Literature
首页医源资料库在线期刊美国病理学杂志2006年第168卷第11期

Localization of Ubiquitin C-Terminal Hydrolase L in Mouse Ova and Its Function in the Plasma Membrane to Block Polyspermy

来源:《美国病理学杂志》
摘要:Ubiquitincarboxyl-terminalhydrolaseL1(UCH-L1)isexclusivelyexpressedinneurons,testis,ovary,andplacenta,eachofwhichhasuniquebiologicalactivities。--------------------------------------------------------------------------------UbiquitinC-terminalhydrolaseL1(UCH-L1)isoneof......

点击显示 收起

【摘要】  Protein degradation is essential for oogenesis and embryogenesis. The ubiquitin-proteasome system regulates many cellular processes via the rapid degradation of specific proteins. Ubiquitin carboxyl-terminal hydrolase L1 (UCH-L1) is exclusively expressed in neurons, testis, ovary, and placenta, each of which has unique biological activities. However, the functional role of UCH-L1 in mouse oocytes remains unknown. Here, we report the expression pattern of UCH-L1 and its isozyme UCH-L3 in mouse ovaries and embryos. Using immunocytochemistry, UCH-L1 was selectively detected on the plasma membrane, whereas UCH-L3 was mainly detected in the cytoplasm, suggesting that these isozymes have distinct functions in mouse eggs. To further investigate the functional role of UCH-L1 in mouse eggs, we analyzed the fertilization rate of UCH-L1-deficient ova of gad female mice. Female gad mice had a significantly increased rate of polyspermy in in vitro fertilization assays, although the rate of fertilization did not differ significantly from wild-type mice. In addition, the litter size of gad female mice was significantly reduced compared with wild-type mice. These results may identify UCH-L1 as a candidate for a sperm-oocyte interactive binding or fusion protein on the plasma membrane that functions during the block to polyspermy in mouse oocytes.
--------------------------------------------------------------------------------
Ubiquitin C-terminal hydrolase L1 (UCH-L1) is one of many deubiquitinating enzymes and is selectively and abundantly expressed in the ovary, placenta, testis, and neuronal cells.1-6 Recent studies suggest that UCH-L1 associates with monoubiquitin and prolongs ubiquitin half-life in neurons.7 Our previous work on UCH-L1 function in gad mice suggests that these mice are resistant to apoptotic stress in retinal cells and testicular germ cells.8,9 This observation is consistent with a recent report that the overexpression of UCH-L1 induces testicular germ cell apoptosis in Uchl1 transgenic mice.10 Furthermore, both UCH-L1 and UCH-L3, the predominant functional UCHs, are differentially expressed in testis during spermatogenesis. These results demonstrate that these enzymes have distinct functions in the testis and epididymis after apoptotic stress,9,11 even though they have high (52%) amino acid sequence identity.12
The above data are in accordance with a number of studies that have linked inhibition of the ubiquitin-proteasome system (UPS) with suppression of apoptosis.8,13-15 UCH-L1 is an important enzyme for UPS-dependent proteolysis and plays a regulatory role in the cell cycle and cellular proliferation. Thus, its expression in placenta is of considerable interest.3,4 Recent studies reported that UPS controls the degradation of various substrates during gametogenesis and fertilization,16-19 but relatively little is known about the functional role of the UPS in fertilization. UCH-L1 is expressed in oocytes in ovaries.5,20 Oocytes, as well as spermatogonia in testis, have multiple potentials and activities for development. However, the function of UCH-L1 during oogenesis is unclear. RFPL4 (ret finger protein-like 4) and FAM (fat facets in mouse) are involved in regulating oogenesis.21,22 RFPL4 is highly expressed during oogenesis and functions as an E3 ubiquitin ligase to target proteins for proteasomal degradation.21 FAM is a developmentally regulated substrate-specific deubiquitinating enzyme that is required for preimplantation of the mouse embryo.22 Thus, the UPS might be important during oocyte development and differentiation of the embryo after fertilization.
Here, we analyzed the functional role of UCH-L1 using mouse oocytes and embryos. Our results indicate that UCH-L1 is selectively expressed on the plasma membrane of mouse ova, where it may regulate membrane penetration by spermatozoa. In addition, the unique expression patterns of UCH-L1 and UCH-L3 suggest that these proteins have distinct functions during oogenesis and embryogenesis. Our results therefore provide strong evidence that UCH-L1 functions in the polyspermy block during mammalian fertilization.

【关键词】  localization ubiquitin c-terminal hydrolase function membrane polyspermy



Materials and Methods


Animals


We used 8-week-old BDF1, gad (CBA/RFM),23,24 and Uchl3 knockout (C57BL/6J)12,25 female and male mice. BDF1 mice were purchased from Nihon SLC, Inc. (Hamamatsu, Japan). The gad mouse is an autosomal recessive mutant that was obtained by crossing CBA and RFM mice. The gad line was maintained by intercrossing for more than 20 generations.23,24 The Uchl3 knockout mouse was generated by standard methods using homologously recombinant ES cells from 129SV mice.12,25 The knockout line was back-crossed several times with C57BL/6J mice. gad mice were maintained at our institute, and Uchl3 knockout mice were maintained at the National Institute of Neuroscience, National Center of Neurology and Psychiatry (Tokyo, Japan). Animal care and handling were in accordance with institutional regulations and were approved by the Animal Care and Use Committee of the University of Tokyo.


Oocyte Collection and in Vitro Fertilization


Female mice were superovulated by intraperitoneal injections with 5 IU of pregnant mare serum gonadotropin (Sankyo, Tokyo, Japan) for 48 hours, followed by 5 IU of human chorionic gonadotropin (Sankyo). Ovulated eggs were collected from the ampullae of oviducts by the scratching method 16 hours after human chorionic gonadotropin injection and placed in 400-µl droplets of Toyoda, Yokoyama, and Hoshi (TYH)26 containing 0.4 mg/ml bovine serum albumin (Sigma-Aldrich, St. Louis, MO). Spermatozoa were collected from the cauda epididymis of male mice and preincubated for 1 hour in 400 µl of TYH to allow capacitation before insemination. After capacitation, the sperms were introduced into the fertilization medium at a final concentration of 150 spermatozoa/µl. At 4 hours after insemination, 0.05% hyaluronidase (Sigma-Aldrich) was added to the medium for 5 minutes. The eggs were washed thoroughly three times and then cultured in potassium simplex optimized medium (KSOM).26 After fertilization, all embryos were incubated in a humidified atmosphere of 5% CO2 in air at 37??C in 100-µl drops of KSOM overlaid with mineral oil. To analyze fertilization in gad mice, gad (n = 5) and wild-type (CBA/RFM) (n = 5) female mice were superovulated. Ovulated oocytes of gad and wild-type mice were fertilized with wild-type spermatozoa.


Western Blotting


Total protein of ovary extracts (10 µg/lane), oocytes, or embryos (20 oocytes or embryos per lane) was subjected to sodium dodecyl sulfate-polyacrylamide gel electrophoresis using 12.5% gels. Proteins were electrophoretically transferred to polyvinylidene difluoride membranes (Bio-Rad, Hercules, CA) and blocked with 1% bovine serum albumin in TBS-T . The membranes were incubated individually with primary antibodies against UCH-L1, UCH-L3,2 monoubiquitin (U5379, Sigma-Aldrich), zona pellucida 2 (ZP2; Santa Cruz Biotechnology, Santa Cruz, CA), and zona pellucida 3 (ZP3) (Santa Cruz Biotechnology). After thorough rinsing, blots were further incubated with peroxidase-conjugated goat anti-rabbit IgG (DakoCytomation, Glostrup, Denmark) for 1 hour at room temperature. Immunoreactions were visualized by enhanced chemiluminescence (ECL Plus; GE Healthcare UK Ltd. Amersham Place, Little Chalfont, Buckinghamshire, UK). Each immunoreactive band was quantified using commercially available software (Quantity One; PDI, Upper Saddle River, NJ). Negative control extracts of ovary or oocytes were obtained from gad and Uchl3 knockout mice.


Histological and Immunochemical Assessment


Ovaries of BDF1 female and gad mice were fixed in 4% paraformaldehyde, embedded in paraffin wax, and then sectioned at 4-µm thickness. Ovary sections of gad mice were stained with hematoxylin and eosin (H&E). Light microscopy was used for routine observations. For immunohistochemical staining, the sections were incubated with Block Ace (Dainippon Sumitomo Pharma, Osaka, Japan) for 1 hour at room temperature followed by incubation overnight at 4??C with a rabbit polyclonal antibody against UCH-L1 and UCH-L3.2 The sections were then incubated with biotinylated goat anti-rabbit IgG (DAKO), which was followed by incubation with streptavidin-biotin-horseradish peroxidase complex (sABC kit; DAKO). Immunoreactivity was visualized by treating the sections with 3,3'-diaminobenzidine tetroxide (Dojin Kagaku, Kumamoto, Japan). Finally, the sections were counterstained with hematoxylin. Negative control ovaries were obtained from gad and Uchl3 knockout mice.


For immunocytochemical staining, whole oocytes or embryos were fixed for 30 minutes with 4% paraformaldehyde in phosphate-buffered saline (PBS) and 0.2% (w/v) Triton X-100 (ICN Biomedicals, Aurora, OH) in PBS for 30 minutes. Nonspecific binding of immunoglobulins was blocked by incubation with Block Ace (Dainippon Pharmaceutical, Ltd.) for 1 hour at room temperature. The sections were then incubated with primary antibodies against UCH-L1, UCH-L3, and lectin with Rhodamine-Lens Culinaris Agglutinin (Vector Laboratories, Burlingame, CA). The sections were then incubated with Alexa 488-conjugated goat anti-rabbit IgG (Molecular Probes, Eugene, OR) and propidium iodide (Molecular Probes). Stained sections were observed under a confocal laser microscope (Laser Scanning Microscope 510; Carl Zeiss, Jena, Germany). Negative control oocytes were obtained from gad and Uchl3 knockout mice by superovulation.


Quantitative Analysis


Normal oocytes were identified by the presence of the first polar body.27,28 The frequency of normal oocytes was calculated by counting the normal oocytes in the total superovulated oocytes of gad (n = 173) and wild-type (n = 148) mice. To determine the fertilization rate, putative fertilized eggs (by in vitro fertilization) were fixed in acetic alcohol (1:3, glacial acetic acid/ethanol) and then stained with 1% aceto-orcein to visualize pronuclei and assess sperm penetration and incidence of monospermic and polyspermic fertilization. Polyspermic fertilization was defined as the presence of three or more pronuclei. The rate of polyspermic fertilization was calculated by counting the polyspermic eggs among the total fertilized eggs of gad (n = 71) and wild-type (n = 56) mice.


Electron Microscopic Analysis


Ovulated mature oocytes and zygotes of BDF1 mice were fixed with 4% paraformaldehyde, and frozen sections were prepared for electron microscopy. The sections were incubated with an antibody against UCH-L1.2 Subsequently, the ABC method was performed as indicated by the supplier, and the peroxidase reaction was developed in diaminobenzidine. Immunostained sections were fixed in 2.5% glutaraldehyde, postfixed in 1% OsO4, dehydrated in a graded series of ethanol, and embedded in Epon 812.29 Ultrathin sections were cut with an ultramicrotome, stained with uranyl acetate,29 and examined with an electron microscope H-7100 (Hitachi, Hitachinaka, Japan).


Breeding Test of gad Female Mice


gad (n = 12) and wild-type (CBA/RFM) (n = 15) female mice and wild-type (CBA/RFM) (n = 9) male mice were subjected to a breeding study. Three female mice of the same genotype were housed with one male mouse per cage. Cages were monitored daily at midday, and the appearance of a vaginal plug was recognized as day 0.5 of gestation. The number of pups, litters, and litter size of gad and wild-type female mice were recorded.


Statistical Analysis


The mean and SD were calculated for all data (presented as mean ?? SD). The Student??s t-test was used for all statistical analyses.


Results


Expression of UCH-L1 and UCH-L3 in Mouse Ovaries


We first used Western blotting to address whether both UCH-L1 and UCH-L3 levels are expressed in ovaries during proestrus, estrus, metestrus, and diestrus (Figure 1A) . Both proteins were detected at all estrous cycle stages in wild-type mice. As expected, UCH-L1 was not detected in gad ovaries and UCH-L3 was not detected in Uchl3 knockout ovaries. The levels of UCH-L1 and UCH-L3 in ovaries did not change throughout the estrous cycle (Figure 1A) .


Figure 1. Expression of UCH-L1 and UCH-L3 in mouse ovaries. A: Western blotting analysis of UCH-L1 and UCH-L3 in ovaries during the estrous cycle. Proestrus (P), estrus (E), metestrus (M), and diestrus (D) ovaries; gad/gad and L3C/C represent gad and Uchl3 knockout mouse ovaries, respectively. B: Immunohistochemical analyses of UCH-L1 (aCe) and UCH-L3 (gCk) in BDF1 mouse ovaries (aCe, gCk). Ovaries from gad (f) and Uchl3 knockout mice (l) are negative controls. UCH-L1 is highly expressed on the plasma membrane of oocytes. UCH-L3 is diffusely expressed in the cytoplasm of oocytes. Follicle stages: primordial (a, g), primary (b, h), secondary (c, i), tertiary (d, j), and mature (e, k) from BDF1 mouse ovaries. Scale bars = 10 µm.


We next used immunohistochemistry to address whether both UCH-L1 and UCH-L3 are present in developing follicles (primordial, primary, secondary, tertiary, and mature follicles; Figure 1B ). In the ovary, UCH-L1 staining was most intense at the plasma membrane of oocytes in developing follicles (Figure 1B, aCe) , whereas UCH-L3 staining was concentrated in the cytoplasm of oocytes (Figure 1B, gCk) . The oocytes of gad and Uchl3 knockout mice ovaries were negative for UCH-L1 and UCH-L3, respectively (Figure 1B, f and l) .


Expression of UCH-L1 and UCH-L3 in Mouse Mature Oocytes and Preimplantation Embryos


Using Western blotting, we examined the levels of both UCH-L1 and UCH-L3 in mature oocytes and during embryogenesis (zygote, two cells, four cells, eight cells, morulas, and blastocysts). UCH-L1 and UCH-L3 were detected in mature oocytes and during all of the embryonic stages we tested; the level of UCH-L1 was essentially constant in all cases, but the level of UCH-L3 was lower in the blastocyst stage (Figure 2A) .


Figure 2. Expression of UCH-L1 and UCH-L3 in mature oocytes and fertilized eggs. A: Western blotting analysis of UCH-L1 and UCH-L3 in ovulated mature oocytes and developing eggs of BDF1 mice. B: Immunocytochemical analysis of UCH-L1 (aCg) and UCH-L3 (iCo) in ovulated mature oocytes and developing embryos of BDF1 mice (aCg, iCo). Ovulated mature oocytes of gad (h) and Uchl3 knockout mice (p) are negative controls. UCH-L1 immunoreactivity was seen only on the plasma membrane of mature oocytes (a) and developing eggs (bCg). UCH-L3 immunoreactivity was seen in the cytoplasm of oocytes (j) and developing eggs (jCo). Stages: oocytes (a, i), zygotes (b, j), two cells (c, k), four cells (d, l), eight cells (e, m), morulas (f, n), and blastocyst (g, o). Nuclei are shown in red. Scale bar = 20 µm.


We further analyzed the distribution of UCH-L1 and UCH-L3 in ovulated mature oocytes and during embryogenesis using immunocytochemistry (Figure 2B) . UCH-L1 immunoreactivity was intense on the plasma membrane of oocytes and developing embryos but not in the cytoplasm of eggs (Figure 2B, aCg) . Furthermore, UCH-L1 immunoreactivity was observed continuously during embryogenesis (Figure 2B, cCg) . In the blastocyst stage, UCH-L1 was observed in the outer layer cells of the trophectoderm (Figure 2B, g) . UCH-L3 immunoreactivity was seen in the cytoplasm of oocytes and developing embryos (Figure 2B, iCo) , as was detected in ovaries (Figure 2A) . In the blastocyst stage, UCH-L3 was observed in the inner cells (Figure 2B, o) . The oocytes and developing embryos of gad and Uchl3 knockout mice were negative for UCH-L1 and UCH-L3, respectively (Figure 2B, h and p) .


Immunoelectron Microscopy


The cortical granule is located right under the plasma membrane in oocytes. This organelle is unique to oocytes and plays an important role in fertilization. At the light microscope level, it is difficult to determine whether UCH-L1 immunoreactivity localizes to the plasma membrane or cortical granule. To address this issue, we investigated the subcellular localization of UCH-L1 in ovulated mature oocytes and zygotes at the ultrastructural level (Figure 3) . Intense UCH-L1 immunoreactivity was observed on the plasma membrane of ovulated mature oocytes (Figure 3, A and B) and zygotes (Figure 3, C and D) . However, the intensity of the plasma membrane immunoreactivity was greater in mature oocytes than in zygotes. Therefore, we concluded that the distribution of UCH-L1 may change after fertilization.


Figure 3. Localization of UCH-L1 on the plasma membrane in mature oocytes and zygotes by immunoelectron microscopy. ACD: Mature oocyte and zygote of BDF1 mouse are immunostained with UCH-L1. B and D: Magnified images of the region enclosed by small rectangles in A and C. UCH-L1 immunoreactivity is intense on the plasma membrane (arrowheads) of an ovulated mature oocyte (B) and zygote (D). The position of cytoplasm (Cy) is indicated. Scale bars = 1 µm.


Morphology of the gad Ovaries and Ovulated Oocytes


To assess whether gad mice have morphologically normal ovaries, we used histology to compare ovaries from gad and wild-type (CBA/RFM) mice. gad mouse ovaries had morphologically normal oocytes, follicles, and corpora lutea (Figure 4, A and B) . In addition, gad mice had a normal estrous cycle (data not shown). We further assessed gad mouse ovarian function by comparison with wild-type mice using the ovulation test. The total number of ovulated oocytes and the normal oocyte ovulation rate of superovulated oocytes did not differ significantly between gad mice and wild-type mice (Figure 4, C and D) .


Figure 4. Histology of ovaries and the ovulation rate of gad mice. A and B: Ovarian sections of gad mice stained with H&E. gad mouse ovaries have morphologically normal oocytes, follicles, and corpora lutea (CL). C and D: The average number of superovulated oocytes (C) and the normal oocyte rate (normal oocytes/total ovulated oocytes) (D) in gad mice are not significantly different from wild-type mice. Scale bars = 200 µm.


High Polyspermy Rate in gad Mouse Oocytes


To analyze the fertilization rate in UCH-L1-deficient embryos of gad female mice, we assessed fertilization by in vitro fertilization using wild-type spermatozoa. For mice, the presence of more than three or more pronuclei defines polyspermic fertilization. Fertilized eggs from gad mice had characteristics consistent with this definition of polyspermy (Figure 5, B and C) . By contrast, wild-type eggs showed normal zygotic nuclei (Figure 5A) . The fertilized eggs of gad mice had a significantly higher rate of polyspermy (27 ?? 0.04%) compared with wild-type mice (2 ?? 0.02%) (Figure 5E) . However, the fertility of gad mouse eggs (46 ?? 0.06%) did not differ significantly from that of wild-type mice (43 ?? 0.09%) (Figure 5D) .


Figure 5. Incidence of polyspermy in gad mice. Fertilized eggs were stained with aceto-orcein (A, B) or visualized by inverted microscopy (C). A: Normal fertilized egg with two pronuclei (arrows in ACC) in wild-type mice. B and C: Polyspermic fertilized eggs in gad mice have three or four pronuclei. D: The fertility rate in gad mice is not significantly different from wild-type mice. E: Polyspermic rate in gad mice is significantly higher than that of wild-type mice. *P < 0.01. Scale bars = 20 µm.


Low Breeding Rate of gad Female Mice


To further evaluate the high polyspermic fertilization rate of gad female mice, we characterized the litter size of these mice after mating with wild-type male mice. gad female mice exhibited normal puberty and estrous cycle, as assessed by vaginal opening and vaginal smear, but they had reduced fertility, as evidenced by a significant decrease in litter size (3 ?? 2.0) compared with wild-type mice (6.6 ?? 1.3) (Table 1) .


Table 1. Breeding Study of gad and Wild-Type Female Mice with Wild-Type Male Mice


Monoubiquitin Level in gad Mouse Oocytes


Our previous study demonstrated that UCH-L1 stabilizes monoubiquitin in testes and that the level of monoubiquitin is decreased in male gad mice.8,9 To determine whether the high polyspermy rate in gad mouse oocytes correlated with a reduced level of monoubiquitin in the oocytes, we used Western blotting to measure monoubiquitin levels in oocytes from gad mice and wild-type mice. The monoubiquitin level was substantially lower in gad mouse oocytes (Figure 6A) . However, wild-type and gad mouse oocytes showed the normal zona reaction (postfertilization proteolytic cleavage of ZP2) and normal cortical reaction (postfertilization exocytosis of cortical granules) (Figure 6, B and C) .


Figure 6. Expression of monoubiquitin, zona reaction, and cortical reaction in gad mice. A: Western blotting analysis of monoubiquitin of wild-type and gad mouse oocytes. The monoubiquitin protein level is substantially lower in gad than in wild-type. B: Western blotting analysis of ZP2 and ZP3 of wild-type and gad mouse oocytes and zygotes. Disappearance of ZP2 of zygotes because of postfertilization proteolytic cleavage is observed in wild-type and gad mice. C: Cortical reaction analysis of wild-type and gad mouse oocytes and zygotes. Disappearance of rhodamine-LCA of zygotes because of postfertilization cortical granule exocytosis is observed in wild-type and gad mice. Scale bars = 20 µm.


Discussion


Polyspermy refers to the penetration of more than one sperm into oocytes during fertilization and is currently considered a pathological phenomenon in mammals.30 However, an exceptionally high incidence of polyspermic fertilization has been revealed in pig fertilization.31,32 The reasons for this higher incidence of polyspermy in oocytes are not clear, and relatively little is known about the exact mechanisms that prevent polyspermy. The mechanism that prevents polyspermy is classically illustrated by the zona reaction (cortical reaction) and subsequent plasma membrane block.33,34 After penetration by sperm, the zona reaction occurs, and cortical contents are extruded into the perivitelline space, thereby preventing polyspermic penetration by hardening of the zona pellucida.35 Plasma membrane block is assumed to result from changes (eg, the destruction of sperm receptors in the membrane) that preclude sperm adherence.36,37


Recent studies have demonstrated that the UPS is responsible for extracellular degradation of the sperm receptor on the outer face of zona pellucida and that proteasomal inhibitors block sperm penetration of the zona pellucida during fertilization.17-19 These findings strongly support a role for the UPS in the sperm-oocyte interaction of the zona pellucida. However, our results showed the apparent contradiction that the high incidence of polyspermy was caused by down-regulation of the UPS function distinct from previous studies.17-19 Our present work shows that UCH-L1 is exclusively localized on the oocyte plasma membrane and that UCH-L1-deficient embryos of gad female mice have a significantly increased polyspermy rate in vitro (Figures 2B and 5) . These results suggest that UCH-L1 may modify plasma membrane components, thereby preventing multiple sperm entry. In mammals, it has been suggested that UCH-L1 associates with monoubiquitin7,10 ; moreover, the monoubiquitin pool is reduced in gad mice relative to wild-type mice. We have shown that the monoubiquitin level in gad mouse oocytes is substantially reduced relative to that of wild-type mice (Figure 6A) . These results suggest that the high incidence of polyspermy in gad mice may have its basis in the down-regulation of the UPS because of a reduction in available monoubiquitin at the plasma membrane, even though gad mouse oocytes undergo a normal zona reaction (Figure 6, B and C) .30,38 In addition, gad mice also have morphologically normal ovary development and a normal rate of ovulation compared with wild-type mice (Figure 4) .


CD9 is an integral membrane protein associated with integrins and other membrane proteins.39 CD9 is extensively localized on the oocyte plasma membrane.39-41 Recent studies suggest that CD9 participates in sperm-oocyte fusion in the mouse.39 CD9 knockout female mice ovulate normally, but the oocytes are rarely fertilized because CD9 deficiency on the plasma membrane inhibits sperm-oocyte fusion. Many plasma membrane channels, transporters, and receptors undergo ubiquitination at the extracellular face, which is required for internalization and subsequent entry into the endocytic pathway.42 These studies indicate that ubiquitination (monoubiquitination, multiple monoubiquitination, or polyubiquitination) of plasma membrane proteins might facilitate plasma membrane block.


In the present study, we showed that both UCH-L1 and UCH-L3 are strongly expressed throughout all stages of oogenesis and embryogenesis (Figure 1A) , even though ovarian and uterine functions change periodically because of fluctuations in hormone levels.43-45 Our previous studies suggested that UCH-L1 and UCH-L3, despite their high sequence homology,12 have distinct expression patterns and differential function in testis and epididymis.9,11 Here we show that these two proteins have distinct distributions: UCH-L1 is exclusively expressed on the plasma membrane of oocytes and embryos, whereas UCH-L3 is diffusely expressed in the cytoplasm (Figures 1B and 2B) . Therefore, it is conceivable that UCH-L1 and UCH-L3 have different functional roles in oocytes and embryos, as was shown in the testis/epididymis.9,11 Although our studies show that UCH-L3 is highly expressed in the cytoplasm of oocytes and embryos, we found no difference in the fertilization rate and litter size in Uchl3 knockout female mice compared with wild-type (C57BL/6J) female mice (data not shown). Thus, UCH-L3 may not impact these developmentally related processes; further research is necessary to elucidate the cytoplasmic function of this UCH.


Polyspermy in humans mostly results in embryonic lethality or triploid embryos, which usually mature into infertile offspring.46,47 According to the World Health Organization, 80 million people worldwide are infertile. Infertility may result from female- or male-derived factors, a combination of the two, or as yet unidentified biological factors.48 Our present work shows that a deficiency in UCH-L1 may be a new cause of female infertility. Thus, gad mice may be a useful model for further studies of infertility.


In conclusion, we show that UCH-L1-deficient gad female mice oocytes have a significantly increased rate of polyspermy in an in vitro fertilization assay; consequently, these mice have significantly decreased litter size. These results suggest that UCH-L1 is a crucial factor in the plasma membrane block to polyspermy in mouse oocytes.


Acknowledgements


We thank Kunihiko Naito and Koji Kashima (University of Tokyo) for technical assistance with in vitro fertilization.


【参考文献】
  Kwon J, Kikuchi T, Setsuie R, Ishii Y, Kyuwa S, Yoshikawa Y: Characterization of the testis in congenitally ubiquitin carboxy-terminal hydrolase-1 (Uch-L1) defective (gad) mice. Exp Anim 2003, 52:1-9

Kwon J, Wang YL, Setsuie R, Sekiguchi S, Sakurai M, Sato Y, Lee WW, Ishii Y, Kyuwa S, Noda M, Wada K, Yoshikawa Y: Developmental regulation of ubiquitin C-terminal hydrolase isozyme expression during spermatogenesis in mice. Biol Reprod 2004, 71:515-521

Sekiguchi S, Takatori A, Negishi T, Kwon J, Kokubo T, Ishii Y, Kyuwa S, Yoshikawa Y: Localization of ubiquitin carboxyl-terminal hydrolase-L1 in cynomolgus monkey placentas. Placenta 2005, 26:99-103

Sekiguchi S, Yoshikawa Y, Tanaka S, Kwon J, Ishii Y, Kyuwa S, Wada K, Nakamura S, Takahashi K: Immunohistochemical analysis of protein gene product 9.5, a ubiquitin carboxyl-terminal hydrolase, during placental and embryonic development in the mouse. Exp Anim 2003, 52:365-369

Wilson PO, Barber PC, Hamid QA, Power BF, Dhillon AP, Rode J, Day IN, Thompson RJ, Polak JM: The immunolocalization of protein gene product 9.5 using rabbit polyclonal and mouse monoclonal antibodies. Br J Exp Pathol 1988, 69:91-104

Kwon J, Mochida K, Wang YL, Sekiguchi S, Sankai T, Aoki S, Ogura A, Yoshikawa Y, Wada K: Ubiquitin C-terminal hydrolase L-1 is essential for the early apoptotic wave of germinal cells and for sperm quality control during spermatogenesis. Biol Reprod 2005, 73:29-35

Osaka H, Wang YL, Takada K, Takizawa S, Setsuie R, Li H, Sato Y, Nishikawa K, Sun YJ, Sakurai M, Harada T, Hara Y, Kimura I, Chiba S, Namikawa K, Kiyama H, Noda M, Aoki S, Wada K: Ubiquitin carboxy-terminal hydrolase L1 binds to and stabilizes monoubiquitin in neuron. Hum Mol Genet 2003, 12:1945-1958

Harada T, Harada C, Wang YL, Osaka H, Amanai K, Tanaka K, Takizawa S, Setsuie R, Sakurai M, Sato Y, Noda M, Wada K: Role of ubiquitin carboxy terminal hydrolase-L1 in neural cell apoptosis induced by ischemic retinal injury in vivo. Am J Pathol 2004, 164:59-64

Kwon J, Wang YL, Setsuie R, Sekiguchi S, Sato Y, Sakurai M, Noda M, Aoki S, Yoshikawa Y, Wada K: Two closely related ubiquitin C-terminal hydrolase isozymes function as reciprocal modulators of germ cell apoptosis in cryptorchid testis. Am J Pathol 2004, 165:1367-1374

Wang YL, Liu W, Sun YJ, Kwon J, Setsuie R, Osaka H, Noda M, Aoki S, Yoshikawa Y, Wada K: Overexpression of ubiquitin carboxyl-terminal hydrolase L1 arrests spermatogenesis in transgenic mice. Mol Reprod Dev 2006, 73:40-49

Kwon J, Sekiguchi S, Wang YL, Setsuie R, Yoshikawa Y, Wada K: The region-specific functions of two ubiquitin C-terminal hydrolase isozymes along the epididymis. Exp Anim 2006, 55:35-43

Kurihara LJ, Semenova E, Levorse JM, Tilghman SM: Expression and functional analysis of Uch-L3 during mouse development. Mol Cell Biol 2000, 20:2498-2504

Rasoulpour RJ, Schoenfeld HA, Gray DA, Boekelheide K: Expression of a K48R mutant ubiquitin protects mouse testis from cryptorchid injury and aging. Am J Pathol 2003, 163:2595-2603

Yang Y, Yu X: Regulation of apoptosis: the ubiquitous way. FASEB J 2003, 17:790-799

Baarends WM, Wassenaar E, Hoogerbrugge JW, van Cappellen G, Roest HP, Vreeburg J, Ooms M, Hoeijmakers JH, Grootegoed JA: Loss of HR6B ubiquitin-conjugating activity results in damaged synaptonemal complex structure and increased crossing-over frequency during the male meiotic prophase. Mol Cell Biol 2003, 23:1151-1162

Baarends WM, Roest HP, Grootegoed JA: The ubiquitin system in gametogenesis. Mol Cell Endocrinol 1999, 151:5-16

Sutovsky P: Ubiquitin-dependent proteolysis in mammalian spermatogenesis, fertilization, and sperm quality control: killing three birds with one stone. Microsc Res Tech 2003, 61:88-102

Sutovsky P, Manandhar G, McCauley TC, Caamano JN, Sutovsky M, Thompson WE, Day BN: Proteasomal interference prevents zona pellucida penetration and fertilization in mammals. Biol Reprod 2004, 71:1625-1637

Sutovsky P, McCauley TC, Sutovsky M, Day BN: Early degradation of paternal mitochondria in domestic pig (Sus scrofa) is prevented by selective proteasomal inhibitors lactacystin and MG132. Biol Reprod 2003, 68:1793-1800

Ellederova Z, Halada P, Man P, Kubelka M, Motlik J, Kovarova H: Protein patterns of pig oocytes during in vitro maturation. Biol Reprod 2004, 71:1533-1539

Suzumori N, Burns KH, Yan W, Matzuk MM: RFPL4 interacts with oocyte proteins of the ubiquitin-proteasome degradation pathway. Proc Natl Acad Sci USA 2003, 100:550-555

Pantaleon M, Kanai-Azuma M, Mattick JS, Kaibuchi K, Kaye PL, Wood SA: FAM deubiquitylating enzyme is essential for preimplantation mouse embryo development. Mech Dev 2001, 109:151-160

Kikuchi T, Mukoyama M, Yamazaki K, Moriya H: Axonal degeneration of ascending sensory neurons in gracile axonal dystrophy mutant mouse. Acta Neuropathol (Berl) 1990, 80:145-151

Saigoh K, Wang YL, Suh JG, Yamanishi T, Sakai Y, Kiyosawa H, Harada T, Ichihara N, Wakana S, Kikuchi T, Wada K: Intragenic deletion in the gene encoding ubiquitin carboxy-terminal hydrolase in gad mice. Nat Genet 1999, 23:47-51

Kurihara LJ, Kikuchi T, Wada K, Tilghman SM, Semenova E, Levorse JM: Loss of Uch-L1 and Uch-L3 leads to neurodegeneration, posterior paralysis and dysphagia. Hum Mol Genet 2001, 10:1963-1970

Kito S, Hayao T, Noguchi-Kawasaki Y, Ohta Y, Hideki U, Tateno S: Improved in vitro fertilization and development by use of modified human tubal fluid and applicability of pronucleate embryos for cryopreservation by rapid freezing in inbred mice. Comp Med 2004, 54:564-570

Wang WH, Abeydeera LR, Han YM, Prather RS, Day BN: Morphologic evaluation and actin filament distribution in porcine embryos produced in vitro and in vivo. Biol Reprod 1999, 60:1020-1028

Ju JC, Tseng JK: Nuclear and cytoskeletal alterations of in vitro matured porcine oocytes under hyperthermia. Mol Reprod Dev 2004, 68:125-133

Ishibashi S, Sakaguchi M, Kuroiwa T, Yamasaki M, Kanemura Y, Shizuko I, Shimazaki T, Onodera M, Okano H, Mizusawa H: Human neural stem/progenitor cells, expanded in long-term neurosphere culture, promote functional recovery after focal ischemia in Mongolian gerbils. J Neurosci Res 2004, 78:215-223

Sun QY: Cellular and molecular mechanisms leading to cortical reaction and polyspermy block in mammalian eggs. Microsc Res Tech 2003, 61:342-348

Xia P, Wang Z, Yang Z, Tan J, Qin P: Ultrastructural study of polyspermy during early embryo development in pigs, observed by scanning electron microscope and transmission electron microscope. Cell Tissue Res 2001, 303:271-275

Sherrer ES, Rathbun TJ, Davis DL: Fertilization and blastocyst development in oocytes obtained from prepubertal and adult pigs. J Anim Sci 2004, 82:102-108

Lambert C, Goudeau H, Franchet C, Lambert G, Goudeau M: Ascidian eggs block polyspermy by two independent mechanisms: one at the egg plasma membrane, the other involving the follicle cells. Mol Reprod Dev 1997, 48:137-143

Wang WH, Abeydeera LR, Prather RS, Day BN: Morphologic comparison of ovulated and in vitro-matured porcine oocytes, with particular reference to polyspermy after in vitro fertilization. Mol Reprod Dev 1998, 49:308-316

Hatanaka Y, Nagai T, Tobita T, Nakano M: Changes in the properties and composition of zona pellucida of pigs during fertilization in vitro. J Reprod Fertil 1992, 95:431-440

Sengoku K, Tamate K, Horikawa M, Takaoka Y, Ishikawa M, Dukelow WR: Plasma membrane block to polyspermy in human oocytes and preimplantation embryos. J Reprod Fertil 1995, 105:85-90

Horvath PM, Kellom T, Caulfield J, Boldt J: Mechanistic studies of the plasma membrane block to polyspermy in mouse eggs. Mol Reprod Dev 1993, 34:65-72

Ducibella T: The cortical reaction and development of activation competence in mammalian oocytes. Hum Reprod Update 1996, 2:29-42

Miyado K, Yamada G, Yamada S, Hasuwa H, Nakamura Y, Ryu F, Suzuki K, Kosai K, Inoue K, Ogura A, Okabe M, Mekada E: Requirement of CD9 on the egg plasma membrane for fertilization. Science 2000, 287:321-324

Chen MS, Tung KS, Coonrod SA, Takahashi Y, Bigler D, Chang A, Yamashita Y, Kincade PW, Herr JC, White JM: Role of the integrin-associated protein CD9 in binding between sperm ADAM 2 and the egg integrin alpha6beta1: implications for murine fertilization. Proc Natl Acad Sci USA 1999, 96:11830-11835

Li YH, Hou Y, Ma W, Yuan JX, Zhang D, Sun QY, Wang WH: Localization of CD9 in pig oocytes and its effects on sperm-egg interaction. Reproduction 2004, 127:151-157

Hicke L, Dunn R: Regulation of membrane protein transport by ubiquitin and ubiquitin-binding proteins. Annu Rev Cell Dev Biol 2003, 19:141-172

Gava N, Clarke CL, Byth K, Arnett-Mansfield RL, deFazio A: Expression of progesterone receptors A and B in the mouse ovary during the estrous cycle. Endocrinology 2004, 145:3487-3494

Grasso P, Reichert LE, Jr: In vivo effects of follicle-stimulating hormone-related synthetic peptides on the mouse estrous cycle. Endocrinology 1996, 137:5370-5375

Robertson SA, Mayrhofer G, Seamark RF: Ovarian steroid hormones regulate granulocyte-macrophage colony-stimulating factor synthesis by uterine epithelial cells in the mouse. Biol Reprod 1996, 54:183-196

Dean J, Cohen G, Kemp J, Robson L, Tembe V, Hasselaar J, Webster B, Lammi A, Smith A: Karyotype 69, XXX/47,XX,+15 in a 2 1/2 year old child. J Med Genet 1997, 34:246-249

Roberts HE, Saxe DF, Muralidharan K, Coleman KB, Zacharias JF, Fernhoff PM: Unique mosaicism of tetraploidy and trisomy 8: clinical, cytogenetic, and molecular findings in a live-born infant. Am J Med Genet 1996, 62:243-246

Isaksson R, Tiitinen A: Present concept of unexplained infertility. Gynecol Endocrinol 2004, 18:278-290


作者单位:From the Department of Biomedical Science,* Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan; the Department of Degenerative Neurological Disease and the Section of Laboratory Animal Resources, National Institute of Neuroscience, National Center of Neurology and P

作者: Satoshi Sekiguchi, Jungkee Kwon, Etsuko Yoshida, H 2008-5-29
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具