Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 169
Filtrar
1.
Nat Med ; 1(1): 47-52, 1995 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7584952

RESUMO

Successful fertilization in humans, achieved when parental chromosomes intermix at first mitosis, requires centrosome restoration and microtubule-mediated motility. Imaging of inseminated human oocytes reveals that the sperm introduces the centrosome. The centrosome then nucleates the new microtubule assembly to form the sperm aster--a step essential for successful fertilization. Oocytes from some infertile patients failed to complete fertilization because of defects in uniting the sperm and egg nuclei, indicating that failure to properly effect the cytoplasmic motions uniting the nuclei results in human infertility. These discoveries have important implications for infertility diagnosis and managing reproduction.


Assuntos
Centrossomo , Fertilização , Infertilidade Masculina/patologia , Microtúbulos/fisiologia , Fuso Acromático/ultraestrutura , Fertilização in vitro , Humanos , Masculino , Microscopia de Fluorescência
2.
Nat Med ; 5(4): 431-3, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10202934

RESUMO

Intracytoplasmic sperm injection has begun an era of considerable improvements in treating male infertility. Despite its success, questions remain about the dangers of transmitting traits responsible for male infertility, sex and autosomal chromosome aberrations and possible mental, physical and reproductive abnormalities. We report here the first births of rhesus monkeys produced by intracytoplasmic sperm injection at rates greater or equal to those reported by clinics. Essential assumptions about this process are flawed, as shown by results with the preclinical, nonhuman primate model and with clinically discarded specimens. Dynamic imaging demonstrated the variable position of the second meiotic spindle in relation to the first polar body; consequently, microinjection targeting is imprecise and potentially lethal. Intracytoplasmic sperm injection resulted in abnormal sperm decondensation, with the unusual retention of vesicle-associated membrane protein and the perinuclear theca, and the exclusion of the nuclear mitotic apparatus from the decondensing sperm nuclear apex. Male pronuclear remodeling in the injected oocytes was required before replication of either parental genome, indicating a unique G1-to-S transition checkpoint during zygotic interphase (the first cell cycle). These irregularities indicate that the intracytoplasmic sperm injection itself might lead to the observed increased chromosome anomalies.


Assuntos
Fertilização in vitro/efeitos adversos , Fertilização/fisiologia , Zigoto/citologia , Animais , Ciclo Celular , Núcleo Celular , Feminino , Infertilidade Masculina/terapia , Macaca mulatta , Masculino , Microinjeções , Oócitos/fisiologia , Interações Espermatozoide-Óvulo , Espermatozoides/patologia
3.
J Cell Biol ; 77(2): 517-35, 1978 May.
Artigo em Inglês | MEDLINE | ID: mdl-649659

RESUMO

The nuclear envelope and associated structures from Xenopus laevis oocytes (stage VI) have been examined with the high resolution scanning electron microscope (SEM). The features of the inner and outer surfaces of the nuclear surface complex were revealed by manual isolation , whereas the membranes facing the perinuclear space (the space between the inner and outer nuclear membranes) were observed by fracturing the nuclear envelope in this plane and splaying the corresponding regions apart. Pore complexes were observed on all four membrane surfaces of this double-membraned structure. The densely packed pore complexes (55/micron2) are often clustered into triplets with shared walls (outer diameter = 90 nm; inner diameter = 25 nm; wall thickness = aproximately 30 nm), and project aproximately 20 nm above each membrane except where they are flush with the innermost surface. The pore complex appears to be an aggregate of four 30-nm subunits. The nuclear cortex, a fibrous layer (300 nm thickness) associated with the inner surface of the nuclear envelope, has been revealed by rapid fixation. This cortical layer is interrupted by funnel-shaped intranuclear channels (120-640 nm diam) which narrow towards the pore complexes. Chains of particles, arranged in spirals, are inserted into these intranuclear channels. The fibers associated with the innermost face of the nuclear envelope can be extraced with 0.6 MKI to reveal the pore complexes. A model of the nuclear surface complex, compiled from the visualization of all the membrane faces and the nuclear cortex, demonstrates relations between the intranuclear channels (3.2/micron2) and the numerous pore complexes, and the possibility of their role in nucleocytoplasmic interactions.


Assuntos
Núcleo Celular/ultraestrutura , Membrana Nuclear/ultraestrutura , Oócitos/ultraestrutura , Óvulo/ultraestrutura , Animais , Feminino , Microscopia Eletrônica de Varredura , Modelos Estruturais , Xenopus
4.
J Cell Biol ; 66(1): 198-200, 1975 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-1095595

RESUMO

Cells of many kinds adhere firmly to glass or plastic surfaces which have been pretreated with polylysine. The attachment takes place as soon as the cells make contact with the surfaces, and the flattening of the cells against the surfaces is quite rapid. Cells which do not normally adhere to solid surfaces, such as sea urchin eggs, attach as well as cells which normally do so, such as amebas or mammalian cells in culture. The adhesion is interpreted simply as the interaction between the polyanionic cell surfaces and the polycationic layer of adsorbed polylysine. The attachment of cells to the polylysine-treated surfaces can be exploited for a variety of experimental manipulations. In the preparation of samples for scanning or transmission electron microscopy, the living material may first be attached to a polylysine-coated plate or grid, subjected to some experimental treatment (fertilization of an egg, for example), then transferred rapidly to fixative and further passed through processing for observation; each step involves only the transfer of the plate or grid from one container to the next. The cells are not detached. The adhesion of the cell may be so firm that the body of the cell may be sheared away, leaving attached a patch of cell surface, face up, for observation of its inner aspect. For example, one may observe secretory vesicles on the inner face of the surface (3) or may study the association of filaments with the inner surface (Fig. 1). Subcellular structures may attach to the polylysine-coated surfaces. So far, we have found this to be the case for nuclei isolated from sea urchin embryos and for the microtubules of flagella, which are well displayed after the membrane has been disrupted by Triton X-100 (Fig. 2).


Assuntos
Adesão Celular , Técnicas Histológicas , Microscopia Eletrônica , Peptídeos , Polilisina , Animais , Dictyostelium/ultraestrutura , Feminino , Vidro , Masculino , Microscopia Eletrônica de Varredura , Óvulo/ultraestrutura , Ouriços-do-Mar/ultraestrutura , Cauda do Espermatozoide/ultraestrutura , Tetrahymena/ultraestrutura
5.
J Cell Biol ; 117(4): 799-811, 1992 May.
Artigo em Inglês | MEDLINE | ID: mdl-1577859

RESUMO

During early development, intracellular Ca2+ mobilization is not only essential for fertilization, but has also been implicated during other meiotic and mitotic events, such as germinal vesicle breakdown (GVBD) and nuclear envelope breakdown (NEBD). In this study, the roles of intracellular and extracellular Ca2+ were examined during meiotic maturation and reinitiation at parthenogenetic activation and during first mitosis in a single species using the same methodologies. Cumulus-free metaphase II mouse oocytes immediately resumed anaphase upon the induction of a large, transient Ca2+ elevation. This resumption of meiosis and associated events, such as cortical granule discharge, were not sensitive to extracellular Ca2+ removal, but were blocked by intracellular Ca2+ chelators. In contrast, meiosis I was dependent on external Ca2+; in its absence, the formation and function of the first meiotic spindle was delayed, the first polar body did not form and an interphase-like state was induced. GVBD was not dependent on external Ca2+ and showed no associated Ca2+ changes. NEBD at first mitosis in fertilized eggs, on the other hand, was frequently, but not always associated with a brief Ca2+ transient and was dependent on Ca2+ mobilization. We conclude that GVBD is Ca2+ independent, but that the dependence of NEBD on Ca2+ suggests regulation by more than one pathway. As cells develop from Ca(2+)-independent germinal vesicle oocytes to internal Ca(2+)-dependent pronuclear eggs, internal Ca2+ pools increase by approximately fourfold.


Assuntos
Cálcio/fisiologia , Meiose , Membrana Nuclear/fisiologia , Oócitos/fisiologia , Animais , Compartimento Celular , Cromatina/ultraestrutura , Camundongos , Microtúbulos/ultraestrutura , Partenogênese
6.
J Cell Biol ; 94(2): 455-65, 1982 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-6125518

RESUMO

Taxol blocks the migrations of the sperm and egg nuclei in fertilized eggs and induces asters in unfertilized eggs of the sea urchins Lytechinus variegatus and Arbacia punctulata. Video recordings of eggs inseminated in 10 microM taxol demonstrate that sperm incorporation and sperm tail motility are unaffected, that the sperm aster formed is unusually pronounced, and that the migration of the egg nucleus and pronuclear centration are inhibited. The huge monopolar aster persists for at least 6 h; cleavage attempts and nuclear cycles are observed. Colcemid (10 microM) disassembles both the large taxol-stabilized sperm aster in fertilized eggs and the numerous asters induced in unfertilized eggs. Antitubulin immunofluorescence microscopy demonstrates that in fertilized eggs all microtubules are within the prominent sperm aster. Within 15 min of treatment with 10 microM taxol, unfertilized eggs develop numerous (greater than 25) asters de novo. Transmission electron microscopy of unfertilized eggs reveals the presence of microtubule bundles that do not emanate from centrioles but rather from osmiophilic foci or, at times, the nuclear envelope. Taxol-treated eggs are not activated as judged by the lack of DNA synthesis, nuclear or chromosome cycles, and the cortical reaction. These results indicate that: (a) taxol prevents the normal cycles of microtubule assembly and disassembly observed during development; (b) microtubule disassembly is required for the nuclear movements during fertilization; (c) taxol induces microtubules in unfertilized eggs; and (d) nucleation centers other than centrioles and kinetochores exist within unfertilized eggs; these presumptive microtubule organizing centers appear idle in the presence of the sperm centrioles.


Assuntos
Alcaloides/farmacologia , Fertilização/efeitos dos fármacos , Microtúbulos/fisiologia , Ouriços-do-Mar/efeitos dos fármacos , Animais , Núcleo Celular/fisiologia , Demecolcina/farmacologia , Feminino , Masculino , Microtúbulos/efeitos dos fármacos , Movimento/efeitos dos fármacos , Óvulo/efeitos dos fármacos , Óvulo/ultraestrutura , Paclitaxel , Motilidade dos Espermatozoides/efeitos dos fármacos , Cauda do Espermatozoide/efeitos dos fármacos , Tubulina (Proteína)/metabolismo
7.
J Cell Biol ; 111(4): 1491-504, 1990 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2211822

RESUMO

Kinetochores may perform several functions at mitosis and meiosis including: (a) directing anaphase chromosome separation, (b) regulating prometaphase alignment of the chromosomes at the spindle equator (congression), and/or (c) capturing and stabilizing microtubules. To explore these functions in vivo, autoimmune sera against the centromere/kinetochore complex are microinjected into mouse oocytes during specific phases of first or second meiosis, or first mitosis. Serum E.K. crossreacts with an 80-kD protein in mouse cells and detects the centromere/kinetochore complex in permeabilized cells or when microinjected into living oocytes. Chromosome separation at anaphase is not blocked when these antibodies are microinjected into unfertilized oocytes naturally arrested at second meiotic metaphase, into eggs at first mitotic metaphase, or into immature oocytes at first meiotic metaphase. Microtubule capture and spindle reformation occur normally in microinjected unfertilized oocytes recovering from cold or microtubule disrupting drugs; the chromosomes segregate correctly after parthenogenetic activation. Prometaphase congression is dramatically influenced when antikinetochore/centromere antibodies are introduced during interphase or in prometaphase-stage meiotic or mitotic eggs. At metaphase, these oocytes have unaligned chromosomes scattered throughout the spindle with several remaining at the poles; anaphase is aberrant and, after division, karyomeres are found in the polar body and oocyte or daughter blastomeres. Neither nonimmune sera, diffuse scleroderma sera, nor sham microinjections affect either meiosis or mitosis. These results suggest that antikinetochore/centromere antibodies produced by CREST patients interfere with chromosome congression at prometaphase in vivo.


Assuntos
Centrômero/fisiologia , Cromossomos/fisiologia , Meiose/fisiologia , Mitose/fisiologia , Anáfase/fisiologia , Animais , Anticorpos , Autoanticorpos , Centrômero/imunologia , Reações Cruzadas , Humanos , Immunoblotting , Metáfase/fisiologia , Camundongos , Microinjeções , Microtúbulos/metabolismo , Mitose/efeitos dos fármacos , Peso Molecular , Nocodazol/farmacologia , Oócitos/citologia , Escleroderma Sistêmico/imunologia , Fuso Acromático/metabolismo
8.
J Cell Biol ; 147(2): 351-66, 1999 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-10525540

RESUMO

We have prepared antibodies specific for HSET, the human homologue of the KAR3 family of minus end-directed motors. Immuno-EM with these antibodies indicates that HSET frequently localizes between microtubules within the mammalian metaphase spindle consistent with a microtubule cross-linking function. Microinjection experiments show that HSET activity is essential for meiotic spindle organization in murine oocytes and taxol-induced aster assembly in cultured cells. However, inhibition of HSET did not affect mitotic spindle architecture or function in cultured cells, indicating that centrosomes mask the role of HSET during mitosis. We also show that (acentrosomal) microtubule asters fail to assemble in vitro without HSET activity, but simultaneous inhibition of HSET and Eg5, a plus end-directed motor, redresses the balance of forces acting on microtubules and restores aster organization. In vivo, centrosomes fail to separate and monopolar spindles assemble without Eg5 activity. Simultaneous inhibition of HSET and Eg5 restores centrosome separation and, in some cases, bipolar spindle formation. Thus, through microtubule cross-linking and oppositely oriented motor activity, HSET and Eg5 participate in spindle assembly and promote spindle bipolarity, although the activity of HSET is not essential for spindle assembly and function in cultured cells because of centrosomes.


Assuntos
Proteínas Fúngicas/fisiologia , Cinesinas/fisiologia , Microtúbulos/fisiologia , Mitose/fisiologia , Proteínas de Saccharomyces cerevisiae , Proteínas de Xenopus , Células HeLa , Humanos , Proteínas Associadas aos Microtúbulos/fisiologia , Proteínas Motores Moleculares
9.
J Cell Biol ; 104(5): 1133-42, 1987 May.
Artigo em Inglês | MEDLINE | ID: mdl-3553205

RESUMO

We have used in situ hybridization and cell fractionation methods to follow the distribution of U1 RNA and immunofluorescence microscopy to follow the distribution of snRNP proteins in oocytes, eggs, and embryos of several sea urchin species. U1 RNA and U1-specific snRNP antigens are concentrated in germinal vesicles of oocytes. Both appear to relocate after oocyte maturation because they are found primarily, if not exclusively, in the cytoplasm of mature unfertilized eggs. This cytoplasmic residence is maintained during early cleavage and U1 RNA is first detectable in nuclei of micromeres at the 16-cell stage. Between morula and gastrula stages the steady-state concentrations of both RNA and antigens gradually increase in nuclei and decrease in cytoplasm. Surprisingly, analysis of the distribution of newly synthesized U1 RNA shows that it does not equilibrate with the maternal pool. Instead new transcripts are confined to nuclei, while cytoplasmic U1 RNAs are of maternal origin. This lack of equilibration and the conversion of maternal U1 RNAs from nuclear species in oocytes to cytoplasmic in embryos suggests that these RNPs (or RNAs) are structurally altered when released to the cytoplasm at oocyte maturation.


Assuntos
Embrião não Mamífero/citologia , RNA Nuclear Pequeno/genética , Ouriços-do-Mar/embriologia , Animais , Divisão Celular , Núcleo Celular/ultraestrutura , Feminino , Imunofluorescência , Hibridização de Ácido Nucleico , RNA Nuclear Pequeno/análise , Ouriços-do-Mar/citologia , Transcrição Gênica
10.
Science ; 291(5502): 309-12, 2001 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-11209082

RESUMO

Transgenic rhesus monkeys carrying the green fluorescent protein (GFP) gene were produced by injecting pseudotyped replication-defective retroviral vector into the perivitelline space of 224 mature rhesus oocytes, later fertilized by intracytoplasmic sperm injection. Of the three males born from 20 embryo transfers, one was transgenic when accessible tissues were assayed for transgene DNA and messenger RNA. All tissues that were studied from a fraternal set of twins, miscarried at 73 days, carried the transgene, as confirmed by Southern analyses, and the GFP transgene reporter was detected by both direct and indirect fluorescence imaging.


Assuntos
Animais Geneticamente Modificados , Técnicas de Transferência de Genes , Transferência Genética Horizontal , Proteínas Luminescentes/genética , Macaca mulatta/genética , Animais , Animais Recém-Nascidos , Southern Blotting , Transferência Embrionária , Desenvolvimento Embrionário e Fetal , Feminino , Imunofluorescência , Expressão Gênica , Vetores Genéticos , Proteínas de Fluorescência Verde , Masculino , Vírus da Leucemia Murina de Moloney/genética , Oócitos , Reação em Cadeia da Polimerase , Gravidez , Resultado da Gravidez , Injeções de Esperma Intracitoplásmicas , Transgenes
11.
Science ; 287(5451): 317-9, 2000 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-10634789

RESUMO

Primates that are identical in both nuclear and cytoplasmic components have not been produced by current cloning strategies, yet such identicals represent the ideal model for investigations of human diseases. Here, genetically identical nonhuman embryos were produced as twin and larger sets by separation and reaggregation of blastomeres of cleavage-stage embryos. A total of 368 multiples were created by the splitting of 107 rhesus embryos with four pregnancies established after 13 embryo transfers (31% versus 53% in vitro fertilization controls). The birth of Tetra, a healthy female cloned from a quarter of an embryo, proves that this approach can result in live offspring.


Assuntos
Blastômeros/fisiologia , Fase de Clivagem do Zigoto/fisiologia , Clonagem de Organismos/métodos , Desenvolvimento Embrionário e Fetal , Macaca mulatta/embriologia , Animais , Apoptose , Blastocisto/fisiologia , Transferência Embrionária , Feminino , Gravidez , Gêmeos Monozigóticos , Zona Pelúcida/fisiologia
12.
Mol Biol Cell ; 9(9): 2509-25, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9725909

RESUMO

To explore the role of nonmuscle myosin II isoforms during mouse gametogenesis, fertilization, and early development, localization and microinjection studies were performed using monospecific antibodies to myosin IIA and IIB isotypes. Each myosin II antibody recognizes a 205-kDa protein in oocytes, but not mature sperm. Myosin IIA and IIB demonstrate differential expression during meiotic maturation and following fertilization: only the IIA isoform detects metaphase spindles or accumulates in the mitotic cleavage furrow. In the unfertilized oocyte, both myosin isoforms are polarized in the cortex directly overlying the metaphase-arrested second meiotic spindle. Cortical polarization is altered after spindle disassembly with Colcemid: the scattered meiotic chromosomes initiate myosin IIA and microfilament assemble in the vicinity of each chromosome mass. During sperm incorporation, both myosin II isotypes concentrate in the second polar body cleavage furrow and the sperm incorporation cone. In functional experiments, the microinjection of myosin IIA antibody disrupts meiotic maturation to metaphase II arrest, probably through depletion of spindle-associated myosin IIA protein and antibody binding to chromosome surfaces. Conversely, the microinjection of myosin IIB antibody blocks microfilament-directed chromosome scattering in Colcemid-treated mature oocytes, suggesting a role in mediating chromosome-cortical actomyosin interactions. Neither myosin II antibody, alone or coinjected, blocks second polar body formation, in vitro fertilization, or cytokinesis. Finally, microinjection of a nonphosphorylatable 20-kDa regulatory myosin light chain specifically blocks sperm incorporation cone disassembly and impedes cell cycle progression, suggesting that interference with myosin II phosphorylation influences fertilization. Thus, conventional myosins break cortical symmetry in oocytes by participating in eccentric meiotic spindle positioning, sperm incorporation cone dynamics, and cytokinesis. Although murine sperm do not express myosin II, different myosin II isotypes may have distinct roles during early embryonic development.


Assuntos
Fertilização/fisiologia , Meiose/fisiologia , Mitose/fisiologia , Miosinas/metabolismo , Animais , Afinidade de Anticorpos , Células COS , Divisão Celular , Demecolcina/farmacologia , Desenvolvimento Embrionário e Fetal , Feminino , Regulação da Expressão Gênica , Isoenzimas/metabolismo , Masculino , Camundongos , Microinjeções , Cadeias Pesadas de Miosina/metabolismo , Cadeias Leves de Miosina/metabolismo , Miosinas/genética , Oócitos , Fosforilação , Isoformas de Proteínas , Interações Espermatozoide-Óvulo , Espermatozoides/metabolismo , Espermatozoides/fisiologia
13.
Mol Biol Cell ; 10(9): 2955-69, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10473639

RESUMO

Human sperm centrosome reconstitution and the parental contributions to the zygotic centrosome are examined in mammalian zygotes and after exposure of spermatozoa to Xenopus laevis cell-free extracts. The presence and inheritance of the conserved centrosomal constituents gamma-tubulin, centrin, and MPM-2 (which detects phosphorylated epitopes) are traced, as is the sperm microtubule-nucleating capability on reconstituted centrosomes. gamma-Tubulin is biparentally inherited in humans (maternal >> than paternal): Western blots detect the presence of paternal gamma-tubulin. Recruitment of maternal gamma-tubulin to the sperm centrosome occurs after sperm incorporation in vivo or exposure to cell-free extract, especially after sperm "priming" induced by disulfide bond reduction. Centrin is found in the proximal sperm centrosomal region, demonstrates expected calcium sensitivity, but appears absent from the zygotic centrosome after sperm incorporation or exposure to extracts. Sperm centrosome phosphorylation is detected after exposure of primed sperm to egg extracts as well as during the early stages of sperm incorporation after fertilization. Finally, centrosome reconstitution in cell-free extracts permits sperm aster microtubule assembly in vitro. Collectively, these results support a model of a blended zygotic centrosome composed of maternal constituents attracted to an introduced paternal template after insemination.


Assuntos
Proteínas de Ciclo Celular , Centrossomo/metabolismo , Herança Extracromossômica , Fertilização/genética , Tubulina (Proteína)/metabolismo , Zigoto/citologia , Animais , Cálcio/metabolismo , Bovinos , Extratos Celulares , Centrossomo/química , Centrossomo/ultraestrutura , Citoplasma/metabolismo , Dissulfetos/química , Dissulfetos/metabolismo , Feminino , Humanos , Cinesinas , Masculino , Microtúbulos/metabolismo , Oócitos/química , Oócitos/citologia , Oócitos/metabolismo , Oócitos/ultraestrutura , Pais , Fosfoproteínas/análise , Fosforilação , Espermatozoides/química , Espermatozoides/citologia , Espermatozoides/metabolismo , Espermatozoides/ultraestrutura , Fuso Acromático/metabolismo , Combinação Trimetoprima e Sulfametoxazol/análise , Tubulina (Proteína)/genética , Xenopus laevis , Zigoto/química , Zigoto/metabolismo , Zigoto/ultraestrutura
14.
Sci Rep ; 6: 31816, 2016 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-27534805

RESUMO

The fertilizing sperm's lengthiest unchartered voyage is through the longest, least-investigated organ in a man's body - the Epididymis. Over six meters long in men, ~80 meters in stallions and over one-hundred times a mouse's body length, there are few functions known aside from sperm storage and nutrition. While spermatogenesis is completed in the testes, here we demonstrate sperm centriole reduction occurs within the epididymis. Investigations of GFP-CENTR mice and controls demonstrate both the presence of centriole pairs in the upper caput region of the epididymis and, the destruction, first, of the distal and, then, of the proximal centriole as the sperm transits to the cauda and vas deferens in preparation for its climactic release. These centrioles can neither recruit γ-tubulin nor nucleate microtubules when eggs are inseminated or microinjected, yet numerous maternally-nucleated cytasters are found. These sperm centrioles appear as vestigial basal bodies, destroyed in the mid-to-lower corpus. Post-testicular sperm maturation, in which sperm centrioles found in the caput are destroyed prior to ejaculation, is a newly discovered function for the epididymis.


Assuntos
Centríolos/metabolismo , Ejaculação/fisiologia , Maturação do Esperma/fisiologia , Espermatozoides/metabolismo , Animais , Centríolos/genética , Epididimo/metabolismo , Masculino , Camundongos , Camundongos Transgênicos
15.
Int Rev Cytol ; 195: 1-65, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10603574

RESUMO

Mammalian fertilization has traditionally been regarded as a simple blending of two gametes, during which the haploid genome of the fertilizing spermatozoon constitutes the primary paternal contribution to the resulting embryo. In contrast to this view, new research provides evidence of important cytoplasmic contributions made by the fertilizing spermatozoon to the zygotic makeup, to the organization of preimplantation development, and even reproductive success of new forms of assisted fertilization. The central role of the sperm-contributed centriole in the reconstitution of zygotic centrosome has been established in most mammalian species and is put in contrast with strictly maternal centrosomal inheritance in rodents. The complementary reduction or multiplication of sperm and oocyte organelles during gametogenesis, exemplified by the differences in the biogenesis of centrosome in sperm and oocytes, represents an intriguing mechanism for avoiding their redundancy during early embryogenesis. New studies on perinuclear theca of sperm revealed its importance for both spermatogenesis and fertilization. Remodeling of the sperm chromatin into a male pronucleus is guided by oocyte-produced, reducing peptide glutathione and a number of molecules required for the reconstitution of the functional nuclear envelope and nuclear skeleton. Although some of the sperm structures are transformed into zygotic components, the elimination of others is vital to early stages of embryonic development. Sperm mitochondria, carrying potentially harmful paternal mtDNA, appear to be eliminated by a ubiquitin-dependent mechanism. Other accessory structures of the sperm axoneme, including fibrous sheath, microtubule doublets, outer dense fibers, and the striated columns of connecting piece, are discarded in an orderly fashion. The new methods of assisted fertilization, represented by intracytoplasmic sperm injection and round spermatid injection, bypass multiple steps of natural fertilization by introducing an intact spermatozoon or spermatogenic cell into oocyte cytoplasm. Consequently, the carryover of sperm accessory structures that would normally be eliminated before or during the entry of sperm into oocyte cytoplasm persist therein and may interfere with early embryonic development, thus decreasing the success rate of assisted fertilization and possibly causing severe embryonic anomalies. Similarly, foreign organelles, proteins, messenger RNAs, and mitochondrial DNAs, which may have a profound impact on the embryonic development, are propagated by the nuclear transfer of embryonic blastomeres and somatic cell nuclei. This aspect of assisted fertilization is yet to be explored by a focused effort.


Assuntos
Interações Espermatozoide-Óvulo , Espermatozoides/citologia , Membrana Celular/metabolismo , Núcleo Celular/fisiologia , Centrossomo , Feminino , Humanos , Masculino , Mitocôndrias/genética , Mitocôndrias/fisiologia , Transdução de Sinais , Espermatozoides/ultraestrutura
16.
Mech Dev ; 35(2): 103-11, 1991 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-1760374

RESUMO

During the successive interphases of cleaving mouse embryos the nuclear periphery diminishes its reactivity to anti-lamin A and C antibodies. This developmentally regulated characteristic can be modified by exposure of the blastomere nuclei to metaphase II (M II) oocyte cytoplasm followed by activation. In the current study we define the cytoplasmic conditions necessary for this modification of 8-cell and 16-cell stage nuclei in hybrids obtained by fusion with metaphase II arrested oocytes, oocytes at various time points after parthenogenetic activation, naturally fertilized eggs (zygotes) and interphase 2-cell embryo blastomeres. The intensity of fluorescence obtained with anti-lamins A/C in the blastomere nuclei increases as a result of fusion with freshly activated oocytes or early zygotes (first 3.0-5.5 h in the case of parthenogenetic activation), and not when eggs or 2-cell blastomeres advanced in interphase are used as partners for fusion. This transformation of the A/C lamin pattern is correlated with the ability to promote pronucleus-like growth of blastomere nuclei in hybrids. Blastomere nuclei introduced into M II-arrested oocytes undergo premature chromatin condensation and dissolution of the nuclear lamina. The results are discussed with regard to certain particularities of the first embryonic interphase of the mouse and the potential involvement of nuclear lamins in pronuclear growth.


Assuntos
Blastômeros/ultraestrutura , Núcleo Celular/ultraestrutura , Laminina/metabolismo , Animais , Ciclo Celular , Fusão Celular , Feminino , Células Híbridas/ultraestrutura , Camundongos , Camundongos Endogâmicos ICR/embriologia , Oócitos/metabolismo , Partenogênese , Gravidez , Zigoto/metabolismo
17.
Eur J Cell Biol ; 27(1): 74-87, 1982 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7084254

RESUMO

Griseofulvin (4-6 X10-5 and 1 X 10-4 M) prevents the formation of any microtubule-based structures of sea urchin (Strongylocentrotus purpuratus, Lytechinus variegatus, Arbacia punctulata) eggs at fertilization. Sperm incorporation occurs, though the migrations of the pronuclei, dependent on the formation of the sperm aster, are arrested. Similarly in "streak" and the mitotic apparatus fail to assemble. Cycles of DNA synthesis, chromosome activity, nuclear breakdown and reconstitution, and even cleavage attempts occur on schedule in the absence of any mitotic movements. The action of griseofulvin, unlike that of colchicine, is readily reversible by the removal of the drug. Microtubules are formed, and the chromosome are separated. At 1 X 10-6 M, diminutive microtubule-based structures (e.g. sperm aster, mitotic apparatus) are observed though syngamy and division are arrested. These results demonstrate an independence of the cycle of microtubule-mediated events from other cyclical processes during the first cell cycles.


Assuntos
Fertilização/efeitos dos fármacos , Griseofulvina/farmacologia , Microtúbulos/efeitos dos fármacos , Mitose/efeitos dos fármacos , Ouriços-do-Mar/embriologia , Animais , Ciclo Celular/efeitos dos fármacos , Cromossomos/metabolismo , DNA/biossíntese
18.
Eur J Cell Biol ; 36(1): 116-27, 1985 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-4038941

RESUMO

The regulation of the microtubule-mediated motions within eggs during fertilization was investigated in relation to the shift in intracellular pH (pHi) that occurs during the ionic sequence of egg activation in the sea urchins Lytechinus variegatus and Arbacia punctulata. Microtubule assembly during formation of the sperm aster and mitotic apparatus was detected by anti-tubulin immunofluorescence microscopy, and the microtubule-mediated migrations of the sperm and egg nuclei were studied with time-lapse video differential interference contrast microscopy. Manipulations of intracellular pH were verified by fluorimetric analyses of cytoplasmic fluorescein incorporated as fluorescein diacetate. The ionic sequence of egg activation was manipulated i) to block the pHi shift at fertilization or reduce the pHi of fertilized eggs to unfertilized values, ii) to elevate artificially the pHi of unfertilized eggs to fertilized values, and iii) to elevate artificially or permit the normal pHi shift in fertilized eggs in which the pHi shift at fertilization was previously prevented. Fertilized eggs in which the pHi shift was suppressed did not assemble microtubules or undergo the normal microtubule-mediated motions. In fertilized eggs in which the pHi was reduced to unfertilized levels after the assembly of the sperm aster, no motions were detected. If the intracellular pH was later permitted to rise, normal motile events leading to division and development occurred, delayed by the time during which the pH elevation was blocked. Microtubule-mediated events occurred in eggs in which the intracellular pH was elevated, even in unfertilized eggs in which the pH was artificially increased. These results indicate that the formation and normal functioning of the egg microtubules is initiated, either directly or indirectly, by the shift in intracellular pH that occurs during fertilization.


Assuntos
Fertilização , Concentração de Íons de Hidrogênio , Microtúbulos/ultraestrutura , Óvulo/ultraestrutura , Zigoto/ultraestrutura , Animais , Feminino , Microtúbulos/fisiologia , Óvulo/fisiologia , Ouriços-do-Mar , Tubulina (Proteína)/fisiologia , Zigoto/fisiologia
19.
Cloning Stem Cells ; 3(3): 157-61, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11945225

RESUMO

Ubiquitination is a universal protein degradation pathway in which the molecules of 8.5-kDa proteolytic peptide ubiquitin are covalently attached to the epsilon-amino group of the substrate's lysine residues. Little is known about the importance of this highly conserved mechanism for protein recycling in mammalian gametogenesis and fertilization. The data obtained by the students and faculty of the international training course Window to the Zygote 2000 demonstrate the accumulation of ubiquitin-cross-reactive structures in the trophoblast, but not in the inner cell mass of the expanding bovine and mouse blastocysts. This observation suggests that a major burst of ubiquitin-dependent proteolysis occurs in the trophoblast of mammalian peri-implantation embryos. This event may be important for the success of blastocyst hatching, differentiation of embryonic stem cells into soma and germ line, and/or implantation in both naturally conceived and reconstructed mammalian embryos.


Assuntos
Mamíferos/embriologia , Trofoblastos/metabolismo , Ubiquitina/metabolismo , Animais , Biomarcadores/análise , Blastocisto/metabolismo , Bovinos , Células Cultivadas , Camundongos , Camundongos Endogâmicos ICR
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA