiferous tubules was significantly increased, and the structure of the small intestine and the composition of the gut microbiota were altered in the busulfan-treated mice after the CPs treatment. The 16s rRNA analysis results showed that the Firmicutes was the predominant phylum in all groups followed by Proteobacteria, Bacteroidetes, Actinobacteria, Tenericutes, Cyanobacteria and unidentified bacteria. Interestingly, the subsequent functional analysis implied that the steroid hormone biosynthesis process is the major metabolic pathway in the CPs-mediated restoration process and the experimental results confirmed this speculation. In conclusion, this study confirmed that CPs can restore the impaired spermatogenesis process by adjusting the gut microbiota and intestinal structure, which will also provide technical support and a theoretical basis for the subsequent treatment of male infertility.


Subject(s)
Aesculus/chemistry , Gastrointestinal Microbiome/drug effects , Nuts/chemistry , Polysaccharides/pharmacology , Spermatogenesis/drug effects , Animals , Infertility, Male/metabolism , Intestines/drug effects , Male , Mice
20.
Chem Biol Interact ; 351: 109743, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34774840

ABSTRACT

Cannabidiol (CBD) is a natural cannabinoid present in the Cannabis sativa plant, widely prescribed as an anticonvulsant drug, especially for pediatric use. However, its effects on male reproduction are still little investigated. Therefore, the present study assessed the effects of CBD on the spermatogenesis and sperm quality. For this, twenty-one-day-old Swiss mice received CBD for 34 consecutive days by gavage at doses of either 15 or 30 mg/kg. Chronic exposure to CBD decreased the frequency of stages VII-VIII and XII of spermatogenesis and an increase in the frequency of stage IX were noted. Furthermore, the seminiferous epithelium height reduced at stage IX and increased at stage XII in both CBD-treated groups. There was a significant rise of sperm DNA damage, while no genotoxic effects were observed in leukocytes. The activities of superoxide dismutase and catalase decreased, while malondialdehyde levels increased in the sperm of mice treated with a higher dose of CBD. Mice exposed to 30 mg/kg of CBD showed a reduction in the mobile spermatozoa percentage and in curvilinear velocity, while straight line and average path velocity decreased in both treated groups. The number of acrosome-intact spermatozoa declined in the CBD 30 group, and the number of abnormal acrosomes raised in both CBD groups. On the other hand, the weight of reproductive organs, sperm count, and hormone levels were not affected by CBD treatment. These findings show that dysregulation of the endocannabinoid system by CBD can reduce sperm quality. The mechanisms responsible may be associated with disorders during spermatogenesis, especially during the final stages of nuclear remodelling and assembly of acrosome. However, changes in mitochondrial function, as well as the reduction on the antioxidant enzyme activities during epididymal transit, at least partly, may also be involved.


Subject(s)
Cannabidiol/toxicity , Spermatozoa/drug effects , Acrosome/drug effects , Animals , DNA Damage/drug effects , Lipid Peroxidation/drug effects , Male , Mice , Sperm Motility/drug effects , Spermatogenesis/drug effects , Spermatozoa/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...