Célia Tebbakh, Anne-Laure Barbotin, Guillaume Martinez, Angèle Boursier, Zeina Wehbe, Asma Hammouda, Nicolas Thierry-Mieg, Christophe Arnoult, Selima Fourati Ben Mustapha, Raoudha Zouari, Pierre F Ray, Zine-Eddine Kherraf
Background: Asthenozoospermia, characterized by reduced sperm motility, is a common cause of male infertility. Multiple morphological abnormalities of the sperm flagella (MMAF) represent a severe and genetically heterogeneous form of asthenozoospermia. Over 50 genes have been associated, but approximately half of MMAF cases remain unexplained. DRC1, a gene involved in the nexin-dynein regulatory complex (N-DRC), has been linked to MMAF and primary ciliary dyskinesia (PCD), often with significant variability in clinical presentation.
Objectives: His study aimed to identify novel pathogenic DRC1 variants in MMAF patients, assess their impact on sperm flagellar structure, and evaluate intracytoplasmic sperm injection (ICSI) and pregnancy outcomes.
Materials and methods: A cohort of 196 non-syndromic MMAF patients was analyzed using whole exome sequencing (WES). Functional validation of candidate variants included immunofluorescence to assess protein expression and transmission electron microscopy (TEM) to identify ultrastructural abnormalities. Assisted reproductive therapy outcomes were also evaluated.
Results: WES identified a recurrent homozygous frameshift variant in DRC1 NM_145038.5: c.109dup; p.(Gln37ProfsTer30) in four patients (2%), all of North African origin, none of whom suffer from PCD-related symptoms. The variant caused a complete absence of DRC1 protein in spermatozoa. TEM showed flagellar abnormalities, with 10% of axonemal sections revealing peripheral doublet dissociation, suggesting N-DRC instability. ICSI resulted in a 68.5% fertilization rate, with three out of four couples successfully delivering healthy children.
Discussion and conclusion: The identification of a novel and recurrent pathogenic DRC1 variant broadens the mutation spectrum associated with MMAF. The absence of systemic PCD symptoms suggests that DRC1 deficiency may primarily affect spermatogenesis. Notably, the phenotypic spectrum might be influenced by the genetic background, varying across populations. Favorable ICSI outcomes, with a 68.5% fertilization rate and successful pregnancies in three out of four couples, highlight the effectiveness of assisted reproductive techniques for patients with this genetic defect.
{"title":"A recurrent loss-of-function variant in DRC1 causes non-syndromic severe asthenozoospermia with favorable intracytoplasmic sperm injection and pregnancy outcomes.","authors":"Célia Tebbakh, Anne-Laure Barbotin, Guillaume Martinez, Angèle Boursier, Zeina Wehbe, Asma Hammouda, Nicolas Thierry-Mieg, Christophe Arnoult, Selima Fourati Ben Mustapha, Raoudha Zouari, Pierre F Ray, Zine-Eddine Kherraf","doi":"10.1111/andr.13837","DOIUrl":"https://doi.org/10.1111/andr.13837","url":null,"abstract":"<p><strong>Background: </strong>Asthenozoospermia, characterized by reduced sperm motility, is a common cause of male infertility. Multiple morphological abnormalities of the sperm flagella (MMAF) represent a severe and genetically heterogeneous form of asthenozoospermia. Over 50 genes have been associated, but approximately half of MMAF cases remain unexplained. DRC1, a gene involved in the nexin-dynein regulatory complex (N-DRC), has been linked to MMAF and primary ciliary dyskinesia (PCD), often with significant variability in clinical presentation.</p><p><strong>Objectives: </strong>His study aimed to identify novel pathogenic DRC1 variants in MMAF patients, assess their impact on sperm flagellar structure, and evaluate intracytoplasmic sperm injection (ICSI) and pregnancy outcomes.</p><p><strong>Materials and methods: </strong>A cohort of 196 non-syndromic MMAF patients was analyzed using whole exome sequencing (WES). Functional validation of candidate variants included immunofluorescence to assess protein expression and transmission electron microscopy (TEM) to identify ultrastructural abnormalities. Assisted reproductive therapy outcomes were also evaluated.</p><p><strong>Results: </strong>WES identified a recurrent homozygous frameshift variant in DRC1 NM_145038.5: c.109dup; p.(Gln37ProfsTer30) in four patients (2%), all of North African origin, none of whom suffer from PCD-related symptoms. The variant caused a complete absence of DRC1 protein in spermatozoa. TEM showed flagellar abnormalities, with 10% of axonemal sections revealing peripheral doublet dissociation, suggesting N-DRC instability. ICSI resulted in a 68.5% fertilization rate, with three out of four couples successfully delivering healthy children.</p><p><strong>Discussion and conclusion: </strong>The identification of a novel and recurrent pathogenic DRC1 variant broadens the mutation spectrum associated with MMAF. The absence of systemic PCD symptoms suggests that DRC1 deficiency may primarily affect spermatogenesis. Notably, the phenotypic spectrum might be influenced by the genetic background, varying across populations. Favorable ICSI outcomes, with a 68.5% fertilization rate and successful pregnancies in three out of four couples, highlight the effectiveness of assisted reproductive techniques for patients with this genetic defect.</p>","PeriodicalId":7898,"journal":{"name":"Andrology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142942848","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Across six decades, androgenetics has consistently concentrated on discovering genetic causes and enhancing the molecular diagnostics of male infertility, disorders of sex development, and their broader implications on health, such as cancer and other comorbidities. Despite vast clinical knowledge, the training of andrologists often lacks fundamental basics in medical genetics. This work, as part of the Special Issue of Andrology "Genetics in Andrology", provides the core terminology in medical genetics and technological advancements in genomics, required to understand the ever-progressing research in the field. It also gives an overview of study designs and approaches that have frequently led to discoveries in androgenetics. The rapid progress in the methodological toolbox in human genetics is illustrated by numerous examples of impactful androgenetic studies over 60 years, and their clinical implications.
{"title":"Introduction to androgenetics: terminology, approaches, and impactful studies across 60 years.","authors":"Arvand Akbari, Laura Kasak, Maris Laan","doi":"10.1111/andr.13835","DOIUrl":"https://doi.org/10.1111/andr.13835","url":null,"abstract":"<p><p>Across six decades, androgenetics has consistently concentrated on discovering genetic causes and enhancing the molecular diagnostics of male infertility, disorders of sex development, and their broader implications on health, such as cancer and other comorbidities. Despite vast clinical knowledge, the training of andrologists often lacks fundamental basics in medical genetics. This work, as part of the Special Issue of Andrology \"Genetics in Andrology\", provides the core terminology in medical genetics and technological advancements in genomics, required to understand the ever-progressing research in the field. It also gives an overview of study designs and approaches that have frequently led to discoveries in androgenetics. The rapid progress in the methodological toolbox in human genetics is illustrated by numerous examples of impactful androgenetic studies over 60 years, and their clinical implications.</p>","PeriodicalId":7898,"journal":{"name":"Andrology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142942849","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<p><strong>Background: </strong>Androgen deprivation is associated with erectile dysfunction (ED). In different animal models, sulfur dioxide (SO<sub>2</sub>) donors Na<sub>2</sub>SO<sub>3</sub> and NaHSO<sub>3</sub> reduced oxidative stress, fibrosis, and inflammation which contribute to the pathogenesis of androgen deprivation-induced ED, however the effect of SO<sub>2</sub> donors on ED in castrated rats were not known.</p><p><strong>Objective: </strong>To investigate the therapeutic effect of SO<sub>2</sub> donors, Na<sub>2</sub>SO<sub>3</sub>/NaHSO<sub>3</sub>, on ED in castrated rat model.</p><p><strong>Materials and methods: </strong>Sprague-Dawley male rats (n = 30) were divided into four groups; control, control-treated with Na<sub>2</sub>SO<sub>3</sub>/NaHSO<sub>3</sub>, castrated, and castrated-treated with Na<sub>2</sub>SO<sub>3</sub>/NaHSO<sub>3</sub>. Castration was induced by bilateral scrotal incisions. Four weeks after castration, rats were treated with Na<sub>2</sub>SO<sub>3</sub>/NaHSO<sub>3</sub> (0.54/0.18 mmol/kg) intraperitoneally (i.p.) for 4 weeks. Intracavernosal pressure/mean arterial pressure ratio (ICP/MAP) and total ICP were measured to evaluate in vivo erectile responses in cavernosal tissue. In vitro relaxant and contractile responses were measured in all groups. Endothelial nitric oxide synthase (eNOS), neuronal NOS (nNOS), PI3 kinase p85 alpha + gamma (PI3K), protein kinase B (AKT 1/2/3), cysteine dioxygenase-1 (CDO), and aspartate aminotransferase (AAT) expressions and localizations were evaluated by Western blotting and immunohistochemical staining. The smooth muscle/collagen ratio was evaluated by Masson's trichrome staining.</p><p><strong>Results: </strong>Prostate (p < 0.001) and penis weight (p < 0.001), total serum testosterone (T) level (p < 0.001), and in vivo erectile responses (p < 0.001 at 7.5 and 5 V, p < 0.05 at 2.5 V for ICP/MAP and total ICP) of castrated rats were decreased compared with control. SO<sub>2</sub> donors improved reduced ICP/MAP ratio and total ICP (p < 0.01 at 7.5, 5, and 2.5 V for ICP/MAP and total ICP) nitrergic (p < 0.05 at 20 Hz), and endothelium-independent relaxation (p < 0.05 at 1 nM, p < 0.01 at 10 µM and 100 µM) in the castrated group. Decreased eNOS (p < 0.01) and AKT (p < 0.001) protein expressions in the castrated group were normalized by SO<sub>2</sub>. SO<sub>2</sub> donors partially restored the reduced smooth muscle/collagen ratio in the castrated group (p < 0.001). The expressions and locations of nNOS, PI3K, CDO, and AAT proteins in penile tissue did not alter among all groups (p > 0.05).</p><p><strong>Discussion and conclusion: </strong>SO<sub>2</sub> donors significantly improve erectile functions and relaxation responses in a castrated rats via ameliorating endothelial damage and fibrosis. Androgen deprivation inhibits the AKT/eNOS signaling while SO<sub>2</sub> activates this pathway. SO<sub>2</sub> donors may be promising for the treatment of ED in hypoandrog
{"title":"Sulfur dioxide (SO<sub>2</sub>) donors, a new gasotransmitter, improve erectile dysfunction after castration in a rat model.","authors":"Seyma Tetik-Rama, Didem Yilmaz-Oral, Damla Turkcan, Cetin Volkan Oztekin, Omer Faruk Kirlangic, Fatma Zeynep Kirlangic, Serap Gur","doi":"10.1111/andr.13839","DOIUrl":"https://doi.org/10.1111/andr.13839","url":null,"abstract":"<p><strong>Background: </strong>Androgen deprivation is associated with erectile dysfunction (ED). In different animal models, sulfur dioxide (SO<sub>2</sub>) donors Na<sub>2</sub>SO<sub>3</sub> and NaHSO<sub>3</sub> reduced oxidative stress, fibrosis, and inflammation which contribute to the pathogenesis of androgen deprivation-induced ED, however the effect of SO<sub>2</sub> donors on ED in castrated rats were not known.</p><p><strong>Objective: </strong>To investigate the therapeutic effect of SO<sub>2</sub> donors, Na<sub>2</sub>SO<sub>3</sub>/NaHSO<sub>3</sub>, on ED in castrated rat model.</p><p><strong>Materials and methods: </strong>Sprague-Dawley male rats (n = 30) were divided into four groups; control, control-treated with Na<sub>2</sub>SO<sub>3</sub>/NaHSO<sub>3</sub>, castrated, and castrated-treated with Na<sub>2</sub>SO<sub>3</sub>/NaHSO<sub>3</sub>. Castration was induced by bilateral scrotal incisions. Four weeks after castration, rats were treated with Na<sub>2</sub>SO<sub>3</sub>/NaHSO<sub>3</sub> (0.54/0.18 mmol/kg) intraperitoneally (i.p.) for 4 weeks. Intracavernosal pressure/mean arterial pressure ratio (ICP/MAP) and total ICP were measured to evaluate in vivo erectile responses in cavernosal tissue. In vitro relaxant and contractile responses were measured in all groups. Endothelial nitric oxide synthase (eNOS), neuronal NOS (nNOS), PI3 kinase p85 alpha + gamma (PI3K), protein kinase B (AKT 1/2/3), cysteine dioxygenase-1 (CDO), and aspartate aminotransferase (AAT) expressions and localizations were evaluated by Western blotting and immunohistochemical staining. The smooth muscle/collagen ratio was evaluated by Masson's trichrome staining.</p><p><strong>Results: </strong>Prostate (p < 0.001) and penis weight (p < 0.001), total serum testosterone (T) level (p < 0.001), and in vivo erectile responses (p < 0.001 at 7.5 and 5 V, p < 0.05 at 2.5 V for ICP/MAP and total ICP) of castrated rats were decreased compared with control. SO<sub>2</sub> donors improved reduced ICP/MAP ratio and total ICP (p < 0.01 at 7.5, 5, and 2.5 V for ICP/MAP and total ICP) nitrergic (p < 0.05 at 20 Hz), and endothelium-independent relaxation (p < 0.05 at 1 nM, p < 0.01 at 10 µM and 100 µM) in the castrated group. Decreased eNOS (p < 0.01) and AKT (p < 0.001) protein expressions in the castrated group were normalized by SO<sub>2</sub>. SO<sub>2</sub> donors partially restored the reduced smooth muscle/collagen ratio in the castrated group (p < 0.001). The expressions and locations of nNOS, PI3K, CDO, and AAT proteins in penile tissue did not alter among all groups (p > 0.05).</p><p><strong>Discussion and conclusion: </strong>SO<sub>2</sub> donors significantly improve erectile functions and relaxation responses in a castrated rats via ameliorating endothelial damage and fibrosis. Androgen deprivation inhibits the AKT/eNOS signaling while SO<sub>2</sub> activates this pathway. SO<sub>2</sub> donors may be promising for the treatment of ED in hypoandrog","PeriodicalId":7898,"journal":{"name":"Andrology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142942860","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Vivian Fuguhara, Gabriel Augusto Oliveira Stocco, José Britto-Júnior, Luiz Fernando Ribeiro, João Figueira Scarini, Fernanda Viviane Mariano, Valéria B de Souza, Andre Almeida Schenka, Edson Antunes, Gilberto De Nucci
Background: 6-Nitrodopamine (6-ND) released from rat vas deferens acts an endogenous modulator of vas deferens contractility.
Objectives: To investigate whether rat isolated seminal vesicles (RISV) releases 6-ND, the mechanisms involved in the release, and the modulatory role of 6-ND on tissue contractility.
Methods: Rat seminal vesicles were removed and placed in Krebs-Henseleit's solution at 37°C for 30 min, and an aliquot was used to analyze the concentrations of 6-ND, dopamine, noradrenaline, and adrenaline by liquid chromatography with tandem mass spectrometry (LC-MS/MS). The effect of mechanical removal of the epithelium and the effects of pre-incubation of RISV strips with Nω-nitro-L-arginine methyl ester (L-NAME; in combination or not with L-arginine), tetrodotoxin (TTX), GKT137831, and hydrogen peroxide (H2O2) on 6-ND release were evaluated. For functional studies, RISV strips were mounted in an organ bath and tied to an isometric force transducer. The expressions of tyrosine hydroxylase and endothelial nitric oxide synthase (eNOS, type III NOS) were investigated by immunohistochemistry and/or fluorescence in situ hybridization (FISH).
Results: 6-ND was the major released catecholamine from RISV strips compared with noradrenaline, adrenaline, and dopamine. Epithelium removal significantly reduced the release of 6-ND, noradrenaline and dopamine. In RISV strips obtained from animals chronically treated with L-NAME, the 6-ND release significantly reduced. Pre-incubation with L-NAME reduced 6-ND release, which was partly restored by co-incubation with L-arginine. Pre-incubation with TTX had no effect on the release of any catecholamine, whereas GKT137831 and H2O2 significantly increased 6-ND release. All catecholamines produced concentration-dependent RISV contractions, but 6-ND was approximately 30× less potent than the others. 6-ND (0.1 nM) significantly potentiated noradrenaline-, adrenaline-, and dopamine-induced contractions, with such potentiations inhibited by TTX. Immunohistochemistry and FISH assays in RISV tissues identified tyrosine hydroxylase in epithelial cells and eNOS expression in both epithelial and endothelial cells.
Discussion and conclusion: This is the first demonstration that epithelial-derived 6-ND modulates rat seminal vesicle contractility.
背景:大鼠输精管释放的6-硝基多巴胺(6-ND)是输精管收缩性的内源性调节剂。目的:研究大鼠离体精囊(RISV)是否释放6-ND、释放机制以及6-ND对组织收缩性的调节作用。方法:取大鼠精囊,置于Krebs-Henseleit溶液中,37℃保存30 min,用液相色谱-串联质谱法(LC-MS/MS)分析6-ND、多巴胺、去甲肾上腺素和肾上腺素的浓度。机械去除上皮的作用及n ω-硝基- l -精氨酸甲酯(L-NAME)预孵育RISV条的效果;分别与l -精氨酸、河豚毒素(TTX)、GKT137831和过氧化氢(H2O2)联用或不联用对6-ND的释放进行了评价。对于功能研究,RISV条带被安装在一个器官浴中,并绑在一个等距力传感器上。采用免疫组织化学和/或荧光原位杂交(FISH)方法检测酪氨酸羟化酶和内皮型一氧化氮合酶(eNOS, III型NOS)的表达。结果:与去甲肾上腺素、肾上腺素和多巴胺相比,RISV条释放的儿茶酚胺主要为6-ND。上皮切除显著降低6-ND、去甲肾上腺素和多巴胺的释放。在长期用L-NAME处理的RISV条中,6-ND的释放明显减少。与L-NAME共孵育可减少6-ND释放,与l -精氨酸共孵育可部分恢复6-ND释放。TTX对儿茶酚胺的释放没有影响,而GKT137831和H2O2显著增加了6-ND的释放。所有儿茶酚胺都产生浓度依赖性的RISV收缩,但6-ND的效力比其他儿茶酚胺低约30倍。6-ND (0.1 nM)显著增强去甲肾上腺素、肾上腺素和多巴胺诱导的收缩,这种增强作用被TTX抑制。RISV组织的免疫组织化学和FISH检测发现上皮细胞中有酪氨酸羟化酶,上皮细胞和内皮细胞中都有eNOS表达。讨论与结论:这是上皮源性6-ND调节大鼠精囊收缩性的首次证明。
{"title":"6-Nitrodopamine is an endogenous mediator of rat seminal vesicles contractility.","authors":"Vivian Fuguhara, Gabriel Augusto Oliveira Stocco, José Britto-Júnior, Luiz Fernando Ribeiro, João Figueira Scarini, Fernanda Viviane Mariano, Valéria B de Souza, Andre Almeida Schenka, Edson Antunes, Gilberto De Nucci","doi":"10.1111/andr.13836","DOIUrl":"https://doi.org/10.1111/andr.13836","url":null,"abstract":"<p><strong>Background: </strong>6-Nitrodopamine (6-ND) released from rat vas deferens acts an endogenous modulator of vas deferens contractility.</p><p><strong>Objectives: </strong>To investigate whether rat isolated seminal vesicles (RISV) releases 6-ND, the mechanisms involved in the release, and the modulatory role of 6-ND on tissue contractility.</p><p><strong>Methods: </strong>Rat seminal vesicles were removed and placed in Krebs-Henseleit's solution at 37°C for 30 min, and an aliquot was used to analyze the concentrations of 6-ND, dopamine, noradrenaline, and adrenaline by liquid chromatography with tandem mass spectrometry (LC-MS/MS). The effect of mechanical removal of the epithelium and the effects of pre-incubation of RISV strips with N<sup>ω</sup>-nitro-L-arginine methyl ester (L-NAME; in combination or not with L-arginine), tetrodotoxin (TTX), GKT137831, and hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>) on 6-ND release were evaluated. For functional studies, RISV strips were mounted in an organ bath and tied to an isometric force transducer. The expressions of tyrosine hydroxylase and endothelial nitric oxide synthase (eNOS, type III NOS) were investigated by immunohistochemistry and/or fluorescence in situ hybridization (FISH).</p><p><strong>Results: </strong>6-ND was the major released catecholamine from RISV strips compared with noradrenaline, adrenaline, and dopamine. Epithelium removal significantly reduced the release of 6-ND, noradrenaline and dopamine. In RISV strips obtained from animals chronically treated with L-NAME, the 6-ND release significantly reduced. Pre-incubation with L-NAME reduced 6-ND release, which was partly restored by co-incubation with L-arginine. Pre-incubation with TTX had no effect on the release of any catecholamine, whereas GKT137831 and H<sub>2</sub>O<sub>2</sub> significantly increased 6-ND release. All catecholamines produced concentration-dependent RISV contractions, but 6-ND was approximately 30× less potent than the others. 6-ND (0.1 nM) significantly potentiated noradrenaline-, adrenaline-, and dopamine-induced contractions, with such potentiations inhibited by TTX. Immunohistochemistry and FISH assays in RISV tissues identified tyrosine hydroxylase in epithelial cells and eNOS expression in both epithelial and endothelial cells.</p><p><strong>Discussion and conclusion: </strong>This is the first demonstration that epithelial-derived 6-ND modulates rat seminal vesicle contractility.</p>","PeriodicalId":7898,"journal":{"name":"Andrology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142942847","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Endocannabinoids like anandamide (AEA), among other lipids, are recognized signaling molecules that participate in reproductive events.
Objectives: Our aims were to characterize orphan G protein-coupled receptor (GPR55) presence; investigate GPR55 activation by AEA and determine GPR55 role in the bovine sperm function.
Materials and methods: GPR55 presence was assessed by immunocytochemistry. Protein kinase A (pPKA) and PKC (pPKC) substrates, pERK1/2, G/F-actin were determined by Western blotting, activation of RAC-1 by pull-down assay, F-actin and acrosomal exocytosis by fluorescence microscopy, sperm motility by optic microscopy and computer-aided sperm analysis and fertilizing ability by in vitro fertilization (IVF).
Results: We detected GPR55 in spermatozoa at T0, after incubation in non-capacitating and capacitating (presence of AEA) conditions and upon release from oviductal epithelia. AEA induced an increase in pPKA and pPKC, while CID16020046 (CID), selective GPR55 antagonist, prevented this effect. Incubation with H89, PKA inhibitor, significantly decreased pPKC, while Gö6983, a PKC inhibitor, partially reduced pPKA. pPKA remained elevated at 15- and 45-min incubation, while pPKC decreased at 15 and increased at 45 min. CID prevented pPKC increase at 5 and 45 min and decreased pPKA at 45 min. RAC-1 and F-actin increase induced by AEA was prevented by CID. Variations in two progressive motility kinematic parameters were observed with AEA and/or CID. Sperm pretreatment with AEA increased the rate of cleaved embryos and CID prevented this effect.
Discussion: We demonstrated that GPR55 activation by AEA induces time-dependent signaling pathways involving pPKA and pPKC during bovine sperm capacitation. AEA regulates actin polymerization through GPR55 activation, suggesting the receptor participates in cytoskeleton remodeling, and yielded higher IVF rates. Also, sperm pre-incubation with molecules like AEA involved in capacitation could improve the embryo development.
Conclusion: We have demonstrated GPR55 presence in bovine spermatozoa. The regulation of PKA and PKC and of molecules associated with cytoskeletal dynamics, such as RAC-1 and actin, by GPR55 is closely related to sperm motility and acrosomal exocytosis.
{"title":"Role of GPR55 receptor in bovine sperm capacitation.","authors":"Raquel Lottero-Leconte, Angela Lara, Jessica Plaza, Camila Arroyo-Salvo, María Eugenia Bogetti, Amada Eugenia Ynsaurralde Rivolta, Franco Dellavalle, Fiamma Sengiali, Pablo Cetica, Sofía Rio, Lucia Zalazar, Andreína Cesari, Marcelo Miragaya, Sergio Morado, Silvina Perez-Martinez","doi":"10.1111/andr.13823","DOIUrl":"https://doi.org/10.1111/andr.13823","url":null,"abstract":"<p><strong>Background: </strong>Endocannabinoids like anandamide (AEA), among other lipids, are recognized signaling molecules that participate in reproductive events.</p><p><strong>Objectives: </strong>Our aims were to characterize orphan G protein-coupled receptor (GPR55) presence; investigate GPR55 activation by AEA and determine GPR55 role in the bovine sperm function.</p><p><strong>Materials and methods: </strong>GPR55 presence was assessed by immunocytochemistry. Protein kinase A (pPKA) and PKC (pPKC) substrates, pERK1/2, G/F-actin were determined by Western blotting, activation of RAC-1 by pull-down assay, F-actin and acrosomal exocytosis by fluorescence microscopy, sperm motility by optic microscopy and computer-aided sperm analysis and fertilizing ability by in vitro fertilization (IVF).</p><p><strong>Results: </strong>We detected GPR55 in spermatozoa at T0, after incubation in non-capacitating and capacitating (presence of AEA) conditions and upon release from oviductal epithelia. AEA induced an increase in pPKA and pPKC, while CID16020046 (CID), selective GPR55 antagonist, prevented this effect. Incubation with H89, PKA inhibitor, significantly decreased pPKC, while Gö6983, a PKC inhibitor, partially reduced pPKA. pPKA remained elevated at 15- and 45-min incubation, while pPKC decreased at 15 and increased at 45 min. CID prevented pPKC increase at 5 and 45 min and decreased pPKA at 45 min. RAC-1 and F-actin increase induced by AEA was prevented by CID. Variations in two progressive motility kinematic parameters were observed with AEA and/or CID. Sperm pretreatment with AEA increased the rate of cleaved embryos and CID prevented this effect.</p><p><strong>Discussion: </strong>We demonstrated that GPR55 activation by AEA induces time-dependent signaling pathways involving pPKA and pPKC during bovine sperm capacitation. AEA regulates actin polymerization through GPR55 activation, suggesting the receptor participates in cytoskeleton remodeling, and yielded higher IVF rates. Also, sperm pre-incubation with molecules like AEA involved in capacitation could improve the embryo development.</p><p><strong>Conclusion: </strong>We have demonstrated GPR55 presence in bovine spermatozoa. The regulation of PKA and PKC and of molecules associated with cytoskeletal dynamics, such as RAC-1 and actin, by GPR55 is closely related to sperm motility and acrosomal exocytosis.</p>","PeriodicalId":7898,"journal":{"name":"Andrology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142920561","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Mustfa Kabi, Aditi Khamamkar, Kwaku Kyei-Baffour, Michel Weïwer, Srinivasan Vijayaraghavan, Souvik Dey
Background and objectives: Epididymal transit renders key competence to mammalian spermatozoa for fertilizing eggs. Generally, the two paralogs of glycogen synthase kinase 3, GSK3α and GSK3β, functionally overlap except in testis and sperm. We showed that GSK3α is essential for epididymal sperm maturation and fertilization. Male infertility is the only phenotype of mice with a global or testis-specific knockout (KO) of Gsk3α. Their sperm maturation is impaired, and sperm cannot fertilize eggs in vitro and in vivo. This suggests that GSK3α is a "male fertility kinase" in mammals and that GSK3α-selective inhibitor is a potential male contraceptive.
Materials and methods: A set of eight heterozygous Gsk3α(±) male mice received daily intraperitoneal injections of BRD0705, an isoform-selective GSK3α inhibitor, at 20 mg/kg body weight for 1 week. Five vehicle-treated and BRD0705-treated mice were tested for in vivo fertility and the remaining mice were sacrificed; their caudal spermatozoa were examined for motility and biochemical properties.
Results: The treated mice did not sire any pups while the control group sired 46 pups with a normal gestation period of 19-23 days. Continued fertility testing up to 6 weeks post-treatment, showed that the treated mice regained fertility siring 56 pups, with 76 in the control group. Sperm motility was impaired, its abnormal morphology increased during epididymal transit, Adenosine triphosphate (ATP) levels were low, and tyrosine-phosphorylation of hexokinase was absent: these phenotypes imitated those observed in Gsk3α KO mice. Tyrosine279-phosphorylation of GSK3α was reduced in sperm from the treated mice showing that the GSK3α activity was inhibited. The altered sperm phenotypes returned to normal following recovery of fertility.
Conclusions: Complete infertility resulted after 1 week of BRD0705-treatment and fertility recovered after cessation of the treatment. Work is ongoing to determine the minimum dose and treatment time and the testing of new compounds with increased selectivity and inhibitory activity against GSK3α.
{"title":"A non-hormonal reversible contraceptive targeting GSK3α, a protein kinase, essential for epididymal sperm maturation.","authors":"Mustfa Kabi, Aditi Khamamkar, Kwaku Kyei-Baffour, Michel Weïwer, Srinivasan Vijayaraghavan, Souvik Dey","doi":"10.1111/andr.13838","DOIUrl":"https://doi.org/10.1111/andr.13838","url":null,"abstract":"<p><strong>Background and objectives: </strong>Epididymal transit renders key competence to mammalian spermatozoa for fertilizing eggs. Generally, the two paralogs of glycogen synthase kinase 3, GSK3α and GSK3β, functionally overlap except in testis and sperm. We showed that GSK3α is essential for epididymal sperm maturation and fertilization. Male infertility is the only phenotype of mice with a global or testis-specific knockout (KO) of Gsk3α. Their sperm maturation is impaired, and sperm cannot fertilize eggs in vitro and in vivo. This suggests that GSK3α is a \"male fertility kinase\" in mammals and that GSK3α-selective inhibitor is a potential male contraceptive.</p><p><strong>Materials and methods: </strong>A set of eight heterozygous Gsk3α(±) male mice received daily intraperitoneal injections of BRD0705, an isoform-selective GSK3α inhibitor, at 20 mg/kg body weight for 1 week. Five vehicle-treated and BRD0705-treated mice were tested for in vivo fertility and the remaining mice were sacrificed; their caudal spermatozoa were examined for motility and biochemical properties.</p><p><strong>Results: </strong>The treated mice did not sire any pups while the control group sired 46 pups with a normal gestation period of 19-23 days. Continued fertility testing up to 6 weeks post-treatment, showed that the treated mice regained fertility siring 56 pups, with 76 in the control group. Sperm motility was impaired, its abnormal morphology increased during epididymal transit, Adenosine triphosphate (ATP) levels were low, and tyrosine-phosphorylation of hexokinase was absent: these phenotypes imitated those observed in Gsk3α KO mice. Tyrosine<sup>279</sup>-phosphorylation of GSK3α was reduced in sperm from the treated mice showing that the GSK3α activity was inhibited. The altered sperm phenotypes returned to normal following recovery of fertility.</p><p><strong>Conclusions: </strong>Complete infertility resulted after 1 week of BRD0705-treatment and fertility recovered after cessation of the treatment. Work is ongoing to determine the minimum dose and treatment time and the testing of new compounds with increased selectivity and inhibitory activity against GSK3α.</p>","PeriodicalId":7898,"journal":{"name":"Andrology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142920514","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Although some studies have revealed the close relationship between leptin and premature ejaculation in clinical practice, whether and how leptin participates in the regulation of ejaculatory behaviors are still unknown.
Objective: To explore the role of leptin on ejaculatory behaviors and its underlying mechanism.
Materials and methods: Copulation behavior tests were performed after acute and chronic leptin administration at peripheral and central levels. To compare changes in sympathetic nervous system activity, lumbar sympathetic nervous activity, serum noradrenaline levels, and the distribution of sympathetic fibers in vas deferens and seminal vesicles were analyzed. Construction of virus vector, immunohistochemistry, and optogenetics techniques were used to explore the neural circuit mechanism. The density of dendritic spines in parvocellular region of paraventricular nucleus was measured by Golgi staining.
Results: Acute administration of leptin had no effect on ejaculation behavior in male mice. However, both mount latency and ejaculation latency were significantly shortened, even if serum leptin decreased to normal level, after chronic administration of leptin at peripheral or central level. Additionally, sympathetic fibers in vas deferens and seminal vesicles obviously increased, in which arcuate nucleus‒paraventricular nucleus circuit and glutamatergic neurons in paraventricular nucleus played an important role. Dendritic spine density in parvocellular region increased after chronic leptin administration.
Discussion and conclusion: The role of leptin in regulating ejaculation behavior was chronic, not acute, in which leptin chronically modulated sympathetic neuroplasticity via arcuate nucleus‒paraventricular nucleus circuit and glutamatergic neurons in paraventricular nucleus and promoted ejaculatory behaviors. Increased dendritic spine density in parvocellular region of paraventricular nucleus may be involved as well.
{"title":"Leptin action on ARC-PVN neural circuit regulates ejaculation behavior by altering sympathetic neuroplasticity.","authors":"Qi-Jie Zhang, Jiao-Chen Luan, Qi Gu, Ning-Hong Song, Jia-Dong Xia","doi":"10.1111/andr.13833","DOIUrl":"https://doi.org/10.1111/andr.13833","url":null,"abstract":"<p><strong>Background: </strong>Although some studies have revealed the close relationship between leptin and premature ejaculation in clinical practice, whether and how leptin participates in the regulation of ejaculatory behaviors are still unknown.</p><p><strong>Objective: </strong>To explore the role of leptin on ejaculatory behaviors and its underlying mechanism.</p><p><strong>Materials and methods: </strong>Copulation behavior tests were performed after acute and chronic leptin administration at peripheral and central levels. To compare changes in sympathetic nervous system activity, lumbar sympathetic nervous activity, serum noradrenaline levels, and the distribution of sympathetic fibers in vas deferens and seminal vesicles were analyzed. Construction of virus vector, immunohistochemistry, and optogenetics techniques were used to explore the neural circuit mechanism. The density of dendritic spines in parvocellular region of paraventricular nucleus was measured by Golgi staining.</p><p><strong>Results: </strong>Acute administration of leptin had no effect on ejaculation behavior in male mice. However, both mount latency and ejaculation latency were significantly shortened, even if serum leptin decreased to normal level, after chronic administration of leptin at peripheral or central level. Additionally, sympathetic fibers in vas deferens and seminal vesicles obviously increased, in which arcuate nucleus‒paraventricular nucleus circuit and glutamatergic neurons in paraventricular nucleus played an important role. Dendritic spine density in parvocellular region increased after chronic leptin administration.</p><p><strong>Discussion and conclusion: </strong>The role of leptin in regulating ejaculation behavior was chronic, not acute, in which leptin chronically modulated sympathetic neuroplasticity via arcuate nucleus‒paraventricular nucleus circuit and glutamatergic neurons in paraventricular nucleus and promoted ejaculatory behaviors. Increased dendritic spine density in parvocellular region of paraventricular nucleus may be involved as well.</p>","PeriodicalId":7898,"journal":{"name":"Andrology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142920471","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Emma Cavarocchi, Maëva Drouault, Joao C Ribeiro, Violaine Simon, Marjorie Whitfield, Aminata Touré
Background: In mammals, sperm fertilization potential relies on efficient progression within the female genital tract to reach and fertilize the oocyte. This fundamental property is supported by the flagellum, an evolutionarily conserved organelle, which contains dynein motor proteins that provide the mechanical force for sperm propulsion and motility. Primary motility of the sperm cells is acquired during their transit through the epididymis and hyperactivated motility is acquired throughout the journey in the female genital tract by a process called capacitation. These activation processes rely on the micro-environment of the genital tracts. In particular, during capacitation, a panoply of ion transporters located at the surface of the sperm cells mediate complex ion exchanges, which induce an increase in plasma membrane fluidity, the alkalinization of the cytoplasm and protein phosphorylation cascades that are compulsory for sperm hyperactivation and fertilization potential. As a consequence, both structural and functional defects of the sperm flagellum can affect sperm motility, resulting in asthenozoospermia, which constitutes the most predominant pathological condition associated with human male infertility.
Objectives: Herein, we have performed a literature review to provide a comprehensive description of the recent advances in the genetics of human asthenozoospermia.
Results and discussion: We describe the currently knowledge on gene mutations that affect sperm morphology and motility, namely, asthenoteratozoospermia; we also specify the gene mutations that exclusively affect sperm function and activation, resulting in functional asthenozoospermia. We discuss the benefit of this knowledge for patient and couple management, in terms of genetic counselling and diagnosis of male infertility as a sole phenotype or in association with ciliary defects. Last, we discuss the current strategies that have been initiated for the development of potential therapeutical and contraceptive strategies targeting genes that are essential for sperm function and activation.
{"title":"Human asthenozoospermia: Update on genetic causes, patient management, and clinical strategies.","authors":"Emma Cavarocchi, Maëva Drouault, Joao C Ribeiro, Violaine Simon, Marjorie Whitfield, Aminata Touré","doi":"10.1111/andr.13828","DOIUrl":"https://doi.org/10.1111/andr.13828","url":null,"abstract":"<p><strong>Background: </strong>In mammals, sperm fertilization potential relies on efficient progression within the female genital tract to reach and fertilize the oocyte. This fundamental property is supported by the flagellum, an evolutionarily conserved organelle, which contains dynein motor proteins that provide the mechanical force for sperm propulsion and motility. Primary motility of the sperm cells is acquired during their transit through the epididymis and hyperactivated motility is acquired throughout the journey in the female genital tract by a process called capacitation. These activation processes rely on the micro-environment of the genital tracts. In particular, during capacitation, a panoply of ion transporters located at the surface of the sperm cells mediate complex ion exchanges, which induce an increase in plasma membrane fluidity, the alkalinization of the cytoplasm and protein phosphorylation cascades that are compulsory for sperm hyperactivation and fertilization potential. As a consequence, both structural and functional defects of the sperm flagellum can affect sperm motility, resulting in asthenozoospermia, which constitutes the most predominant pathological condition associated with human male infertility.</p><p><strong>Objectives: </strong>Herein, we have performed a literature review to provide a comprehensive description of the recent advances in the genetics of human asthenozoospermia.</p><p><strong>Results and discussion: </strong>We describe the currently knowledge on gene mutations that affect sperm morphology and motility, namely, asthenoteratozoospermia; we also specify the gene mutations that exclusively affect sperm function and activation, resulting in functional asthenozoospermia. We discuss the benefit of this knowledge for patient and couple management, in terms of genetic counselling and diagnosis of male infertility as a sole phenotype or in association with ciliary defects. Last, we discuss the current strategies that have been initiated for the development of potential therapeutical and contraceptive strategies targeting genes that are essential for sperm function and activation.</p>","PeriodicalId":7898,"journal":{"name":"Andrology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142920525","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Giuseppe Lisco, Anna De Tullio, Maima Matin, Vito Angelo Giagulli, Edoardo Guastamacchia, Giovanni De Pergola, Giuseppina Piazzolla, Vincenzo Triggiani
Background: Evidence indicates a wide range of andrological alterations in patients with the Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV-2) infection and Coronavirus Disease 2019 (COVID-19).
Aim: To provide an update on the andrological effects of SARS-CoV-2 infection and COVID-19.
Methods: PubMed/MEDLINE and Institutional websites were searched for randomized clinical trials, non-systematic reviews, systematic reviews, and meta-analyses.
Results: Fifty-four records were included from 2020 to 2024. The most representative paper categories were non-systematic reviews (n = 26) and systematic reviews/meta-analyses (n = 27). One randomized, prospective, phase 2 trial was also included. Eight topics were identified and discussed as follows: short- and long-term seminal changes attributable to SARS-CoV-2 infection or COVID-19; andrological effects of anti-SARS-CoV-2 vaccines; the potential impact of SARS-CoV-2 infection and COVID-19 on male fertility; the relationship between serum testosterone levels and COVID-19 prognosis in men; fertility care during the pandemic; urinary/genital male system tract impairment in SARS-CoV-2 infection and COVID-19; the effect of SARS-CoV-2 infection and COVID-19 on circulating levels of sex steroid hormones; the impact of SARS-CoV-2 and COVID-19 on sexual function and activity.
Discussion: SARS-CoV-2 can affect the whole testicular function through direct and indirect mechanisms, with a positive relation between the severity of SARS-CoV-2 infection and the level of deterioration of testicular function. Testicular function recovers along with the recovery from the disease. In vitro fertilization techniques ensure similar results in patients with or without previous SARS-CoV-2 infection or COVID-19. Immunization with anti-SARS-CoV-2 vaccines prevents andrological complaints due to naturally occurring infection. Erectile dysfunction and sexual dysfunction are frequently diagnosed in COVID-19 patients due to several contributing factors, including hormonal imbalance and psychosocial complaints related to the pandemic.
{"title":"Update on andrological effects of SARS-CoV-2 infection and COVID-19: An overview review.","authors":"Giuseppe Lisco, Anna De Tullio, Maima Matin, Vito Angelo Giagulli, Edoardo Guastamacchia, Giovanni De Pergola, Giuseppina Piazzolla, Vincenzo Triggiani","doi":"10.1111/andr.13830","DOIUrl":"https://doi.org/10.1111/andr.13830","url":null,"abstract":"<p><strong>Background: </strong>Evidence indicates a wide range of andrological alterations in patients with the Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV-2) infection and Coronavirus Disease 2019 (COVID-19).</p><p><strong>Aim: </strong>To provide an update on the andrological effects of SARS-CoV-2 infection and COVID-19.</p><p><strong>Methods: </strong>PubMed/MEDLINE and Institutional websites were searched for randomized clinical trials, non-systematic reviews, systematic reviews, and meta-analyses.</p><p><strong>Results: </strong>Fifty-four records were included from 2020 to 2024. The most representative paper categories were non-systematic reviews (n = 26) and systematic reviews/meta-analyses (n = 27). One randomized, prospective, phase 2 trial was also included. Eight topics were identified and discussed as follows: short- and long-term seminal changes attributable to SARS-CoV-2 infection or COVID-19; andrological effects of anti-SARS-CoV-2 vaccines; the potential impact of SARS-CoV-2 infection and COVID-19 on male fertility; the relationship between serum testosterone levels and COVID-19 prognosis in men; fertility care during the pandemic; urinary/genital male system tract impairment in SARS-CoV-2 infection and COVID-19; the effect of SARS-CoV-2 infection and COVID-19 on circulating levels of sex steroid hormones; the impact of SARS-CoV-2 and COVID-19 on sexual function and activity.</p><p><strong>Discussion: </strong>SARS-CoV-2 can affect the whole testicular function through direct and indirect mechanisms, with a positive relation between the severity of SARS-CoV-2 infection and the level of deterioration of testicular function. Testicular function recovers along with the recovery from the disease. In vitro fertilization techniques ensure similar results in patients with or without previous SARS-CoV-2 infection or COVID-19. Immunization with anti-SARS-CoV-2 vaccines prevents andrological complaints due to naturally occurring infection. Erectile dysfunction and sexual dysfunction are frequently diagnosed in COVID-19 patients due to several contributing factors, including hormonal imbalance and psychosocial complaints related to the pandemic.</p>","PeriodicalId":7898,"journal":{"name":"Andrology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142906324","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jie Chen, Jie-Jie He, Shan Wu, Zhao-Yang Deng, Yu-Peng Liu, Jian-Fan Chen, Yue Xu, Han-Kai Fang, Ya-Lan Wei, Zhen-Yu She
Background: The establishment of kinetochore-microtubule attachment is essential for error-free chromosome alignment and segregation during cell division. Defects in chromosome alignment result in chromosome instability, birth defects, and infertility. Kinesin-7 CENP-E mediates kinetochore-microtubule capture, chromosome alignment, and spindle assembly checkpoint in somatic cells, however, mechanisms of CENP-E in germ cells remain poorly understood.
Objectives: This study aimed to explore the functions of CENP-E in the proliferation and self-renewal of spermatogonia.
Materials and methods: A CENP-E heterozygous knockout strain was established in C57BL/6J mice using the CRISPR/Cas9 and Cre/LoxP system. Hematoxylin-eosin staining was performed to study the histology. The inhibition of CENP-E in the GC-1 spg cells was performed using the specific inhibitor GSK923295. The expression and localization of spermatogonial marker proteins were determined by immunofluorescence using confocal microscopy in the control and CENP-E+/- heterozygous mouse testes. The protein expression level was analyzed using Western blot. The cell-cycle and apoptosis assay were measured using flow cytometry. In addition, karyotype analysis was performed using hypotonic preparation and chromosome spreading.
Results: Here, we reveal that CENP-E haploinsufficiency results in chromosome misalignment, spindle disorganization, and metaphase arrest in spermatogonia, which leads to the loss of spermatogonia, chromosomal instability, and spermatogenic disorders. Notably, CENP-E ablation leads to the activation of spindle assembly checkpoint and aneuploidy, which impairs the proliferation and self-renewal of spermatogonia.
Discussion and conclusion: CENP-E depletion disrupts the recruitment of key checkpoint proteins, including BubR1, Bub1, KIF2C, and Aurora B, indicating a causal relationship between chromosome misalignment and spindle assembly checkpoint activation in spermatogonia. Our findings demonstrate that CENP-E regulates kinetochore-microtubule attachment, chromosome alignment, and spindle assembly checkpoint in spermatogonia.
{"title":"CENP-E haploinsufficiency causes chromosome misalignment and spindle assembly checkpoint activation in the spermatogonia.","authors":"Jie Chen, Jie-Jie He, Shan Wu, Zhao-Yang Deng, Yu-Peng Liu, Jian-Fan Chen, Yue Xu, Han-Kai Fang, Ya-Lan Wei, Zhen-Yu She","doi":"10.1111/andr.13834","DOIUrl":"https://doi.org/10.1111/andr.13834","url":null,"abstract":"<p><strong>Background: </strong>The establishment of kinetochore-microtubule attachment is essential for error-free chromosome alignment and segregation during cell division. Defects in chromosome alignment result in chromosome instability, birth defects, and infertility. Kinesin-7 CENP-E mediates kinetochore-microtubule capture, chromosome alignment, and spindle assembly checkpoint in somatic cells, however, mechanisms of CENP-E in germ cells remain poorly understood.</p><p><strong>Objectives: </strong>This study aimed to explore the functions of CENP-E in the proliferation and self-renewal of spermatogonia.</p><p><strong>Materials and methods: </strong>A CENP-E heterozygous knockout strain was established in C57BL/6J mice using the CRISPR/Cas9 and Cre/LoxP system. Hematoxylin-eosin staining was performed to study the histology. The inhibition of CENP-E in the GC-1 spg cells was performed using the specific inhibitor GSK923295. The expression and localization of spermatogonial marker proteins were determined by immunofluorescence using confocal microscopy in the control and CENP-E<sup>+/-</sup> heterozygous mouse testes. The protein expression level was analyzed using Western blot. The cell-cycle and apoptosis assay were measured using flow cytometry. In addition, karyotype analysis was performed using hypotonic preparation and chromosome spreading.</p><p><strong>Results: </strong>Here, we reveal that CENP-E haploinsufficiency results in chromosome misalignment, spindle disorganization, and metaphase arrest in spermatogonia, which leads to the loss of spermatogonia, chromosomal instability, and spermatogenic disorders. Notably, CENP-E ablation leads to the activation of spindle assembly checkpoint and aneuploidy, which impairs the proliferation and self-renewal of spermatogonia.</p><p><strong>Discussion and conclusion: </strong>CENP-E depletion disrupts the recruitment of key checkpoint proteins, including BubR1, Bub1, KIF2C, and Aurora B, indicating a causal relationship between chromosome misalignment and spindle assembly checkpoint activation in spermatogonia. Our findings demonstrate that CENP-E regulates kinetochore-microtubule attachment, chromosome alignment, and spindle assembly checkpoint in spermatogonia.</p>","PeriodicalId":7898,"journal":{"name":"Andrology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2024-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142906306","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}