Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Biol Reprod ; 108(3): 447-464, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36617158

ABSTRACT

The formation of fertilisation-competent sperm requires spermatid morphogenesis (spermiogenesis), a poorly understood program that involves complex coordinated restructuring and specialised cytoskeletal structures. A major class of cytoskeletal regulators are the actin-related proteins (ARPs), which include conventional actin variants, and related proteins that play essential roles in complexes regulating actin dynamics, intracellular transport, and chromatin remodeling. Multiple testis-specific ARPs are well conserved among mammals, but their functional roles are unknown. One of these is actin-like 7b (Actl7b) that encodes an orphan ARP highly similar to the ubiquitously expressed beta actin (ACTB). Here we report ACTL7B is expressed in human and mouse spermatids through the elongation phase of spermatid development. In mice, ACTL7B specifically localises to the developing acrosome, within the nucleus of early spermatids, and to the flagellum connecting region. Based on this localisation pattern and high level of sequence conservation in mice, humans, and other mammals, we examined the requirement for ACTL7B in spermiogenesis by generating and characterising the reproductive phenotype of male Actl7b KO mice. KO mice were infertile, with severe and variable oligoteratozoospermia (OAT) and multiple morphological abnormalities of the flagellum (MMAF) and sperm head. These defects phenocopy human OAT and MMAF, which are leading causes of idiopathic male infertility. In conclusion, this work identifies ACTL7B as a key regulator of spermiogenesis that is required for male fertility.


Subject(s)
Actins , Infertility, Male , Male , Mice , Humans , Animals , Actins/genetics , Actins/metabolism , Avena , Seeds/metabolism , Spermatids/metabolism , Spermatogenesis/genetics , Flagella/metabolism , Infertility, Male/genetics , Infertility, Male/metabolism , Mammals
2.
Endocrinology ; 159(9): 3403-3420, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30099545

ABSTRACT

Retinoic acid receptor α (RARA), a retinoic acid-dependent transcription factor, is expressed in both somatic and germ cells of the testis. Rara-null male mice with global Rara mutations displayed severely degenerated testis and infertility phenotypes. To elucidate the specific responsibility of germ cell RARA in spermatogenesis, Rara was deleted in germ cells, generating germ cell-specific Rara conditional knockout (cKO) mice. These Rara cKO animals exhibited phenotypes of quantitatively reduced epididymal sperm counts and disorganized germ cell layers in the seminiferous tubules, which worsened with aging. Abnormal tubules lacked lumen, contained vacuoles, and showed massive germ cell sloughing, all characteristics similar to those observed in Rara-null tubules. Spermatocyte chromosomal spreads revealed a novel role for germ cell RARA in modulating the integrity of synaptonemal complexes and meiotic progression. Furthermore, the initiation of spermatogenesis from spermatogonial stem cells was decreased in Rara cKO testes following busulfan treatment, supporting a role of germ cell RARA in spermatogonial proliferation. Collectively, the evidence in this study indicates that RARA produced in male germ cells has a broad spectrum of functions throughout spermatogenesis, which includes the maintenance of seminiferous epithelium organization, the integrity of the meiotic genome, and spermatogonial proliferation and differentiation. The results further suggest that germ cell RARA has dual functions: intrinsically in germ cells, balancing proliferation and differentiation of spermatogonia, and controlling genome integrity during meiosis; and extrinsically in the crosstalks with Sertoli cells, controlling the cell junctional physiology for coordinating proper spatial and temporal development of germ cells during spermatogenesis.


Subject(s)
Meiosis/genetics , Retinoic Acid Receptor alpha/genetics , Spermatogenesis/genetics , Spermatozoa/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Epididymis , Germ Cells , Intercellular Junctions/metabolism , Male , Mice , Mice, Knockout , Retinoic Acid Receptor alpha/metabolism , Seminiferous Epithelium/metabolism , Seminiferous Tubules/metabolism , Sertoli Cells/metabolism , Signal Transduction , Sperm Count , Spermatocytes/growth & development , Spermatocytes/metabolism , Spermatogonia/growth & development , Spermatogonia/metabolism , Testis
3.
Reproduction ; 152(5): 379-88, 2016 11.
Article in English | MEDLINE | ID: mdl-27486269

ABSTRACT

We previously described a novel germ cell-specific X-linked reproductive homeobox gene (Rhox13) that is upregulated at the level of translation in response to retinoic acid (RA) in differentiating spermatogonia and preleptotene spermatocytes. We hypothesize that RHOX13 plays an essential role in male germ cell differentiation, and have tested this by creating a Rhox13 gene knockout (KO) mouse. Rhox13 KO mice are born in expected Mendelian ratios, and adults have slightly reduced testis weights, yet a full complement of spermatogenic cell types. Young KO mice (at ~7-8 weeks of age) have a ≈50% reduction in epididymal sperm counts, but numbers increased to WT levels as the mice reach ~17 weeks of age. Histological analysis of testes from juvenile KO mice reveals a number of defects during the first wave of spermatogenesis. These include increased apoptosis, delayed appearance of round spermatids and disruption of the precise stage-specific association of germ cells within the seminiferous tubules. Breeding studies reveal that both young and aged KO males produce normal-sized litters. Taken together, our results indicate that RHOX13 is not essential for mouse fertility in a controlled laboratory setting, but that it is required for optimal development of differentiating germ cells and progression of the first wave of spermatogenesis.


Subject(s)
Apoptosis , Epididymis/cytology , Fertilization/physiology , Homeodomain Proteins/physiology , Spermatocytes/cytology , Spermatogenesis/physiology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Cell Calcium ; 59(4): 189-97, 2016 04.
Article in English | MEDLINE | ID: mdl-26969191

ABSTRACT

Store-operated calcium entry (SOCE) is an important Ca(2+) influx pathway in somatic cells. In addition to maintaining endoplasmic reticulum (ER) Ca(2+) stores, Ca(2+) entry through store-operated channels regulates essential signaling pathways in numerous cell types. Patients with mutations in the store-operated channel subunit ORAI1 exhibit defects in store-operated Ca(2+) influx, along with severe immunodeficiency, congenital myopathy and ectodermal dysplasia. However, little is known about the functional role of ORAI1 in germ cells and reproductive function in mice, or in men, since men with loss-of-function or null mutations in ORAI1 rarely survive to reproductive age. In this study, we investigated the role of ORAI1 in male reproductive function. We reveal that Orai1(-/-) male mice are sterile and have severe defects in spermatogenesis, with prominent deficiencies in mid- to late-stage elongating spermatid development. These studies establish an essential in vivo role for store-operated ORAI1 channels in male reproductive function and identify these channels as potential non-steroidal regulators of male fertility.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Infertility, Male/metabolism , ORAI1 Protein/metabolism , Animals , Cell Separation/methods , Female , Male , Mice , ORAI1 Protein/deficiency
5.
Biol Reprod ; 93(6): 137, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26490841

ABSTRACT

While cyclin dependent kinase 1 (CDK1) has a critical role in controlling resumption of meiosis in oocytes, its role has not been investigated directly in spermatocytes. Unique aspects of male meiosis led us to hypothesize that its role is different in male meiosis than in female meiosis. We generated a conditional knockout (cKO) of the Cdk1 gene in mouse spermatocytes to test this hypothesis. We found that CDK1-null spermatocytes undergo synapsis, chiasmata formation, and desynapsis as is seen in oocytes. Additionally, CDK1-null spermatocytes relocalize SYCP3 to centromeric foci, express H3pSer10, and initiate chromosome condensation. However, CDK1-null spermatocytes fail to form condensed bivalent chromosomes in prophase of meiosis I and instead are arrested at prometaphase. Thus, CDK1 has an essential role in male meiosis that is consistent with what is known about the role of CDK1 in female meiosis, where it is required for formation of condensed bivalent metaphase chromosomes and progression to the first meiotic division. We found that cKO spermatocytes formed fully condensed bivalent chromosomes in the presence of okadaic acid, suggesting that cKO chromosomes are competent to condense, although they do not do so in vivo. Additionally, arrested cKO spermatocytes exhibited irregular cell shape, irregular large nuclei, and large distinctive nucleoli. These cells persist in the seminiferous epithelium through the next seminiferous epithelial cycle with a lack of stage XII checkpoint-associated cell death. This indicates that CDK1 is required upstream of a checkpoint-associated cell death as well as meiotic metaphase progression in mouse spermatocytes.


Subject(s)
CDC2 Protein Kinase/genetics , Infertility, Male/genetics , Meiosis/genetics , Spermatocytes/metabolism , Spermatogenesis/genetics , Animals , CDC2 Protein Kinase/metabolism , Cell Nucleus/metabolism , Cell Shape/genetics , Infertility, Male/metabolism , Male , Mice , Mice, Knockout , Spermatocytes/cytology
6.
Biol Reprod ; 88(4): 90, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23446454

ABSTRACT

Sperm utilize glycolysis to generate ATP required for motility, and several spermatogenic cell-specific glycolytic isozymes are associated with the fibrous sheath (FS) in the principal piece of the sperm flagellum. We used proteomics and molecular biology approaches to confirm earlier reports that a novel enolase is present in mouse sperm. We then found that a pan-enolase antibody, but not antibodies to ENO2 and ENO3, recognized a protein in the principal piece of the mouse sperm flagellum. Database analyses identified two previously uncharacterized enolase family-like candidate genes, 64306537H0Rik and Gm5506. Northern analysis indicated that 64306537H0Rik (renamed Eno4) was transcribed in testes of mice by Postnatal Day 12. To determine the role of ENO4, we generated mice using embryonic stem cells in which an Eno4 allele was disrupted by a gene trap containing a beta galactosidase (beta-gal) reporter (Eno4(+/Gt)). Expression of beta-gal occurred in the testis, and male mice homozygous for the gene trap allele (Eno4(Gt/Gt)) were infertile. Epididymal sperm numbers were 2-fold lower and sperm motility was reduced substantially in Eno4(Gt/Gt) mice compared to wild-type mice. Sperm from Eno4(Gt/Gt) mice had a coiled flagellum and a disorganized FS. The Gm5506 gene encodes a protein identical to ENO1 and also is transcribed at a low level in testis. We conclude that ENO4 is required for normal assembly of the FS and provides most of the enolase activity in sperm and that Eno1 and/or Gm5506 may encode a minor portion of the enolase activity in sperm.


Subject(s)
Infertility, Male/genetics , Phosphopyruvate Hydratase/genetics , Spermatogenesis/genetics , Spermatozoa/abnormalities , Animals , Female , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutagenesis/physiology , Organ Specificity/genetics , Phosphopyruvate Hydratase/physiology , Spermatozoa/enzymology , Spermatozoa/ultrastructure
7.
Proc Natl Acad Sci U S A ; 109(51): 21140-5, 2012 Dec 18.
Article in English | MEDLINE | ID: mdl-23213263

ABSTRACT

Estrogen receptor alpha (ERα) is a ligand-dependent transcription factor containing two transcriptional activation function (AF) domains. AF-1 is in the N terminus of the receptor protein, and AF-2 activity is dependent on helix 12 of the C-terminal ligand-binding domain. We recently showed that two point mutations converting leucines 543 and 544 to alanines in helix 12 (AF2ER) minimized estrogen-dependent AF-2 transcriptional activation. A characteristic feature of AF2ER is that the estrogen antagonists ICI182780 and tamoxifen (TAM) act as agonists through intact AF-1, but not through mutated AF-2. Here we report the reproductive phenotype of male AF2ER knock-in (AF2ERKI) mice and demonstrate the involvement of ERα in male fertility. The AF2ERKI male homozygotes are infertile because of seminiferous tubular dysmorphogenesis in the testis, similar to ERα KO males. Sperm counts and motility did not differ at age 6 wk in AF2ERKI and WT mice, but a significant testis defect was observed in adult AF2ERKI male mice. The expression of efferent ductal genes involved in fluid reabsorption was significantly lower in AF2ERKI males. TAM treatment for 3 wk beginning at age 21 d activated AF-2-mutated ERα (AF2ER) and restored expression of efferent ductule genes. At the same time, the TAM treatment reversed AF2ERKI male infertility compared with the vehicle-treated group. These results indicate that the ERα AF-2 mutation results in male infertility, suggesting that the AF-1 is regulated in an AF-2-dependent manner in the male reproductive tract. Activation of ERα AF-1 is capable of rescuing AF2ERKI male infertility.


Subject(s)
Estrogen Receptor alpha/metabolism , Animals , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/chemistry , Fertility , Fulvestrant , Homozygote , Ligands , Male , Mice , Mice, Knockout , Point Mutation , Protein Structure, Tertiary , Reproduction , Sperm Count , Sperm Motility , Tamoxifen/pharmacology , Testis/metabolism , Transcriptional Activation
8.
Reprod Toxicol ; 34(4): 512-21, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22989549

ABSTRACT

ERΔ3 transgenic mice expressing a dominant negative estrogen receptor α (ERα) variant lacking the second zinc finger in the DNA binding domain were developed to examine its potential to inhibit estrogen action in vivo. To investigate if ERΔ3 expression influences uterine carcinogenesis, ERΔ3 transgenic mice were exposed to diethylstilbestrol (DES) on post-natal days 1-5. Neonatal DES treatment induced uterine adenocarcinomas in 81% of 8-month-old ERΔ3 mice compared to 49% of wild-type females (p<0.016). ERΔ3 did not inhibit the expression of the estrogen-responsive progesterone receptor and lactoferrin genes in the presence of ERα or modify their expression in ERα knockout (αERKO) mice. Higher circulating 17ß-estradiol levels and non-classical signaling by ERΔ3 may be related to the earlier incidence of uterine cancer. These findings indicate that expression of this ERα variant can influence determining events in uterine cancer development and its natural occurrence in the human uterus would unlikely be protective.


Subject(s)
Carcinogens/toxicity , Diethylstilbestrol/toxicity , Estrogen Receptor alpha/genetics , Estrogens/toxicity , Uterine Neoplasms/genetics , Animals , Animals, Newborn , Estradiol/blood , Estrogen Receptor alpha/metabolism , Female , Male , Mice , Mice, Transgenic , Progesterone/blood , Uterine Neoplasms/chemically induced , Uterine Neoplasms/metabolism
9.
J Endocrinol ; 207(3): 281-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20833731

ABSTRACT

Disruption of the Esr1 gene encoding estrogen receptor α (ERα) by insertion of a neomycin resistance gene (neo) into exon 2 (αERKO mice) was shown previously to cause infertility in male mice. While full-length ERα protein was not expressed in αERKO mice, alternative splicing resulted in the low-level expression of a truncated form lacking the N-terminus A/B domain and containing the DNA- and ligand-binding domains. Thus, it was unclear whether the reproductive phenotype in αERKO males was only due to the lack of full-length ERα or was affected by the presence of the variant ERα isoform. The present study examined male mice with deletion of exon 3 of Esr1 gene, lacking the DNA-binding domain, and null for ERα (Ex3αERKO). Dilation of some seminiferous tubules was apparent in male Ex3αERKO mice as early as postnatal day 10 and was pronounced in all tubules from day 20 onward. At 6 weeks of age, sperm numbers and sperm motility were lower in Ex3αERKO mice than in wild-type (WT) mice, and the rete testis and efferent ductules were dilated. Mating studies determined that adult Ex3αERKO males were infertile and failed to produce copulatory plugs. Serum testosterone levels and Hsd17b3 and Cyp17a1 transcript levels were significantly higher, but serum estradiol, progesterone, LH, and FSH levels and Cyp19a1 transcript levels were not significantly different from those in WT mice. These results confirm and extend those seen in other studies on male mice with deletion of exon 3 of Esr1 gene. In addition, the reproductive phenotype of male Ex3αERKO mice recapitulated the phenotype of αERKO mice, strongly suggesting that the αERKO male infertility was not due to the presence of the DNA-binding domain in the truncated form of ERα and that full-length ERα is essential for maintenance of male fertility.


Subject(s)
Estrogen Receptor alpha/genetics , Infertility, Male/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , Animals , Epididymis , Estradiol/blood , Estradiol/genetics , Exons/genetics , Follicle Stimulating Hormone/blood , Luteinizing Hormone/blood , Male , Mice , Mice, Inbred C57BL , Phenotype , Progesterone/blood , Rete Testis , Seminiferous Tubules , Sequence Deletion , Sperm Count , Sperm Motility/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Testosterone/blood , Testosterone/genetics
10.
J Am Assoc Lab Anim Sci ; 49(4): 423-6, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20819387

ABSTRACT

The objective of this study was to evaluate the effect on litter size of 2 analgesics used perioperatively during mouse embryo transfer surgery. Day 2.5 pseudopregnant CD1 mice (n = 96) were divided equally into 2 analgesic treatment groups and a saline control group. Each mouse received a single, subcutaneous dose of buprenorphine hydrochloride (0.1 mg/kg), flunixin meglumine (2.5 mg/kg), or saline immediately after induction of anesthesia with 2.5% isoflurane. Each mouse then was prepared for aseptic surgery. Blastocysts had previously been collected from C57BL/6NCrl female mice that were synchronized and superovulated by using pregnant mare serum gonadotropin and human chorionic gonadotropin and mated with C57BL/6NTac male mice 3.5 d before collection. Viable blastocysts were pooled, and 8 were selected arbitrarily and transplanted into the right uterine horn of each pseudopregnant CD1 mouse. Mice were monitored throughout pregnancy, and the number of pups at birth was documented. No statistically significant difference was found between the 3 groups. These results indicate that perioperative analgesic treatment with buprenorphine or flunixin in the CD1 mouse undergoing embryo transfer is not associated with increased embryonic loss.


Subject(s)
Analgesia/adverse effects , Buprenorphine/adverse effects , Clonixin/analogs & derivatives , Embryo Transfer/veterinary , Litter Size/drug effects , Animals , Clonixin/adverse effects , Female , Maternal Exposure , Mice , Mice, Inbred ICR , Perioperative Care/veterinary , Pregnancy , Pregnancy Rate
11.
Biol Reprod ; 82(1): 136-45, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19759366

ABSTRACT

Phosphoglycerate kinase 2 (PGK2), an isozyme that catalyzes the first ATP-generating step in the glycolytic pathway, is encoded by an autosomal retrogene that is expressed only during spermatogenesis. It replaces the ubiquitously expressed phosphoglycerate kinase 1 (PGK1) isozyme following repression of Pgk1 transcription by meiotic sex chromosome inactivation during meiotic prophase and by postmeiotic sex chromatin during spermiogenesis. The targeted disruption of Pgk2 by homologous recombination eliminates PGK activity in sperm and severely impairs male fertility, but does not block spermatogenesis. Mating behavior, reproductive organ weights (testis, excurrent ducts, and seminal vesicles), testis histology, sperm counts, and sperm ultrastructure were indistinguishable between Pgk2(-/-) and wild-type mice. However, sperm motility and ATP levels were markedly reduced in males lacking PGK2. These defects in sperm function were slightly less severe than observed in males lacking glyceraldehyde-3-phosphate dehydrogenase, spermatogenic (GAPDHS), the isozyme that catalyzes the step preceding PGK2 in the sperm glycolytic pathway. Unlike Gapdhs(-/-) males, the Pgk2(-/-) males also sired occasional pups. Alternative pathways that bypass the PGK step of glycolysis exist. We determined that one of these bypass enzymes, acylphosphatase, is active in mouse sperm, perhaps contributing to phenotypic differences between mice lacking GAPDHS or PGK2. This study determined that PGK2 is not required for the completion of spermatogenesis, but is essential for sperm motility and male fertility. In addition to confirming the importance of the glycolytic pathway for sperm function, distinctive phenotypic characteristics of Pgk2(-/-) mice may provide further insights into the regulation of sperm metabolism.


Subject(s)
Fertility , Isoenzymes/metabolism , Phosphoglycerate Kinase/metabolism , Spermatogenesis , Spermatozoa/enzymology , Testis/enzymology , Acid Anhydride Hydrolases/metabolism , Adenosine Triphosphate/metabolism , Animals , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sperm Count , Sperm Motility , Spermatozoa/ultrastructure , Acylphosphatase
12.
Genesis ; 48(2): 114-20, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20027617

ABSTRACT

We generated transgenic mouse line C57BL/6-Tg(Hspa2-cre)1Eddy/J (Hspa2-cre), which expresses cre-recombinase under the control of a 907-bp fragment of the heat shock protein 2 (Hspa2) gene promoter. Transgene expression was determined using Gt(ROSA)26Sor(tm1Sor)/J (ROSA26) and Tg(CAG-Bgeo/GFP)21Lbe/J (Z/EG) reporter strains and RT-PCR and immunohistochemistry assays. Hspa2-cre expression mimicked the spermatogenic cell-specific expression of endogenous HSPA2 within the testis, being first observed in leptotene/zygotene spermatocytes. Expression of the transgene also was detected at restricted sites in the brain, as occurs for endogenous HSPA2. Although the results of mating the Hspa2-cre mice to mice with a floxed Cdc2a allele indicated that some expression of the transgene occurs during embryogenesis, the Hspa2-cre mice provide a valuable new tool for assessing the roles of genes during and after meiotic prophase in pachytene spermatocytes.


Subject(s)
HSP27 Heat-Shock Proteins/genetics , Integrases/metabolism , Promoter Regions, Genetic , Spermatocytes/metabolism , Animals , Genes, Reporter , HSP27 Heat-Shock Proteins/metabolism , Immunohistochemistry , Male , Mice , Mice, Transgenic , Mutation , RNA, Messenger/metabolism , Recombination, Genetic , Testis/cytology , Transgenes
13.
Biol Reprod ; 80(3): 425-31, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19005167

ABSTRACT

Female mice treated neonatally with the phytoestrogen genistein (50 mg/kg/day) have multioocyte follicles, lack regular estrous cyclicity, and are infertile even after superovulation. To determine the cause of their infertility, we examined oocyte developmental competence and timing of embryo loss. Eggs obtained by superovulation of genistein-treated or control females were equally capable of being fertilized in vitro and cultured to the blastocyst stage. However, if eggs were fertilized in vivo, retrieved at the pronucleus stage, and cultured, there was a significant reduction in the percentage of embryos from genistein-treated females reaching the blastocyst stage. When these blastocysts were transferred to pseudopregnant recipients, the number of live pups produced was similar to that in controls. Preimplantation embryo development in vivo was examined by flushing embryos from the oviduct and/or uterus. Similar numbers of one-cell and two-cell embryos were obtained from genistein-treated and control females. However, significantly fewer embryos (<50%) were obtained from genistein-treated females on postcoital Days 3 and 4. To determine if neonatal genistein treatment altered the ability of the uterus to support implantation, blastocysts from control donors were transferred to control and genistein-treated pseudopregnant recipients. These experiments demonstrated that genistein-treated females are not capable of supporting normal implantation of control embryos. Taken together, these results suggest that oocytes from mice treated neonatally with genistein are developmentally competent; however, the oviductal environment and the uterus have abnormalities that contribute to the observed reproductive failure.


Subject(s)
Embryo Implantation/drug effects , Embryonic Development/drug effects , Genistein/toxicity , Infertility, Female/chemically induced , Phytoestrogens/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Animals , Cells, Cultured , Disease Models, Animal , Embryo Implantation/physiology , Embryo Transfer , Embryonic Development/physiology , Female , Fertilization/drug effects , Fertilization/physiology , Genistein/pharmacology , Infertility, Female/physiopathology , Mice , Mice, Inbred ICR , Oocytes/drug effects , Oocytes/physiology , Ovary/drug effects , Ovary/physiopathology , Oviducts/drug effects , Oviducts/physiopathology , Phytoestrogens/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Uterus/drug effects , Uterus/physiopathology
14.
Dev Biol ; 320(1): 12-8, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18571638

ABSTRACT

Mammalian fertilization is a complex multi-step process mediated by different molecules present on both gametes. Epididymal protein CRISP1, a member of the Cysteine-RIch Secretory Protein (CRISP) family, was identified by our laboratory and postulated to participate in both sperm-zona pellucida (ZP) interaction and gamete fusion by binding to egg-complementary sites. To elucidate the functional role of CRISP1 in vivo, we disrupted the Crisp1 gene and evaluated the effect on animal fertility and several sperm parameters. Male and female Crisp1(-/-) animals exhibited no differences in fertility compared to controls. Sperm motility and the ability to undergo a spontaneous or progesterone-induced acrosome reaction were neither affected in Crisp1(-/-) mice. However, the level of protein tyrosine phosphorylation during capacitation was clearly lower in mutant sperm than in controls. In vitro fertilization assays showed that Crisp1(-/-) sperm also exhibited a significantly reduced ability to penetrate both ZP-intact and ZP-free eggs. Moreover, when ZP-free eggs were simultaneously inseminated with Crisp1(+/+) and Crisp1(-/-) sperm in a competition assay, the mutant sperm exhibited a greater disadvantage in their fusion ability. Finally, the finding that the fusion ability of Crisp1(-/-) sperm was further inhibited by the presence of CRISP1 or CRISP2 during gamete co-incubation, supports that another CRISP cooperates with CRISP1 during fertilization and might compensate for its lack in the mutant mice. Together, these results indicate that CRISP proteins are players in the mammalian fertilization process. To our knowledge this is the first knockout mice generated for a CRISP protein. The information obtained might have important functional implications for other members of the widely distributed and evolutionarily conserved CRISP family.


Subject(s)
Fertilization/physiology , Membrane Glycoproteins/deficiency , Spermatozoa/physiology , Acrosome Reaction , Animals , Fertility , Gene Targeting , Male , Membrane Glycoproteins/metabolism , Mice , Sperm Capacitation
15.
Biol Reprod ; 79(1): 26-34, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18367675

ABSTRACT

The lactate dehydrogenase (LDH) protein family members characteristically are distributed in tissue- and cell type-specific patterns and serve as the terminal enzyme of glycolysis, catalyzing reversible oxidation reduction between pyruvate and lactate. They are present as tetramers, and one family member, LDHC, is abundant in spermatocytes, spermatids, and sperm, but also is found in modest amounts in oocytes. We disrupted the Ldhc gene to determine whether LDHC is required for spermatogenesis, oogenesis, and/or sperm and egg function. The targeted disruption of Ldhc severely impaired fertility in male Ldhc(-/-) mice but not in female Ldhc(-/-) mice. Testis and sperm morphology and sperm production appeared to be normal. However, total LDH enzymatic activity was considerably lower in Ldhc(-/-) sperm than in wild type sperm, indicating that the LDHC homotetramer (LDH-C(4)) is responsible for most of the LDH activity in sperm. Although initially motile when isolated, there was a more rapid reduction in the level of ATP and in motility in Ldhc(-)(/-) sperm than in wild-type sperm. Moreover, Ldhc(-/-) sperm did not acquire hyperactivated motility, were unable to penetrate the zona pellucida in vitro, and failed to undergo the phosphorylation events characteristic of capacitation. These studies showed that LDHC plays an essential role in maintenance of the processes of glycolysis and ATP production in the flagellum that are required for male fertility and sperm function.


Subject(s)
Fertility/genetics , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/physiology , Adenosine Triphosphate/metabolism , Animals , Female , Gene Expression/physiology , Glycolysis/genetics , Infertility, Male/genetics , Infertility, Male/pathology , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/physiology , L-Lactate Dehydrogenase/metabolism , Lactic Acid/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size/genetics , RNA, Messenger/metabolism , Sperm Motility/genetics , Spermatozoa/pathology , Spermatozoa/physiology , Testis/metabolism , Testis/pathology
16.
Proc Natl Acad Sci U S A ; 101(47): 16501-6, 2004 Nov 23.
Article in English | MEDLINE | ID: mdl-15546993

ABSTRACT

Although glycolysis is highly conserved, it is remarkable that several unique isozymes in this central metabolic pathway are found in mammalian sperm. Glyceraldehyde 3-phosphate dehydrogenase-S (GAPDS) is the product of a mouse gene expressed only during spermatogenesis and, like its human ortholog (GAPD2), is the sole GAPDH isozyme in sperm. It is tightly bound to the fibrous sheath, a cytoskeletal structure that extends most of the length of the sperm flagellum. We disrupted Gapds expression by gene targeting to selectively block sperm glycolysis and assess its relative importance for in vivo sperm function. Gapds(-/-) males were infertile and had profound defects in sperm motility, exhibiting sluggish movement without forward progression. Although mitochondrial oxygen consumption was unchanged, sperm from Gapds(-/-) mice had ATP levels that were only 10.4% of those in sperm from WT mice. These results imply that most of the energy required for sperm motility is generated by glycolysis rather than oxidative phosphorylation. Furthermore, the critical role of glycolysis in sperm and its dependence on this sperm-specific enzyme suggest that GAPDS is a potential contraceptive target, and that mutations or environmental agents that disrupt its activity could lead to male infertility.


Subject(s)
Fertility/physiology , Glyceraldehyde-3-Phosphate Dehydrogenases/physiology , Sperm Motility/physiology , Adenosine Triphosphate/metabolism , Animals , Base Sequence , DNA/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/deficiency , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Glycolysis , Humans , Infertility, Male/enzymology , Infertility, Male/genetics , Isoenzymes/deficiency , Isoenzymes/genetics , Isoenzymes/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Mitochondria/metabolism , Oxygen Consumption , Spermatozoa/metabolism , Spermatozoa/ultrastructure
17.
Biol Reprod ; 69(1): 211-7, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12620939

ABSTRACT

Cytokinesis is incomplete in spermatogenic cells, and the descendants of each stem cell form a clonal syncytium. As a result, a heterozygous mutation in a gene expressed postmeiotically affects all of the haploid spermatids within a syncytium. Previously, we have found that disruption of one copy of the gene for either protamine 1 (PRM1) or protamine 2 (PRM2) in the mouse results in a reduction in the amount of the respective protein, abnormal processing of PRM2, and inability of male chimeras to transmit either the mutant or wild-type allele derived from the 129-genotype embryonic stem cells to the next generation. Although it is believed that protamines are essential for compaction of the sperm nucleus and to protect the DNA from damage, this has not been proven experimentally. To test the hypothesis that failure of chimeras to transmit the 129 genotype to offspring was due to alterations in the organization and integrity of sperm DNA, we used the single-cell DNA electrophoresis (comet) assay, ultrastructural analysis, and the intracytoplasmic sperm injection (ICSI) procedure. Comet assay demonstrated a direct correlation between the fraction of sperm with haploinsufficiency of PRM2 and the frequency of sperm with damaged DNA. Ultrastructural analysis revealed reduced compaction of the chromatin. ICSI with PRM2-deficient sperm resulted in activation of most metaphase II-arrested mouse eggs, but few were able to develop to the blastocyst stage. These findings suggest that development fails because of damage to paternal DNA and that PRM2 is crucial for maintaining the integrity of sperm chromatin.


Subject(s)
DNA Damage , Embryo Loss/etiology , Protamines/genetics , Protamines/metabolism , Spermatozoa/metabolism , Animals , Chimera/genetics , Chromatin/metabolism , Chromatin/ultrastructure , Embryo Loss/genetics , Embryo Loss/metabolism , Female , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Electron , Microscopy, Electron, Scanning , Pregnancy , Spermatozoa/ultrastructure
18.
Biol Reprod ; 67(4): 1268-77, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12297545

ABSTRACT

Ovarian hormonal signaling is essential for proper functioning of the uterus in the establishment of pregnancy. Previous studies have demonstrated that decidualization, a stromal transformation that occurs in response to embryo implantation, can be elicited in the uterus of estrogen receptor alpha knockout (alphaERKO) mice in the absence of the estrogen dependence normally seen in wild-type (WT) mice for this response. While the alphaERKO stromal compartment demonstrated the necessary decidual response, embryo implantation is a process initiated in the epithelial layer, a uterine component that lacks estrogen responsiveness in the alphaERKO. To determine if the alphaERKO uterus would be competent for implantation, donor embryos were transferred into the uterine lumen of WT and alphaERKO females that had been ovariectomized and treated with exogenous estradiol and progesterone to mimic early pregnancy. No implantation occurred in the alphaERKO, while implantation sites containing live embryos were seen in similarly treated WT uteri, indicating that functional estrogen receptor alpha (ERalpha) is required for implantation. Previous observations of estrogen-independent decidualization in the alphaERKO prompted investigation of the mechanism leading to estrogen independence of this process. The disruption of progesterone receptor (PR), Hoxa10, Cox2, or LIF in transgenic mice results in the loss of decidualization response. Therefore, the expression of these genes was studied in WT and alphaERKO uteri by comparing expression following vehicle, progesterone alone (P), or estradiol priming followed by progesterone with nidatory estradiol (E+Pe) and by comparing expression following the above hormonal manipulations in addition to luminal infusion of oil used previously as decidualization-initiating stimulus. The whole-uterus level of PR and Hoxa10 mRNAs did not vary; however, the PR protein was induced in the stroma 24 h after oil infusion. Interestingly, in the WT, this induction was most apparent in samples receiving E+Pe, while in the alphaERKO samples, the induction occurred independent of any hormone priming. Cox2 protein and mRNA increased in both WT and alphaERKO samples 2 h after oil infusion in all three of the treatment groups. In the WT samples, Cox2 levels remained elevated 24 h after oil infusion only in the E+Pe treatment group; however, the elevated Cox2 was seen in samples taken 24 h after oil infusion in all three alphaERKO treatment groups. The alphaERKO uterine tissue appeared to sustain more extensive damage when examined 24 h after oil infusion. Severe trauma, such as crushing of the uterine tissue, has previously been shown to remove the requirement for nidatory estradiol for deciduomas to develop, indicating that the greater susceptibility of alphaERKO uterine tissue to damage from intraluminal oil infusion is contributing to decidualization in the absence of ERalpha. Leukemia inhibitory factor (LIF) mRNA was also induced following estradiol treatment in the WT, but also following oil infusion in WT samples that were not treated with estradiol. In contrast, estradiol does not induce LIF mRNA in the alphaERKO, but oil infusion leads to a robust increase in LIF in all alphaERKO sample groups. LIF binds and activates its membrane receptor, which initiates responses including the phosphorylation and nuclear translocation of Stat3 transcription factor. Thus, Stat3 phosphorylation was studied in WT and alphaERKO samples and found to be induced following oil infusion in all samples. Together, these and previous observations illustrate that estrogen is essential for epithelial proliferation and embryo implantation and that estrogen is dispensable for stromal decidualization in the alphaERKO, as the essential genes and signals required for the response are still induced.


Subject(s)
Decidua/physiology , Embryo Implantation/physiology , Estrogens/physiology , Interleukin-6 , Receptors, Estrogen/deficiency , Signal Transduction/physiology , Uterus/physiology , Animals , Cyclooxygenase 2 , DNA-Binding Proteins/metabolism , Embryo Transfer , Estradiol/pharmacology , Estrogen Receptor alpha , Female , Gene Expression/drug effects , Growth Inhibitors/genetics , Homeobox A10 Proteins , Homeodomain Proteins/genetics , Immunohistochemistry , Isoenzymes/biosynthesis , Isoenzymes/genetics , Leukemia Inhibitory Factor , Lymphokines/genetics , Mice , Mice, Knockout , Phosphorylation , Pregnancy , Progesterone/pharmacology , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/genetics , Receptors, Estrogen/physiology , Receptors, Progesterone/genetics , STAT3 Transcription Factor , Sesame Oil/administration & dosage , Trans-Activators/metabolism
19.
Dev Biol ; 248(2): 331-42, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12167408

ABSTRACT

A-kinase anchoring proteins (AKAPs) tether cyclic AMP-dependent protein kinases and thereby localize phosphorylation of target proteins and initiation of signal-transduction processes triggered by cyclic AMP. AKAPs can also be scaffolds for kinases and phosphatases and form macromolecular complexes with other proteins involved in signal transduction. Akap4 is transcribed only in the postmeiotic phase of spermatogenesis and encodes the most abundant protein in the fibrous sheath, a novel cytoskeletal structure present in the principal piece of the sperm flagellum. Previous studies indicated that cyclic AMP-dependent signaling processes are important in the regulation of sperm motility, and gene targeting was used here to test the hypothesis that AKAP4 is a scaffold for protein complexes involved in regulating flagellar function. Sperm numbers were not reduced in male mice lacking AKAP4, but sperm failed to show progressive motility and male mice were infertile. The fibrous sheath anlagen formed, but the definitive fibrous sheath did not develop, the flagellum was shortened, and proteins usually associated with the fibrous sheath were absent or substantially reduced in amount. However, the other cytoskeletal components of the flagellum were present and appeared fully developed. We conclude that AKAP4 is a scaffold protein required for the organization and integrity of the fibrous sheath and that effective sperm motility is lost in the absence of AKAP4 because signal transduction and glycolytic enzymes fail to become associated with the fibrous sheath.


Subject(s)
Carrier Proteins/metabolism , Seminal Plasma Proteins/metabolism , Sperm Motility , Sperm Tail/metabolism , Animals , Carrier Proteins/genetics , Cyclic AMP/metabolism , Gene Deletion , Gene Expression Regulation , Infertility, Male/genetics , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Microscopy, Phase-Contrast , Mutation , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Seminal Plasma Proteins/genetics , Sperm Tail/ultrastructure , Tubulin/metabolism
20.
Cancer Res ; 62(4): 990-4, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11861370

ABSTRACT

Inherited mutations of the human BRCA2 gene confer increased risks for developing breast, ovarian, and several other cancers. Unlike previously described Brca2 knockout mice that display predominantly embryonic lethal phenotypes, we developed mice with a homozygous germ-line deletion of Brca2 exon 27 that exhibit a moderate decrease in perinatal viability and are fertile. We deleted this Brca2 COOH-terminal domain because it interacts directly with the Rad51 protein, contains a nuclear localization signal, and is required to maintain genomic stability in response to various types of DNA damage. These homozygous Brca2-mutant mice have a significantly increased overall tumor incidence and decreased survival compared with their heterozygous littermates. Virgin female mice homozygous for this Brca2 mutation also display an inhibition of ductal side branching in the mammary gland at 6 months of age. Given their substantial viability and cancer predisposition, these mutant mice will be useful to further define the role of the COOH-terminal Brca2 domain in tumorigenesis both in vivo and in vitro.


Subject(s)
Genes, BRCA2 , Neoplasms, Experimental/genetics , Animals , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Exons/genetics , Female , Gene Deletion , Genetic Predisposition to Disease , Inbreeding , Male , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/physiology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/pathology , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...