Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 81
1.
Theranostics ; 14(6): 2622-2636, 2024.
Article En | MEDLINE | ID: mdl-38646657

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.


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.
Ecotoxicol Environ Saf ; 272: 116063, 2024 Mar 01.
Article En | MEDLINE | ID: mdl-38306818

Environmental pollution has emerged as a global concern due to its detrimental effects on human health. One of the critical aspects of this concern is the impact of environmental pollution on sperm quality in males. Male factor infertility accounts for approximately 40%- 50% of all infertility cases. Nonobstructive azoospermia (NOA) is the most severe type of male infertility. Human umbilical cord mesenchymal stem cell (hUCMSC) exosomes enhance proliferation and migration, playing crucial roles in tissue and organ injury repair. However, whether hUCMSC exosomes impacting on NOA caused by chemotherapeutic agents remains unknown. This study aimed to explore the functional restoration and mechanism of hUCMSC exosomes on busulfan-induced injury in GC-1 spg cells and ICR mouse testes. Our results revealed that hUCMSC exosomes effectively promoted the proliferation and migration of busulfan-treated GC-1 spg cells. Additionally, oxidative stress and apoptosis were significantly reduced when hUCMSC exosomes were treated. Furthermore, the injection of hUCMSC exosomes into the testes of ICR mice treated with busulfan upregulated the expression of mouse germ cell-specific genes, such as vasa, miwi, Stra8 and Dazl. Moreover, the expression of cellular junction- and cytoskeleton-related genes, including connexin 43, ICAM-1, ß-catenin and androgen receptor (AR), was increased in the testicular tissues treated with exosomes. Western blot analysis demonstrated significant downregulation of apoptosis-associated proteins, such as bax and caspase-3, and upregulation of bcl-2 in the mouse testicular tissues injected with hUCMSC exosomes. Further, the spermatogenesis in the experimental group of mice injected with exosomes showed partial restoration of spermatogenesis compared to the busulfan-treated group. Collectively, these findings provide evidence for the potential clinical applications of hUCMSC exosomes in cell repair and open up new avenues for the clinical treatment of NOA.


Acetates , Azoospermia , Exosomes , Mesenchymal Stem Cells , Phenols , Mice , Male , Humans , Animals , Busulfan/toxicity , Busulfan/metabolism , Exosomes/genetics , Mice, Inbred ICR , Semen , Umbilical Cord , Azoospermia/chemically induced , Azoospermia/therapy , Azoospermia/metabolism
3.
Sci Data ; 11(1): 163, 2024 Feb 02.
Article En | MEDLINE | ID: mdl-38307907

Chemotherapeutic drugs will affect the process of spermatogenesis. However, most current studies on the effects of chemotherapeutic drugs on spermatogenesis are based on mouse models, with a shortage of human body evidence. In addition, the mechanism of chemotherapeutic drugs causing spermatogenesis disorder is not clear. Therefore, we have collected the testicular tissues of an inguinal-lipoma patient whose testes were resected after chemotherapy and a patient who had normal spermatogenesis disorder and underwent single-nucleus RNA sequencing (snRNA-Seq). After quality control, we obtained a total of 27,957 high-quality cells, including 18,612 normal cells and 9,345 drug-treated cells, which were all used in analyzing the mechanism of chemotherapeutic drugs causing spermatogenesis disorder. This study has provided data resources and references for exploring the mechanism of chemotherapeutic drugs causing spermatogenesis disorder with the insight of protecting the spermatogenic abilities of male tumor patients receiving chemotherapy.


Azoospermia , Testis , Humans , Male , Azoospermia/chemically induced , Azoospermia/pathology , Base Sequence , Cyclophosphamide/adverse effects , Spermatogenesis
4.
Reprod Sci ; 31(2): 375-392, 2024 Feb.
Article En | MEDLINE | ID: mdl-37737972

Non-obstructive azoospermia is a severe form of male infertility, with limited effective treatments. Bone marrow mesenchymal stem cells (BMSCs) can differentiate to different cell lines; therefore, transplantation of these cells is used for treatment of several diseases. Since these cells require induction factors to differentiate into germ cells, we co-transplanted bone marrow stem cells (BMSCs) with Sertoli cell-conditioned medium (SCCM) into the testis of azoospermic mice. This study was carried out in two sections, in vitro and in vivo. For in vitro study, differentiating factors (c-kit and ID4) were examined after 15 days of co-culture of bone marrow cells with Sertoli cell-conditioned medium, while for in vivo study, the azoospermia model was first created by intraperitoneal administration of a single-dose busulfan (40 mg/kg) followed by single-dose CdCl2 (2 mg/kg) after 4 weeks. Mice were divided into 4 groups including control (azoospermia), BMSC, SCCM, and BMSC + SCCM. Eight weeks after transplantation, samples were assessed for proliferation and differentiation via the expression level of MVH, ID4, SCP3, Tp1, Tp2, and Prm1 differentiation markers. The results showed that BMSC co-cultured with SCCM in vitro differentiated BMSC to germ-like cells. Similarly, in vivo studies revealed a higher level of BMSC differentiation into germ-like cells with significant higher expression of differentiation markers in transplanted groups compared to the control. This study confirmed the role of SCCM as an inductive factor for BMSC differentiation to germ cells both in vivo and in vitro conditions.


Azoospermia , Mesenchymal Stem Cells , Humans , Male , Mice , Animals , Sertoli Cells/metabolism , Busulfan/pharmacology , Culture Media, Conditioned , Azoospermia/chemically induced , Azoospermia/metabolism , Bone Marrow , Cell Differentiation , Disease Models, Animal , Antigens, Differentiation , Mesenchymal Stem Cells/metabolism
5.
Fertil Steril ; 120(6): 1203-1209, 2023 12.
Article En | MEDLINE | ID: mdl-37769866

OBJECTIVE: To study sperm parameters recovery and fertility outcomes in men with azoospermia or severe oligospermia caused by anabolic steroid use who underwent a standardized treatment regimen for spermatogenesis recovery. DESIGN AND SUBJECTS: A retrospective analysis of a cohort of men with a prior history of anabolic steroid use and infertility complaints (between 2018 and 2022) was conducted. EXPOSURE: The standardized treatment approach involved discontinuing testosterone replacement therapy and administering a combination regimen of clomiphene citrate and human chorionic gonadotropin for a minimum of 3 to 6 months. MAIN OUTCOME MEASURES: The main outcome measures included changes in sperm parameters, predominantly sperm concentration, and subsequent pregnancy outcomes. RESULTS: A total of 45 men (median age 37 years, IQR 32-45) met the inclusion criteria for this analysis. Median duration of prior T use was 4 years (IQR 1.3-10), with the 2 most common modalities consisting of injection therapy (43.5%) and oral therapy (34.8%). The median initial sperm concentration was 0 million/cc (IQR 0-1.15), and 23 (51.1%) men initially presented with azoospermia. The median duration of combination human chorionic gonadotropin/clomid therapy was 5 months (IQR 3-12). In initially azoospermic men (N: 23), 5 were lost to follow-up, 6 (33.3%) progressed to severe oligospermia (<5 million/cc), 6 (33.3%) to oligospermia (<15 million/cc), 1 (5.6%) to normozoospermia (>15 million/cc), and 5 (27.8%) remained azoospermic after medical treatment for 6 months. Among the 24 couples who responded to the follow-up call, a total of 9 (37.5%) achieved a successful subsequent pregnancy. Of these, 33.3% (3 couples) used assisted reproductive technology, whereas 66.7% (6 couples) conceived naturally. On logistic regression analysis, no significant predictors for improved sperm parameters or successful pregnancy were identified. CONCLUSION: Despite appropriate treatment regimens, a significant proportion of men with a prior history of anabolic steroid use continue to exhibit severe oligospermia, with more than half showing limited improvement in semen parameters after 6 months of treatment. Only a fraction of men achieves normozoospermia after treatment. Further research is needed to explore predictors for improved sperm parameters and successful pregnancy outcomes in men with a history of anabolic steroid use.


Azoospermia , Oligospermia , Pregnancy , Female , Humans , Male , Adult , Oligospermia/chemically induced , Oligospermia/diagnosis , Oligospermia/drug therapy , Azoospermia/chemically induced , Azoospermia/diagnosis , Azoospermia/drug therapy , Anabolic Androgenic Steroids , Testosterone/adverse effects , Retrospective Studies , Semen , Chorionic Gonadotropin , Clomiphene/adverse effects , Fertility
6.
Biol Res ; 56(1): 2, 2023 Jan 19.
Article En | MEDLINE | ID: mdl-36653814

BACKGROUND: The testes are highly susceptible to the adverse effects of chemotherapy and radiation at all stages of life. Exposure to these threats mainly occurs during cancer treatment and as an occupational hazard in radiation centers. The present study investigated the regenerative ability of adipose-derived mesenchymal stem cells (ADMSCs) against the adverse effects of cisplatin on the structure and function of the testes. METHODS: New Zealand white rabbits (N = 15) were divided into three groups of five: a negative control group (no treatment), a cisplatin group (single dose of cisplatin into each testis followed three days later by a PBS injection), and a cisplatin + ADMSCs group (cisplatin injection followed three days later by an ADMSC injection). On day 45 post-treatment, serum testosterone levels were evaluated, and the testes and epididymis were collected for histology, oxidative stress examination, and epididymal sperm analysis. RESULTS: Cisplatin caused damage to the testicular tissue and decreased serum testosterone levels, epididymal sperm counts, and oxidants. An antioxidant imbalance was detected due to increasing malondialdehyde (MDA) and reduced glutathione (GSH) levels in testicular tissue. The ADMSC-treated group displayed a moderate epididymal sperm count, adequate antioxidant protection, suitable hormone levels, and enhanced testicular tissue morphology. CONCLUSIONS: ADMSCs treatment repaired damaged testicular tissue, enhanced biochemical parameters, and modified pathological changes caused by cisplatin.


Azoospermia , Mesenchymal Stem Cells , Male , Animals , Rabbits , Humans , Testis/metabolism , Cisplatin/adverse effects , Antioxidants/pharmacology , Azoospermia/chemically induced , Azoospermia/metabolism , Azoospermia/pathology , Semen , Spermatozoa/metabolism , Spermatozoa/pathology , Oxidative Stress , Testosterone/pharmacology
7.
J Ethnopharmacol ; 301: 115760, 2023 Jan 30.
Article En | MEDLINE | ID: mdl-36209951

ETHNOPHARMACOLOGICAL RELEVANCE: Ethnopharmacological studies for drug discovery from natural compounds play an important role for developing current therapeutical platforms. Plants are a group of natural sources which have been served as the basis in the treatment of many diseases for centuries. In this regard, Ceratonia siliqua (carob) is one of the herbal medicine which is traditionally used for male infertility treatments. But so far the main mechanisms for effects of carob are unknown. Here, we intend to investigate the ability of carob extract to induce spermatogenesis in an azoospermia mouse model and determine the mechanisms that underlie its function. AIM OF THE STUDY: This is a pre-clinical animal model study to evaluate the effect of carob extract in spermatogenesis recovery. METHODS: We established an infertile mouse model with the intent to examine the ability of carob extract as a potential herbal medicine for restoration of male fertility. Sperm parameters, as well as gene expression dynamics and levels of spermatogenesis hormones, were evaluated 35 days after carob administration. RESULTS: Significant enhanced sperm parameters (P < 0.05) showed that the carob extract could induce spermatogenesis in the infertile mouse model. Our data suggested an anti-apototic and inducer role in the expressions of cell cycle regulating genes. Carob extract improved the spermatogenesis niche by considerable affecting Sertoli and Leydig cells (P < 0.05). The carob-treated mice were fertile and contributed to healthy offspring that matured. Our data confirmed that this extract triggered the hormonal system, the spermatogenesis-related gene expression network, and signaling pathways to induce and promote sperm production with notable level (P < 0.05). We found that the aqueous extract consisted of a polar and mainly well water-soluble substance. Carob extract might upregulate spermatogenesis hormones via its amino acid components, which were detected in the extract by liquid chromatography-mass spectrometry (LC-MS). CONCLUSION: Our results strongly suggest that carob extract might be a promising future treatment option for male infertility. This finding could pave the way for clinical trials in infertile men. This is the first study that has provided reliable, strong pre-clinical evidence for carob extract as an effective candidate for fertility recovery in cancer-related azoospermia.


Azoospermia , Fabaceae , Infertility, Male , Humans , Male , Animals , Mice , Azoospermia/chemically induced , Azoospermia/drug therapy , Azoospermia/genetics , Up-Regulation , Spermatogenesis , Infertility, Male/drug therapy , Infertility, Male/metabolism , Disease Models, Animal , Hormones , Seeds/metabolism , RNA-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Protamines/genetics , Protamines/metabolism
8.
Biol. Res ; 56: 2-2, 2023. ilus, tab, graf
Article En | LILACS | ID: biblio-1420300

BACKGROUND: The testes are highly susceptible to the adverse effects of chemotherapy and radiation at all stages of life. Exposure to these threats mainly occurs during cancer treatment and as an occupational hazard in radiation centers. The present study investigated the regenerative ability of adipose-derived mesenchymal stem cells (ADMSCs) against the adverse effects of cisplatin on the structure and function of the testes. METHODS: New Zealand white rabbits (N = 15) were divided into three groups of five: a negative control group (no treatment), a cisplatin group (single dose of cisplatin into each testis followed three days later by a PBS injection), and a cisplatin + ADMSCs group (cisplatin injection followed three days later by an ADMSC injection). On day 45 post-treatment, serum testosterone levels were evaluated, and the testes and epididymis were collected for histology, oxidative stress examination, and epididymal sperm analysis. RESULTS: Cisplatin caused damage to the testicular tissue and decreased serum testosterone levels, epididymal sperm counts, and oxidants. An antioxidant imbalance was detected due to increasing malondialdehyde (MDA) and reduced glutathione (GSH) levels in testicular tissue. The ADMSC-treated group displayed a moderate epididymal sperm count, adequate antioxidant protection, suitable hormone levels, and enhanced testicular tissue morphology. CONCLUSIONS: ADMSCs treatment repaired damaged testicular tissue, enhanced biochemical parameters, and modified pathological changes caused by cisplatin.


Humans , Animals , Male , Rabbits , Azoospermia/chemically induced , Azoospermia/metabolism , Azoospermia/pathology , Semen , Spermatozoa/metabolism , Spermatozoa/pathology , Testis/metabolism , Testosterone/pharmacology , Cisplatin/adverse effects , Oxidative Stress , Mesenchymal Stem Cells , Antioxidants/pharmacology
9.
Turk J Med Sci ; 52(3): 778-787, 2022 Jun.
Article En | MEDLINE | ID: mdl-36326321

BACKGROUND: This study evaluated the treatment procedures for chemotherapy (CT)-induced persistent azoospermia and their outcomes from a different perspective. METHODS: In 63 patients (mean age: 30.16 ± 4.91 years) who had undergone CT 11 ± 5 years earlier, the semen volume, gonadotropins level, FSH level, genetics, micro-testicular sperm extraction (m-TESE) result, sperm DNA fragmentation index (SDFI), semen reactive oxidative stress (ROS) rate, duration of embryonic development, and pregnancy and baby take-home rates were examined. The correlations between the ROS rates and the SDFIs, m-TESE results, sperm motility, pathology scores, time-lapses, and baby take-home rates were evaluated. RESULTS: The semen volumes were 3.5 ± 1.1/ml. The FSH level following CT was 17.87 ± 5.80 mIU/ml. A sperm rate of 34.9% was found from the m-TESE result. The mean SDFI and ROS rate were 4 (<15-30>) and 1.29 ± 0.51, respectively. The time-lapse was calculated as 5h. Pregnancy and live birth were achieved at 20.63% and 12.7%, respectively. In the patients with a low ROS (≤1.42) and SDFI (≤15), the m-TESE success rate was high, the FSH value was low, the pathological score and fertilization rate were elevated, the embryonic cleavage period was normal, and the pregnancy and baby take-home rates were high. DISCUSSION: The sperms may be detected using m-TESE in patients who develop persistent azoospermia associated with CT due to different oncological diagnoses. Our study revealed that a low FSH value and normal ejaculatory ROS rates are positive predictive factors of sperm detection before m-TESE. The motility of the sperms detected after m-TESE and normal SDFI rates were found to be positive predictive criteria of high fertilization, good embryonic cleavage, pregnancy, and live birth.


Azoospermia , Pregnancy , Female , Humans , Male , Adult , Azoospermia/chemically induced , Azoospermia/pathology , Azoospermia/therapy , Sperm Retrieval , Retrospective Studies , Reactive Oxygen Species , Sperm Motility , Semen , Testis/pathology , Spermatozoa/pathology , Follicle Stimulating Hormone
10.
Andrologia ; 54(11): e14595, 2022 Dec.
Article En | MEDLINE | ID: mdl-36102209

Gene expression in meiotic cells in the testis is characterized by intense transcriptional activity and alternative splicing. These processes are mainly controlled by RNA-binding proteins expressed strongly in germ cells. Functional impairments in any of these proteins' functions can lead to defects in meiosis and thus severe male infertility. Here, we have identified a homozygous frameshift mutation (NM_014469.4:c.301dup; p.Ser101LysfsTer29) in the RNA-binding motif protein, X-linked like 2 (RBMXL2) gene in a man with an azoospermia due to meiotic arrest. As RBMXL2 is known to be crucial for safeguarding the meiotic transcriptome in mice testes, we hypothesized that this variant leads to cryptic splice site poisoning. To determine the variant's impact on spermatogenesis, we confirmed the absence of RBMXL2 protein in the patient's testis tissue and then evidenced abnormal expression of several spermatogenesis proteins (e.g. meiosis-specific with coiled-coil domain) known to be altered in rbmxl2 knock-out mice with meiotic arrest. Our results indicate that RBMXL2's function in spermatogenesis is conserved in mammals. We hypothesize that deleterious variant in the RBMXL2 gene can result in male infertility and complete meiotic arrest, due to the disruption of gene expression by cryptic splice site poisoning.


Azoospermia , Infertility, Male , Humans , Mice , Animals , Male , RNA Splice Sites/genetics , Frameshift Mutation , Azoospermia/chemically induced , Azoospermia/genetics , Azoospermia/metabolism , Meiosis/genetics , Infertility, Male/genetics , Infertility, Male/metabolism , Spermatogenesis/genetics , Testis/metabolism , RNA-Binding Proteins/genetics , Mutation , Mammals/genetics , Mammals/metabolism
11.
Reprod Fertil Dev ; 34(17): 1078-1088, 2022 Oct.
Article En | MEDLINE | ID: mdl-36127818

CONTEXT: Approximately 40-50% of all infertility cases are due to male infertility, and one of the most important causes of infertility is azoospermia. AIMS: This study aimed to evaluate the potential effect of elderberry on the spermatogenesis process in the azoospermia mice model. METHOD: Thirty adult male mice were randomised into three groups: control; busulfan (45mg/kg); and busulfan+elderberry (2%), 6mL orally per animal. Sperm samples were collected from the tail of the epididymis, and testis specimens were also collected and then subjected to sperm parameters analysis, histopathological evaluation, reactive oxygen species (ROS), and glutathione (GSH) measurement to determine the mRNA expression and hormonal assay. CONCLUSIONS: It can be concluded that the elderberry diet may be considered a complementary treatment to improve the spermatogenesis process in busulfan-induced azoospermic mice. IMPLICATIONS: Considering some limitations, the elderberry diet can be an alternate option for improving testicular damage following chemotherapy.


Azoospermia , Sambucus , Humans , Male , Mice , Animals , Azoospermia/chemically induced , Azoospermia/genetics , Busulfan/pharmacology , Seeds , Spermatogenesis , Testis/metabolism , Diet
12.
Theriogenology ; 191: 47-53, 2022 Oct 01.
Article En | MEDLINE | ID: mdl-35964476

Busulfan (Bus), is an alkylating agent widely used in chemotherapy which has been proven to possess toxic side effects on testicles. This study was carried out to compare the probable treatment effects of resveratrol (Res) or/and l-carnitine (Lca), as strong antioxidants, on the testicular tissue as well as on the level of sex hormones in busulfan-induced azoospermic rat models. A total of 78 adult male rats, were divided into six different experimental groups including: 1) Control; 2) Lca + Res; 3) BUS; 4) Bus + Lca; 5) BUS + Res and 6) Bus + Lca + Res. Busulfan was intraperitoneally administered in a single dose (10 mg/kg b.w), while resveratrol (20 mg/kg b.w/day) and l-carnitine (200 mg/kg b.w/day) were orally administered by gavage during 48 consecutive days to the rats. At the end of the experiment in all groups the level of LH, FSH, and testosterone were biochemically analyzed by ELISA and the testicular tissue evaluated histologically using stereological technique. Results showed that Lca or/and Res, increased the body and testis weight, the volume of the testis, interstitial tissue, germinal epithelium, and seminiferous tubule, the number of the different cells of germinal epithelium and the level of testosterone. On the other hand, Lca, Res and their combination decreased the concentration of LH and FSH compared to the group treated with Bus. In conclusion, these results suggested that l-carnitine or/and resveratrol treatment significantly attenuated busulfan -induced changes of the rat reproductive system led to the recovery of both testis and sperm parameters. However, co-administration of L-ca and Res was more effective than their individual treatment. This combination may alleviate the side effects of alkylating drugs, such as busulfan and may be beneficial for spermatogenesis.


Azoospermia , Rodent Diseases , Animals , Azoospermia/chemically induced , Azoospermia/veterinary , Busulfan/pharmacology , Carnitine/pharmacology , Follicle Stimulating Hormone/pharmacology , Male , Rats , Resveratrol/pharmacology , Rodent Diseases/chemically induced , Semen , Spermatogenesis , Testis , Testosterone/pharmacology
13.
BMC Pharmacol Toxicol ; 23(1): 50, 2022 07 14.
Article En | MEDLINE | ID: mdl-35831882

BACKGROUND: Administration of antineoplastic drugs may cause azoospermia driving to subfertility. Production of animal azoospermia models is essential for evaluating new treatment methods before therapeutic interventions in human setup. This study aimed to investigate the toxic effects of Busulfan (an anticancer drug) on some vital organs and describe the best method and appropriate dose of Busulfan to induce an animal azoospermia model. METHODS: Rats were randomly assigned into four groups, treatment groups received 10 mg/kg, 40 mg/kg Busulfan intraperitoneally (IP), 5 mg/kg Busulfan intratesticular (IT), and control group. Blood, bone marrow, liver, renal, and testes samples were collected for histological (H&E staining), biochemical (serum levels of ALT, AST, ALP, creatinine, and urea), and hematological analyses. RESULTS: Results revealed severe anemia and leukopenia in rats that received Busulfan via IP. By contrast, injection of 5 mg/kg Busulfan via IT did not cause anemia except with a mild decrease in RBC count. Non-significant differences in the M/E ratio were observed in all groups. The administration of 40 mg/kg of Busulfan led to evacuation and destruction in the spermatogenesis process with thin-walled seminiferous epithelium in most tubules, but in rats treated with 10 mg/kg of Busulfan, the normal spermatogenesis process was notified. IT injection of Busulfan contributed to the complete degradation of spermatogenesis in which all spermatogenic cells degenerated. In the renal tissue, hyperemia, extensive tubular necrosis degeneration, and hyaline casts were found after IP injection of Busulfan. In hepatic tissue, focal hemorrhagic, chronic cholangitis, and hepatocyte degeneration, and swelling were noticed. Biochemical analysis revealed apparent Busulfan toxicity of both hepatic and renal tissues in IP Busulfan-treated rats. CONCLUSIONS: In summary, we found that the intratesticular injection of low doses of Busulfan (5 mg/kg) is a relatively non-invasive and safe method for producing the rat azoospermia model causing the least toxicity on vital organs.


Azoospermia , Animals , Azoospermia/chemically induced , Azoospermia/pathology , Azoospermia/therapy , Busulfan/toxicity , Disease Models, Animal , Humans , Injections , Injections, Intraperitoneal , Male , Rats , Spermatogenesis , Testis
14.
Andrologia ; 53(8): e14144, 2021 Sep.
Article En | MEDLINE | ID: mdl-34143903

This study aimed at the efficacy of sequential treatment of bone marrow-derived mesenchymal stem cell secretion for busulfan-treated azoospermia in mice. The conditioned media (CM) was obtained from bone marrow mesenchymal stem cells (MSCs) or 293 cells. Chemically induced azoospermia mice received 200 µl MSC-CM or 293-CM twice a week intravenously for three consecutive weeks. The histological assessment of spermatogenic recovery quantifying the expression of meiosis-associated genes, and Sertoli cell barrier functional factors were assessed. The characteristics of TM4 cells (Sertoli cell line) after pre-incubation of MSC-CM in vitro were also obtained. The MSC-CM group had the most spermatogenic colonies among the three groups (p < .05), but no spermatids were seen. Expressions of the meiosis-associated genes Dazl, Vasa, Miwi, Stra8, CyclinA1, Pgk2 and Scp3 in MSC-CM testis were remarkably higher compared with 293-CM and busulfan groups respectively (p < .05). The levels of Sertoli cell barrier functional factors, for example ICAM-1 and N-cadherin, were significantly increased during MSC-CM treatment (p < .05). Moreover, pre-incubation of MSC-CM particularly accelerated the CD54 (ICAM-1) and CD44 expressions of TM4 cells and promoted cell inherent adhesion. MSC-CM treatment can significantly improve the short-term restoration of spermatogonial structures of chemically induced azoospermia related to facilitating Sertoli cell adhesion integrity.


Azoospermia , Mesenchymal Stem Cells , Animals , Azoospermia/chemically induced , Azoospermia/therapy , Busulfan/toxicity , Humans , Male , Mice , Sertoli Cells , Spermatogenesis
15.
Eur J Cancer ; 152: 41-48, 2021 07.
Article En | MEDLINE | ID: mdl-34062486

BACKGROUND: Immune checkpoint inhibitors (ICIs) are widely used and may induce long-term survival in various types of cancer. Yet, there is scarce evidence on potential effects on patient fertility and the necessity of cryopreservation before treatment onset. The aim of our study was to assess the prevalence of male infertility after initiation of ICI treatment. METHODS: This is a monocenter, cross-sectional pilot study. Fertility was investigated by spermiogram, analysis of sexual hormones and questionnaires on sexual function and sexual activity. Male patients under the age of 60 years previously or currently treated with ICI for cutaneous malignancies or uveal melanoma were included. RESULTS: Twenty-five patients were included, with a median age of 49 years. Eighteen of 22 (82%) available spermiograms showed no pathologies, all patients reported a normal sexual function and sexual activity. Of four patients with pathological spermiogram, three patients were diagnosed with azoospermia and one with oligoasthenoteratozoospermia. Three patients had significant confounding factors (previous inguinal radiotherapy, chemotherapy and chronic alcohol abuse, and bacterial orchitis). One patient with normal spermiogram before ICI treatment presented 1 year after initiation with azoospermia, showing an asymptomatic, inflammatory infiltrate with predominantly neutrophil granulocytes, macrophages and T-lymphocytes in the ejaculate. Infectious causes were ruled out; andrological examination was unremarkable. A second case with reduced sperm counts during treatment may be ICI-induced also. CONCLUSIONS: Most patients had no restrictions in fertility, yet an inflammatory loss of spermatogenesis seems possible. Cryopreservation should be discussed with all patients with potential future desire for children before treatment.


Azoospermia/diagnosis , Fertility/drug effects , Immune Checkpoint Inhibitors/adverse effects , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Uveal Neoplasms/drug therapy , Adult , Azoospermia/chemically induced , Azoospermia/immunology , Cross-Sectional Studies , Cryopreservation , Fertility/immunology , Fertility Preservation , Humans , Male , Melanoma/immunology , Middle Aged , Pilot Projects , Referral and Consultation , Semen Analysis , Skin Neoplasms/immunology , Spermatogenesis/drug effects , Spermatogenesis/immunology , Uveal Neoplasms/immunology
16.
Life Sci ; 274: 119336, 2021 Jun 01.
Article En | MEDLINE | ID: mdl-33716061

AIMS: This study aimed to explore the therapeutic effects of amniotic fluid-derived extracellular vesicles including exosomes (AF-Exos) on the recovery of sperm production capacity in a rat model of azoospermia. MAIN METHODS: The non-obstructive azoospermia (NOA) was induced in rats using intratesticular administration of Busulfan. Azoospermia was confirmed by testis histology. AF-Exos samples containing 10 or 40 µg exosomal proteins were injected into testicular tissue of NOA rats. After two months, the recovery of spermatogenesis was monitored via histopathological staining, spermiogram, and hormonal analysis. Immunohistochemistry staining for OCT-3/4 was used to identify of spermatogonial progenitors. The expression of DAZL and VASA, was also measured. KEY FINDINGS: AF-Exos exhibited sphere-shaped morphology with the mean diameter and zeta potential of 50 ± 7.521 nm and -7.16 mV. Immunoblots revealed that isolated nanoparticles were CD63, CD9, and CD81 positive. Histopathological evaluation revealed that spermatogenesis was improved significantly in NOA rats after AF-Exos injection. Data showed that the sperm parameters and spermatogenesis index were significantly improved after AF-Exos injection compared to azoospermic groups. OCT-3/4+ cells were increased in NOA rats after AF-Exos injection, showing the restoration of spermatogenesis. In the present study, both doses of exosome (10 and 40 µg) restored the testicular function of NOA rats. DAZL and VASA were increased significantly in animals who received 40 µg exosomal protein compared to azoospermic rats. Except in a high dose of AF-Exos (40 µg) for Testosterone and FSH, no statistically significant differences were found regarding hormones post-exosome injection. SIGNIFICANCE: Our study demonstrated that AF-Exos regenerated spermatogenesis and improved sperm quality in NOA rats.


Amniotic Fluid/chemistry , Azoospermia/therapy , Busulfan/toxicity , Exosomes/metabolism , Spermatogenesis , Alkylating Agents/toxicity , Animals , Azoospermia/chemically induced , Azoospermia/metabolism , Azoospermia/pathology , Exosomes/chemistry , Male , Rats , Rats, Wistar
17.
Tissue Eng Regen Med ; 18(2): 279-295, 2021 04.
Article En | MEDLINE | ID: mdl-33713308

BACKGROUND: Busulfan is an alkylating chemotherapeutic agent that is routinely prescribed for leukemic patients to induce myelo-ablation. However, it also results in azoospermia and infertility in cancer survivors. This research was constructed to explore the possible therapeutic role of amniotic fluid-derived stem cells (AFSCs) in improving busulfan-induced azoospermia in adult rats. METHODS: Forty two adult male albino rats were randomized into: (1) control group, (2) azoospermia group, (3) spontaneous recovery group, and (4) AFSCs-treated group, in which AFSCs were transplanted through their injection into the testicular efferent ducts. The assessment included a histo-pathological examination of the seminiferous tubules by the light and transmission electron microscopes. Additionally, the confocal laser scanning microscope was used for confirmation of homing of the implanted cells. Moreover, we conducted an immuno-fluorescence study for detection of the proliferating cell nuclear antigen (PCNA) in the spermatogenic cells, epididymal sperm count, and a histo-morphometric study. RESULTS: AFSCs successfully homed over the basement membrane of the injured seminiferous tubules. They greatly attenuated busulfan-induced degenerative and oxidative changes. They also caused a re-expression of PCNA in the germ cells, leading to resumption of spermatogenesis and re-appearance of spermatozoa. CONCLUSION: AFSCs could be a promising treatment modality for male infertility induced by chemotherapy, as they possess prominent regenerative, anti-apoptotic, and anti-inflammatory potentials.


Amniotic Fluid , Antineoplastic Agents, Alkylating , Azoospermia , Busulfan , Aged , Animals , Antineoplastic Agents, Alkylating/adverse effects , Azoospermia/chemically induced , Busulfan/adverse effects , Female , Humans , Male , Rats , Rats, Sprague-Dawley , Stem Cells
18.
Andrologia ; 53(2): e13927, 2021 Mar.
Article En | MEDLINE | ID: mdl-33355959

Busulfan-induced testicular injury mouse models are commonly used for experiments on spermatogonial stem cell transplantation, treatments for azoospermia due to spermatogenic failure and preserving male fertility after chemotherapy. Here, we investigated the value of testicular quantitative ultrasound for evaluating spermatogenic function in this model. In this study, testicular ultrasound was performed on mice from day 0 to 126 after busulfan treatment (n = 48), and quantitative data, including the testicular volume, mean pixel intensity and pixel uniformity, were analysed. The results revealed that from day 0 to 36, the testicular volume was positively associated with the testicle-to-body weight ratio (r = .92). On day 63, the pixel uniformity, which remained stable from day 0 to 36, declined significantly compared with that on day 36 (p < .01). On day 126, when the whole progression of spermatogenesis could be observed in most tubules, the mean pixel intensity also returned to normal (p > .05). In conclusion, testicular quantitative ultrasound could be used as a noninvasive and accurate monitoring method for evaluating spermatogenic function in busulfan-induced testicular injury mouse models.


Azoospermia , Testis , Animals , Azoospermia/chemically induced , Azoospermia/diagnostic imaging , Busulfan/toxicity , Humans , Male , Mice , Spermatogenesis , Spermatogonia , Testis/diagnostic imaging
19.
Biomed Pharmacother ; 133: 110868, 2021 Jan.
Article En | MEDLINE | ID: mdl-33181455

Male infertility has become an important health problem that is primarily caused by testicular dysfunction with abnormal spermatogenesis. In this study, we demonstrated that the neuropeptide, substance P (SP), is essential for spermatogonia proliferation in a seminiferous tubule culture system. In addition, SP (5 nmol/kg) treatment markedly restored spermatogenesis, improved sperm quality, and increased the number of ZBTB16+ or LIN28+ undifferentiated spermatogonia as well as STRA8+ differentiated spermatogonia in a busulfan-induced non-obstructive azoospermic mouse model. Furthermore, 100 nM SP treatment in vitro significantly stimulated the proliferation of GC-1 spg cells (a spermatogonia cell line) via activation of the Erk1/2 signaling pathway. Moreover, the sperm quality and the number of spermatogonia were significantly reduced after treatment with RP67580, a selective NK-1 receptor antagonist, suggesting that SP-NK1R signaling plays an important role in spermatogenesis. Taken together, these results suggest that SP may be a potential therapeutic agent for male infertility by accelerating the restoration of spermatogenesis.


Azoospermia/drug therapy , Fertility Agents, Male/pharmacology , Fertility/drug effects , Spermatogenesis/drug effects , Spermatogonia/drug effects , Substance P/pharmacology , Animals , Apoptosis/drug effects , Azoospermia/chemically induced , Azoospermia/metabolism , Azoospermia/physiopathology , Busulfan , Cell Line , Cell Proliferation/drug effects , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Mice, Inbred C57BL , Receptors, Neurokinin-1/agonists , Receptors, Neurokinin-1/metabolism , Signal Transduction , Spermatogonia/metabolism , Tissue Culture Techniques
20.
Hum Reprod Update ; 26(6): 874-885, 2020 11 01.
Article En | MEDLINE | ID: mdl-32935838

BACKGROUND: Children with cancer often face infertility as a long-term complication of their treatment. For boys, compromised testicular function is common after chemotherapy and currently there are no well-established options to prevent this damage. Platinum-based agents are used to treat a wide variety of childhood cancers. However, platinum agents are not currently included in the cyclophosphamide equivalent dose (CED), which is used clinically to assess the risks to fertility posed by combination chemotherapy in children with cancer. OBJECTIVE AND RATIONALE: This was a systematic search of the literature designed to determine the evidence for effects of platinum-based cancer treatment on the prepubertal human testis in relation to subsequent testicular function and fertility. SEARCH METHODS: PubMed and EMBASE were searched for articles published in English between 01 January 1966 and 05 April 2020 using search terms including 'cancer treatment', 'chemotherapy', 'human', 'prepubertal', 'testis', 'germ cells', 'testosterone' and related terms. Abstracts were screened and full-text articles were obtained for those that met the three major inclusion criteria (age ≤12 years at treatment, exposure to platinum-based chemotherapeutic and measure of reproductive function). Screening of bibliographies for full-text articles was used to identify additional studies. OUTCOMES: Our initial search identified 1449 articles of which 20 (1.3%) studies (n = 13 759 males) met all inclusion criteria. A control group (healthy individuals or siblings) was included for 5/20 (25%) studies. A total of 10/20 (50%) studies provided sub-analysis of the relative gonadotoxicity of platinum-based agents.The primary outcome measures were: pregnancies and fatherhood; semen analysis; and hormonal function. For pregnancies and fatherhood, three studies (n = 10 453 males) reported negative associations with platinum-agents, including the largest (n = 5640) controlled study (hazard ratio = 0.56, P = 0.0023), whilst two other studies (n = 1781) with platinum sub-analysis reported no association. For semen analysis (based on World Health Organization criteria), platinum-based chemotherapy was associated with azoospermia in one study (n = 129), whilst another (n = 44) found no association and the remainder did not perform platinum-based sub-analysis. For hormone analysis, conflicting results were obtained regarding potential associations between platinum-based agents and elevated FSH (a proxy for impaired spermatogenesis); however, the majority of these studies were based on low numbers of patients receiving platinum-based chemotherapy. WIDER IMPLICATIONS: Overall, these results indicate that platinum-based chemotherapy should be included in clinical calculators, for example CED, used to determine gonadotoxicity for childhood cancer treatment. These findings have important implications for clinicians regarding counselling patients and their carer(s) on fertility risk, guiding requirements for fertility preservation strategies (e.g. testicular tissue cryopreservation) and modification of treatments to reduce or eliminate the risk of infertility in childhood cancer survivors.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Fertility/drug effects , Neoplasms/drug therapy , Platinum Compounds/administration & dosage , Testis/drug effects , Adult , Age of Onset , Azoospermia/chemically induced , Azoospermia/epidemiology , Cancer Survivors/statistics & numerical data , Child , Female , Fertility Preservation/methods , Humans , Infertility, Male/chemically induced , Infertility, Male/epidemiology , Infertility, Male/prevention & control , Male , Neoplasms/epidemiology , Pregnancy , Testis/physiology
...