Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Ecotoxicol Environ Saf ; 272: 116063, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38306818

ABSTRACT

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.


Subject(s)
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
2.
Biol Res ; 56(1): 2, 2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36653814

ABSTRACT

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.


Subject(s)
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
3.
Reprod Fertil Dev ; 34(17): 1078-1088, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36127818

ABSTRACT

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.


Subject(s)
Azoospermia , Sambucus , Humans , Male , Mice , Animals , Azoospermia/chemically induced , Azoospermia/genetics , Busulfan/pharmacology , Seeds , Spermatogenesis , Testis/metabolism , Diet
4.
Andrologia ; 54(11): e14595, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36102209

ABSTRACT

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.


Subject(s)
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
5.
Turk J Med Sci ; 52(3): 778-787, 2022 Jun.
Article in English | MEDLINE | ID: mdl-36326321

ABSTRACT

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.


Subject(s)
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
6.
Andrologia ; 53(8): e14144, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34143903

ABSTRACT

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.


Subject(s)
Azoospermia , Mesenchymal Stem Cells , Animals , Azoospermia/chemically induced , Azoospermia/therapy , Busulfan/toxicity , Humans , Male , Mice , Sertoli Cells , Spermatogenesis
7.
Andrologia ; 53(2): e13927, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33355959

ABSTRACT

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.


Subject(s)
Azoospermia , Testis , Animals , Azoospermia/chemically induced , Azoospermia/diagnostic imaging , Busulfan/toxicity , Humans , Male , Mice , Spermatogenesis , Spermatogonia , Testis/diagnostic imaging
8.
Andrologia ; 52(1): e13474, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31710394

ABSTRACT

Ubiquitin conjugating enzyme (E2) is crucial for mediating N-terminal ubiquitination. Recent study reports that UBE2W is involved in male infertility. However, the correlation between UBE2W expression and hypospermatogenesis is unclear. The present study is to explore the biological role of UBE2W and its association with hypospermatogenesis. Results showed that the sexpression levels of UBE2W in mouse testes were gradually elevated from 2 to 10 weeks, while were significantly deceased in the testes with hypospermatogenesis. When UBE2W expression was successfully down-regulated in spermatogenic cells, the rate of apoptosis was significantly increased and the P53/Bcl-2/caspase 6/caspase 9 signal pathways were activated. Thus, these data indicate that UBE2W down-regulation promotes cell apoptosis and correlates with hypospermatogenesis, which may be helpful for the diagnosis of male infertility.


Subject(s)
Azoospermia/pathology , Spermatogenesis/physiology , Testis/pathology , Ubiquitin-Conjugating Enzymes/metabolism , Animals , Apoptosis , Azoospermia/chemically induced , Azoospermia/physiopathology , Busulfan/toxicity , Cell Line , Dimethyl Sulfoxide/toxicity , Disease Models, Animal , Down-Regulation , Humans , Male , Mice , RNA, Small Interfering/metabolism , Spermatocytes , Spermatogonia , Ubiquitin-Conjugating Enzymes/genetics
9.
Indian J Med Res ; 150(1): 81-86, 2019 07.
Article in English | MEDLINE | ID: mdl-31571633

ABSTRACT

Background & objectives: For improved male contraception, a new polymeric drug molecule - Reversible Inhibition of Sperm under Guidance (RISUG) has been synthesized and has been found to be effective, safe and reversible in various animal species. Phase-I and phase-II clinical trials have confirmed its safety and contraceptive efficacy. The present study was undertaken as a multicentric-limited phase-III clinical trial to test the efficacy and safety of RISUG in human volunteers. Methods: One hundred and thirty nine young males each having at least two children and living with wife were given 120 µl of RISUG as bilateral vas intraluminal injection. After the single-dose administration, the individuals were followed in respect of general health and semen parameters. Their wives were also followed particularly to determine onset of pregnancy. Results: During the six month follow up, the health of male volunteers and their wives was normal with no significant adverse effects. Temporary scrotal enlargement and mild scrotal and inguinal region pain were manifested in most individuals and resolved within one month without any routine activity impairment. In six individuals, there was injection procedure failure and azoospermia was not achieved. The other 133 individuals had either severe oligozoospermia or azoospermia at the first semen examination one month following RISUG injection; 82.7 per cent individuals had continued azoospermia in the month following first semen examination onwards and the rest 17.3 per cent manifested azoospermia within three to six months. Interpretation & conclusions: RISUG intravasal injection appears to be a safe clinical procedure with no significant adverse effects and has high sustained contraceptive efficacy. The localized intervention and continued contraceptive action on single-dose administration were significant features of the RISUG technology.


Subject(s)
Contraception/methods , Contraceptive Agents, Male/administration & dosage , Polyesters/administration & dosage , Polystyrenes/administration & dosage , Vas Deferens/drug effects , Adult , Animals , Azoospermia/chemically induced , Azoospermia/diagnosis , Azoospermia/pathology , Contraceptive Agents, Male/adverse effects , Female , Humans , Injections , Male , Polyesters/adverse effects , Polystyrenes/adverse effects , Pregnancy , Semen/drug effects , Semen Analysis , Spermatozoa/drug effects , Spermatozoa/pathology , Spouses , Volunteers
10.
Reproduction ; 156(3): 269-282, 2018 09.
Article in English | MEDLINE | ID: mdl-29921625

ABSTRACT

The Big Blue λSelect-cII selection system has been employed along with whole-exome sequencing to examine the susceptibility of the male germ line to mutation in two challenging situations (i) exposure to a chemotherapeutic regime including bleomycin, etoposide and cis-platinum (BEP) and (ii) the ageing process. A 3-week exposure to BEP induced complete azoospermia associated with a loss of developing germ cells and extensive vacuolization of Sertoli cell cytoplasm. Following cessation of treatment, spermatozoa first appeared in the caput epididymis after 6 weeks and by 12 weeks motile spermatozoa could be recovered from the cauda, although the count (P < 0.001) and motility (P < 0.01) of these cells were significantly reduced and superoxide generation was significantly elevated (P < 0.001). Despite this increase in free radical generation, no evidence of chromatin instability was detected in these spermatozoa. Furthermore, embryos obtained from females mated at this 12-week time point showed no evidence of an increased mutational load. Similarly, progressive ageing of Big Blue mice had no impact on the quality of the spermatozoa, fertility or mutation frequency in the offspring despite a significant increase in the mutational load carried by somatic tissues such as the liver (P < 0.05). We conclude that the male germ line is highly resistant to mutation in keeping with the disposable soma hypothesis, which posits that genetic integrity in the germ cells will be maintained at the expense of the soma, in light of the former's sentinel position in safeguarding the stability of the genome.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Genome/physiology , Germ Cells/drug effects , Mutation/genetics , Aging , Animals , Azoospermia/chemically induced , Bleomycin/adverse effects , Chromatin/drug effects , Cisplatin/adverse effects , Etoposide/adverse effects , Female , Fertility , Genome/drug effects , Male , Mice , Pregnancy , Spermatozoa/drug effects , Spermatozoa/physiology , Spermatozoa/ultrastructure
11.
Mol Cell Biochem ; 428(1-2): 179-191, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28120211

ABSTRACT

Currently, azoospermia is one of the most common diseases of male infertility. Stem cell research is the new hope for novel therapy with a higher degree of safety and lower cost. This study aimed to investigate the effect of umbilical cord blood-derived stem cells (" and mesenchymal "UCB-MSCs") and mono-cell layer implanted into the induced azoospermic mice testis. Stem cells were isolated from umbilical cord blood and CD34+ve cells were separated from negative one by Mini MACs column. At 5th week after single injection of busulfan, stained mesenchymal (CD34-ve), hematopoietic stem cells (CD34+ve) and their conjugate (mono-cell layer) were injected locally into testis. At the end of the study, MSCs group showed that mRNA levels of genes related to meiosis (Vasa, SCP3, and PgK2) were increased with significant decrease of FSH and LH levels, compared to control group. Histologically, most of the tubules restored normal architecture. In contrast, HSCs and mono-cell layer groups showed statically insignificant change of FSH, LH, and gene expression, compared to control group. Histologically, distorted seminiferous tubules, with reduction in sperm content, and interstitial mononuclear cellular infiltration were seen. There was significant increase in the optical density of PCNA immune reaction in MSCs group than azoospermia, HSCs, and mono-cell layer, while there was non-significant difference between MSCs and control group. The present study suggested that injection of MSCs into chemotherapeutic-induced azoospermia in mice improved testicular failure; histologically and functionally, by restoration of spermatogenic gene expression while HSC and mono-cell layer showed no effect on spermatogenesis added to that mono-cell layer may induce testicular tissue damage.


Subject(s)
Antigens, CD34/metabolism , Azoospermia/therapy , Cord Blood Stem Cell Transplantation/methods , Fetal Blood/cytology , Meiosis , Animals , Azoospermia/chemically induced , Azoospermia/genetics , Disease Models, Animal , Fetal Blood/immunology , Follicle Stimulating Hormone/metabolism , Gene Expression Regulation , Humans , Luteinizing Hormone/metabolism , Male , Mice
12.
Ecotoxicol Environ Saf ; 144: 450-455, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28667856

ABSTRACT

Certain genetic background (mainly Y chromosome haplogroups, Y-hg) may modify the susceptibility of certain environmental exposure to some diseases. Compared with respective main effects of genetic background or environmental exposure, interactions between them reflect more realistic combined effects on the susceptibility to a disease. To identify the interactions on spermatogenic impairment, we performed Y chromosome haplotyping and measurement of 9 urinary phenols concentrations in 774 infertile males and 520 healthy controls in a Han Chinese population, and likelihood ratio tests were used to examine the interactions between Y-hgs and phenols. Originally, we observed that Y-hg C and Y-hg F* might modify the susceptibility to male infertility with urinary 4-n-octylphenol (4-n-OP) level (Pinter = 0.005 and 0.019, respectively). Subsequently, based on our results, two panels were tested to identify the possible protective sub-branches of Y-hg F* to 4-n-OP exposure, and Y-hg O3* was uncovered to interact with 4-n-OP (Pinter = 0.019). In conclusion, while 4-n-OP shows an adverse effect on spermatogenesis, Y-hg O3* makes individuals more adaptive to such an effect for maintaining basic reproductive capacity.


Subject(s)
Chromosomes, Human, Y/genetics , Environmental Pollutants/toxicity , Infertility, Male/chemically induced , Phenols/toxicity , Spermatogenesis/drug effects , Adult , Asian People/genetics , Azoospermia/chemically induced , Azoospermia/genetics , Azoospermia/urine , Case-Control Studies , China , Environmental Pollutants/urine , Gene-Environment Interaction , Genetic Predisposition to Disease , Haplotypes , Humans , Infertility, Male/genetics , Infertility, Male/urine , Male , Phenols/urine , Spermatogenesis/genetics
13.
Int J Clin Oncol ; 21(6): 1167-1171, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27306218

ABSTRACT

BACKGROUND: Combinations of surgery, radiation therapy, and chemotherapy can achieve high remission rates in patients with cancer, but these treatments can have damaging effects on spermatogenesis. In particular, cytotoxic chemotherapy may lead to irreversible spermatogenic dysfunction. Microdissection testicular sperm extraction (micro-TESE) is the only method that can address infertility in cancer survivors with persistent postchemotherapy azoospermia. METHODS: We included 66 Japanese patients with postchemotherapy azoospermia who underwent micro-TESE for sperm retrieval in this analysis. Age, oncology data, hormone profiles, and outcomes of micro-TESE and subsequent intracytoplasmic sperm injections (ICSIs) were reviewed. RESULTS: The common disease in our patients was testicular cancer (21 patients), followed by acute lymphoblastic leukemia and Hodgkin's lymphoma (nine patients). In this cohort of 66 patients, sperm was successfully retrieved in 31 patients (47 %), and clinical pregnancy occurred in 23 cases (35 %). The live birth rate was 27 %. No significant differences in sperm retrieval, clinical pregnancy, and live birth rates were seen between testicular cancer, Hodgkin's lymphoma, non-Hodgkin's lymphoma, acute lymphoblastic leukemia, acute myeloid leukemia, or sarcoma cases. Multiple logistic regression analysis showed that the chance of retrieving sperm during micro-TESE could not be predicted by any variable. CONCLUSIONS: Cryopreservation of sperm should be offered before any gonadotoxic chemotherapy takes place. However, micro-TESE and subsequent ICSI could be effective treatment options for patients with persistent postchemotherapy azoospermia whose sperm were not frozen before therapy. Our results suggest that micro-TESE-ICSI could benefit 27 % of such Japanese patients.


Subject(s)
Antineoplastic Agents , Azoospermia , Cryopreservation , Infertility, Male , Neoplasms/drug therapy , Sperm Injections, Intracytoplasmic/methods , Sperm Retrieval , Adult , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Azoospermia/chemically induced , Azoospermia/complications , Azoospermia/diagnosis , Azoospermia/epidemiology , Cryopreservation/methods , Cryopreservation/statistics & numerical data , Humans , Infertility, Male/diagnosis , Infertility, Male/etiology , Infertility, Male/therapy , Japan/epidemiology , Male , Microdissection/methods , Middle Aged , Neoplasms/classification , Neoplasms/epidemiology , Retrospective Studies , Treatment Outcome
14.
Andrologia ; 48(2): 238-40, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26032021

ABSTRACT

Recurrent ischaemic priapism also known as stuttering priapism is an uncommon form of ischaemic priapism, and its treatment is not yet clearly defined. If left untreated, it may evolve into classic form of acute ischaemic priapism and lead to erectile dysfunction due to fibrosis of corpora cavernosa. Several drugs have been proposed with variable results and only supported with level three or four of evidence. Hormonal therapy such as cyproterone acetate, oestrogen, bicalutamide or Lh-Rh agonist are often effective but can cause side effects such as hypogonadal state and infertility. Other medical options are 5-alpha-reductase and phosphodiesterase-5 inhibitors, ketoconazole, baclofen, digoxin, gabapentin and beta-2-agonist terbutaline. We report the first case of stuttering priapism treated with beta-2-agonist salbutamol.


Subject(s)
Adrenergic beta-2 Receptor Agonists/therapeutic use , Albuterol/therapeutic use , Ischemia/drug therapy , Priapism/drug therapy , Administration, Oral , Androgen Antagonists/adverse effects , Azoospermia/chemically induced , Cyproterone Acetate/adverse effects , Humans , Male , Penis/blood supply , Recurrence , Young Adult
15.
FASEB J ; 28(6): 2566-76, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24599970

ABSTRACT

Testosterone (T), alone or in combination with progestin, provides a promising approach to hormonal male contraception. Its principle relies on enhanced negative feedback of exogenous T to suppress gonadotropins, thereby blocking the testicular T production needed for spermatogenesis, while simultaneously maintaining the extragonadal androgen actions, such as potency and libido, to avoid hypogonadism. A serious drawback of the treatment is that a significant proportion of men do not reach azoospermia or severe oligozoospermia, commensurate with contraceptive efficacy. We tested here, using hypogonadal luteinizing hormone/choriongonadotropin receptor (LHCGR) knockout (LHR(-/-)) mice, the basic principle of the T-based male contraceptive method, that a specific T dose could maintain extragonadal androgen actions without simultaneously activating spermatogenesis. LHR(-/-) mice were treated with increasing T doses, and the responses of their spermatogenesis and extragonadal androgen actions (including gonadotropin suppression and sexual behavior) were assessed. Conspicuously, all dose responses to T were practically superimposable, and no dose of T could be defined that would maintain sexual function and suppress gonadotropins without simultaneously activating spermatogenesis. This finding, never addressed in clinical contraceptive trials, is not unexpected in light of the same androgen receptor mediating androgen actions in all organs. When extrapolated to humans, our findings may jeopardize the current approach to hormonal male contraception and call for more effective means of inhibiting intratesticular T production or action, to achieve consistent spermatogenic suppression.


Subject(s)
Contraception/methods , Spermatogenesis/drug effects , Testosterone/administration & dosage , Animals , Azoospermia/chemically induced , Gonadotropins/antagonists & inhibitors , Gonadotropins/blood , Luteinizing Hormone/genetics , Male , Mice , Mice, Knockout , Receptors, LH/genetics , Sexual Behavior, Animal/drug effects , Testis/drug effects , Testosterone/pharmacology
16.
Clin Exp Obstet Gynecol ; 42(4): 545-6, 2015.
Article in English | MEDLINE | ID: mdl-26411232

ABSTRACT

The authors report a case of transient azoospermia following hydroxymethylglutaryl-coenzyme A reductase (HMGCR) inhibitor rosuvastatin medication for hypercholesterolemia. While a primary infertile couple with oligoasthenospermia was preparing for an in vitro fertilization program, the male partner had been diagnosed with hypercholesterolemia in a medical check-up and prescribed four-week oral administration of rosuvastatin. No motile spermatozoa were found in the ejaculated semen and urine on the day of follicular aspiration. Azoospermia was confirmed by reexamination in weeks 3 and 7. Spermatozoa appeared in the ejaculated semen in two weeks of drug withdrawal. In week 16, the sperm count and motility increased to the level where intracytoplasmic sperm injection was available.


Subject(s)
Azoospermia/diagnosis , Fluorobenzenes/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hypercholesterolemia/drug therapy , Infertility , Pyrimidines/adverse effects , Sulfonamides/adverse effects , Administration, Oral , Adult , Azoospermia/chemically induced , Diagnosis, Differential , Female , Fluorobenzenes/administration & dosage , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Magnetic Resonance Imaging , Male , Middle Aged , Oocyte Retrieval , Pyrimidines/administration & dosage , Rosuvastatin Calcium , Sulfonamides/administration & dosage , Testis/pathology
17.
Zhonghua Nan Ke Xue ; 21(8): 692-7, 2015 Aug.
Article in Zh | MEDLINE | ID: mdl-26442294

ABSTRACT

OBJECTIVE: To study the ability of bone marrow mesenchymal stem cells (BMSCs) to repair the internal environment of the testis in male azoospermia rats. METHODS: We established azoospermia models in 22 six-week-old male SD rats by intraperitoneal injection of busulfan at 20 mg per kg body weight. We transplanted allogeneic rat BMSCs (rBMSCs) into the testicular seminiferous tubules of the model rats and, 30 days after transplantation, observed the composition and structure of the seminiferous tubular cells by HE staining and detected the expressions of CD44, CD106, and c-kit in the rBMSCs by immunohistochemistry. RESULTS: The number of epididymal sperm was significantly reduced in the model rats as compared with the normal controls (P < 0.01). CD44 and CD106, but not c-kit, were expressed in the isolated rBMSCs. At 30 days after transplantation of rBMSCs, lots of new cells were observed in the seminiferous tubules, some expressing CD106 and some expressing the germ cell surface marker c-kit. CONCLUSION: BMSCs can transdifferentiate into germ cells and repair the damaged seminiferous tubules of sterile rats.


Subject(s)
Azoospermia/therapy , Mesenchymal Stem Cell Transplantation , Seminiferous Tubules/anatomy & histology , Animals , Azoospermia/chemically induced , Biomarkers/metabolism , Bone Marrow Cells , Busulfan , Cell Membrane/metabolism , Epididymis , Hyaluronan Receptors/metabolism , Male , Mesenchymal Stem Cells/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Rats , Rats, Sprague-Dawley , Seminiferous Tubules/metabolism , Spermatozoa , Staining and Labeling , Vascular Cell Adhesion Molecule-1/metabolism
18.
Andrologia ; 46(10): 1098-106, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24325627

ABSTRACT

Busulfan is a chemotherapeutic drug that induces sterility, azoospermia and testicular atrophy. To induce degeneration of spermatogenesis, we used different amounts of busulfan. Adult male C57Bl/6 mice were treated with 15, 30 and 45 mg kg(-1) of busulfan. After 5 weeks, animals had daily injections of 7.5 IU human follicle-stimulating hormone (hFSH) and 12.5 µg kg(-1) oestradiol benzoate (EB), separately or simultaneously. After this time, the animals were killed and blood samples were taken through cardiac puncture. Testes were used for histopathology experiments, DNA flow cytometry and RNA extraction for expression of c-kit and cyclin B1 genes. EB unlike FSH has induced stimulatory effects on spermatogenesis, increased the level of serum testosterone 2-fold and caused a 2-fold increase in the number of haploid cells. The result showed that hFSH with EB multiplied EB stimulatory effects on spermatogenesis up to four times. Expression of c-kit and cyclin B1 genes increased in EB and hFSH+EB groups. These findings suggest that EB regulates spermatogonial stem cells via hFSH. hFSH with EB had synergistic effect on regeneration of spermatogenesis.


Subject(s)
Azoospermia/drug therapy , Estradiol/therapeutic use , Follicle Stimulating Hormone/therapeutic use , Spermatogenesis/drug effects , Testis/drug effects , Animals , Azoospermia/chemically induced , Azoospermia/metabolism , Azoospermia/pathology , Busulfan , Cyclin B1/genetics , Cyclin B1/metabolism , Disease Models, Animal , Estradiol/pharmacology , Follicle Stimulating Hormone/pharmacology , Male , Mice , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Testis/metabolism , Testis/pathology , Testosterone/blood
19.
Int J Mol Sci ; 15(8): 13151-65, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-25062349

ABSTRACT

Non-obstructive azoospermia is the most challenging type of male infertility. Stem cell based therapy provides the potential to enhance the recovery of spermatogenesis following cancer therapy. Bone marrow-derived mesenchymal stem cells (BMSCs) possess the potential to differentiate or trans-differentiate into multi-lineage cells, secrete paracrine factors to recruit the resident stem cells to participate in tissue regeneration, or fuse with the local cells in the affected region. In this study, we tested whether spermatogenically-induced BMSCs can restore spermatogenesis after administration of an anticancer drug. Allogeneic BMSCs were co-cultured in conditioned media derived from cultured testicular Sertoli cells in vitro, and then induced stem cells were transplanted into the seminiferous tubules of a busulfan-induced azoospermatic rat model for 8 weeks. The in vitro induced BMSCs exhibited specific spermatogonic gene and protein markers, and after implantation the donor cells survived and located at the basement membranes of the recipient seminiferous tubules, in accordance with what are considered the unique biological characteristics of spermatogenic stem cells. Molecular markers of spermatogonial stem cells and spermatogonia (Vasa, Stella, SMAD1, Dazl, GCNF, HSP90α, integrinß1, and c-kit) were expressed in the recipient testis tissue. No tumor mass, immune response, or inflammatory reaction developed. In conclusion, BMSCs might provide the potential to trans-differentiate into spermatogenic-like-cells, enhancing endogenous fertility recovery. The present study indicates that BMSCs might offer alternative treatment for the patients with azoospermatic infertility after cancer chemotherapy.


Subject(s)
Bone Marrow Cells/cytology , Mesenchymal Stem Cells/cytology , Animals , Antineoplastic Agents/toxicity , Azoospermia/chemically induced , Azoospermia/pathology , Busulfan/toxicity , Cell Transdifferentiation , Cells, Cultured , Coculture Techniques , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Disease Models, Animal , Male , Mesenchymal Stem Cells/metabolism , Rats , Rats, Sprague-Dawley , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sertoli Cells/cytology , Sertoli Cells/metabolism , Sertoli Cells/pathology , Spermatogenesis/drug effects , Spermatogonia/metabolism , Testis/metabolism , Testis/pathology , Transplantation, Homologous
20.
Med Mol Morphol ; 47(1): 21-30, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23512148

ABSTRACT

Neonatal estrogen treatment (NET) induces morphological changes in male reproductive organs. NET with ß-estradiol 17-cypionate is reported to induce inflammation with stromal-epithelial abnormalities in the prostate and seminal vesicles in post-pubertal mice. The aim of this study was to investigate the histopathology of the testis, ductuli efferentes, epididymis, and vas deferens in mice after NET with ß-estradiol 17-cypionate. No morphological changes in these organs were observed until 4 weeks after NET. However, some inflammatory cells were found in epididymis and vas deferens 6 weeks after NET. Eight weeks after NET, inflammatory cells spread to the ductuli efferentes and inflammation was severe from 6 to 12 weeks after NET. Inflammatory cells were never seen in the whole testis, but cystic dilatation of the rete testes with spermatogenic disturbance was found around the mediastinum testis. Many inflammatory cells emigrated into the lumen of the epididymis, resulting in complete absence of spermatozoa in the vas deferens. Most of the inflammatory cells penetrating into the epithelial layers of epididymal ducts were neutrophils. These results indicate that in post-pubertal mice, NET with ß-estradiol 17-cypionate induces inflammation in the ductuli efferentes, epididymis, and vas deferens, but not in the testis, provoking obstructive azoospermia.


Subject(s)
Azoospermia/chemically induced , Epididymis/drug effects , Estradiol/analogs & derivatives , Inflammation/chemically induced , Vas Deferens/drug effects , Animals , Animals, Newborn , Azoospermia/pathology , Ejaculatory Ducts/drug effects , Ejaculatory Ducts/pathology , Epididymis/pathology , Epididymis/ultrastructure , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/ultrastructure , Estradiol/toxicity , Female , Infertility, Male/chemically induced , Infertility, Male/pathology , Inflammation/pathology , Male , Mice , Mice, Inbred BALB C , Microscopy, Electron , Neutrophil Infiltration/drug effects , Sexual Maturation , Spermatogenesis/drug effects , Testis/drug effects , Testis/pathology , Testis/ultrastructure , Time Factors , Vas Deferens/pathology , Vas Deferens/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL