Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cryobiology ; 114: 104841, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38104854

RESUMO

The cryopreservation of immature testicular tissue (ITT) prior to gonadotoxic therapy is crucial for fertility preservation in prepubertal boys with cancer. However, the optimal holding time between tissue collection and cryopreservation has yet to be elucidated. Using the bovine model, we investigated four holding times (1, 6, 24, and 48 h) for ITTs before cryopreservation. Biopsies from two-week-old calves were stored in transport medium and cryopreserved following a standard slow-freezing clinical protocol. Thawed samples were then assessed for viability, morphology, and gene expression by haematoxylin and eosin (H&E) staining, immunohistochemistry and real-time quantitative reverse transcription-polymerase chain reaction (RT-qPCR). Analysis failed to identify any significant changes in cell viability when compared between the different groups. Sertoli (Vimentin+) and proliferating cells (Ki67+) were well-preserved. The expression of genes related to germ cells, spermatogenesis (STRA8, PLZF, GFRα-1, C-KIT, THY1, UCHL-1, NANOG, OCT-4, CREM), and apoptosis (HSP70-2) remained stable over 48 h. However, seminiferous cord detachment increased significantly in the 48-h group (p < 0.05), with associated cord and SSC shrinkage. Collectively, our analyses indicate that bovine ITTs can be stored for up to 48 h prior to cryopreservation with no impact on cell viability and the expression levels of key genes. However, to preserve the morphology of frozen-thawed tissue, the ideal processing time would be within 24 h. Testicular tissues obtained from patients for fertility preservation often need to be transported over long distances to be cryopreserved in specialist centres. Our findings highlight the importance of determining optimal tissue transport times to ensure tissue quality in cryopreservation.


Assuntos
Preservação da Fertilidade , Testículo , Masculino , Humanos , Animais , Bovinos , Criopreservação/métodos , Espermatogônias/metabolismo , Espermatogênese , Preservação da Fertilidade/métodos
2.
Development ; 147(19)2020 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-32928909

RESUMO

Precise patterning within the three-dimensional context of tissues, organs and embryos implies that cells can sense their relative position. During preimplantation development, outside and inside cells rely on apicobasal polarity and the Hippo pathway to choose their fate. Despite recent findings suggesting that mechanosensing might be central to this process, the relationship between blastomere geometry (i.e. shape and position) and the Hippo pathway effector YAP remains unknown. We used a highly quantitative approach to analyse information on the geometry and YAP localisation of individual blastomeres of mouse and human embryos. We identified the proportion of exposed cell surface area as most closely correlating with the nuclear localisation of YAP. To test this relationship, we developed several hydrogel-based approaches to alter blastomere geometry in cultured embryos. Unbiased clustering analyses of blastomeres from such embryos revealed that this relationship emerged during compaction. Our results therefore pinpoint the time during early embryogenesis when cells acquire the ability to sense changes in geometry and provide a new framework for how cells might integrate signals from different membrane domains to assess their relative position within the embryo.


Assuntos
Blastocisto/citologia , Blastocisto/metabolismo , Blastômeros/metabolismo , Animais , Blastômeros/citologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Feminino , Humanos , Camundongos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
3.
Reprod Biomed Online ; 38(5): 725-739, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30922556

RESUMO

To improve success rates, assisted reproductive technology (ART) procedures continually undergo optimization and enhancement such that the best quality gametes and embryos can be identified and manipulated, thus improving clinical outcomes. Laser technology is now being applied across ART to reduce procedure times and increase the consistency and reproducibility of traditional ART techniques such as assisted hatching, embryo biopsy, intracytoplasmic sperm injection cryopreservation and sperm immobilization/selection. This review examines the current status of cutting-edge laser-assisted reproductive technologies, investigates experimental techniques that are increasingly being applied clinically. It highlights the benefits of lasers as a powerful technology at the forefront of both diagnostic and therapeutic treatments for general subfertility and male-factor infertility. However, it is important to note that although lasers are becoming increasingly commonplace in ART units, there is comparatively little information in the existing literature pertaining to the potential negative effects that laser application might have on the developing human embryo, thus creating the need for further investigative research.


Assuntos
Lasers , Técnicas de Reprodução Assistida , Animais , Blastocisto , Criopreservação , Humanos
4.
Birth Defects Res C Embryo Today ; 108(1): 19-32, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26969610

RESUMO

Successful pregnancy is dependent upon the implantation of a competent embryo into a receptive endometrium. Despite major advancement in our understanding of reproductive medicine over the last few decades, implantation failure still occurs in both normal pregnancies and those created artificially by assisted reproductive technology (ART). Consequently, there is significant interest in elucidating the etiology of implantation failure. The complex multistep process of implantation begins when the developing embryo first makes contact with the plasma membrane of epithelial cells within the uterine environment. However, although this biological interaction marks the beginning of a fundamental developmental process, our knowledge of the intricate physiological and molecular processes involved remains sparse. In this synopsis, we aim to provide an overview of our current understanding of the morphological changes which occur to the plasma membrane of the uterine endothelium, and the molecular mechanisms that control communication between the early embryo and the endometrium during implantation. A multitude of molecular factors have been implicated in this complex process, including endometrial integrins, extracellular matrix molecules, adhesion molecules, growth factors, and ion channels. We also explore the development of in vitro models for embryo implantation to help researchers investigate mechanisms which may underlie implantation failure. Understanding the precise molecular pathways associated with implantation failure could help us to generate new prognostic/diagnostic biomarkers, and may identify novel therapeutic targets.


Assuntos
Implantação do Embrião/fisiologia , Animais , Endométrio/fisiologia , Células Epiteliais/fisiologia , Matriz Extracelular/fisiologia , Membranas Extraembrionárias/fisiologia , Feminino , Humanos , Gravidez , Técnicas de Reprodução Assistida
5.
Biol Reprod ; 93(2): 50, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26157070

RESUMO

The beginning of embryogenesis is preceded by a sequence of events mediated by the release of intracellular calcium in the ooplasm, a multifaceted process known as oocyte activation. It is now well established that a sperm protein factor introduced into the oocyte at the time of gamete fusion is responsible for initiating the cascade of signaling events involved. Several sperm proteins have been hypothesized as the sperm oocyte-activating factor (SOAF) over the years, with phospholipase C zeta 1 (PLCZ1 or PLCzeta) emerging as the strongest candidate. A large body of consistent and reproducible evidence, from both biochemical and clinical settings, has accumulated in support of PLCzeta, and data clearly demonstrate that oocyte activation ability can be rescued in PLCzeta-deficient sperm by either PLCzeta cRNA or recombinant PLCzeta protein. However, a series of recent publications has challenged the dominance of PLCzeta and proposed an alternative candidate protein, WBP2 N-terminal like (WBP2NL or PAWP). These events have led to significant debate, fueled by the opposing views of two independent laboratories, each defending its own respective SOAF candidate. This raises important questions with regards to the relative importance of these two proteins in diagnostic and therapeutic medicine, and invites urgent research attention. Here, it is our intention to reflect upon this now very controversial area in order to engage the scientific and clinical communities in addressing the true importance of these two sperm proteins.


Assuntos
Oócitos/fisiologia , Interações Espermatozoide-Óvulo/fisiologia , Espermatozoides/fisiologia , Animais , Sinalização do Cálcio/fisiologia , Feminino , Fertilização , Humanos , Masculino , Gravidez
6.
Nanomedicine ; 10(5): 921-38, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24444494

RESUMO

In the last decade, nanotechnology has been extensively introduced for biomedical applications, including bio-detection, drug delivery and diagnostic imaging, particularly in the field of cancer diagnostics and treatment. However, there is a growing trend towards the expansion of nanobiotechnological tools in a number of non-cancer applications. In this review, we discuss the emerging uses of nanotechnology in reproductive medicine and reproductive biology. For the first time, we summarise the available evidence regarding the use of nanomaterials as experimental tools for the detection and treatment of malignant and benign reproductive conditions. We also present an overview of potential applications for nanomaterials in reproductive biology, discuss the benefits and concerns associated with their use in a highly delicate system of reproductive tissues and gametes, and address the feasibility of this innovative and potentially controversial approach in the clinical setting and for investigative research into the mechanisms underlying reproductive diseases. FROM THE CLINICAL EDITOR: This unique review paper focuses on the emerging use of nanotechnology in reproductive medicine and reproductive biology, highlighting the role of nanomaterials in the detection and treatment of various reproductive conditions, keeping in mind the benefits and potential concerns associated with nanomaterial use in the delicate system of reproductive tissue and gametes.


Assuntos
Nanoestruturas/química , Nanoestruturas/uso terapêutico , Nanotecnologia/métodos , Medicina Reprodutiva/métodos , Feminino , Humanos , Masculino
7.
Nanomedicine ; 10(4): 859-70, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24200525

RESUMO

Nanomaterial-mediated delivery represents a promising technique for reproductive biology with a potential to improve the safety and efficacy of existing methodologies, including experimental gene therapy and sperm-mediated gene transfer. Mesoporous silica nanoparticles (MSNPs) have been characterised as a powerful and safe delivery tool, rendering them an excellent candidate for use in reproductive research. However, their effects upon mammalian gametes with highly specialised structure and functionality remain untested. Here, we show for the first time, that spherical MSNPs with hexagonal pore symmetry, functionalised with polyethileneimine and aminopropyltriethoxysilane, and optionally loaded with two common types of cargo (nucleic acid/protein), form strong associations with boar sperm following incubation in vitro and do not exert negative effect upon the main parameters of sperm function, including motility, viability, acrosomal status and DNA fragmentation index. Our findings provide a rationale for the use of MSNPs for the transfer of investigative, diagnostic and/or therapeutic compounds into mammalian sperm. FROM THE CLINICAL EDITOR: Functionalized mesoporous silica nanoparticles (MSNPs) are demonstrated as efficient agents for the transfer of investigative, diagnostic, and/or therapeutic compounds into mammalian sperm. This promising technique has the potential to improve the safety and efficacy of existing methodologies, including experimental gene therapy and sperm-mediated gene transfer.


Assuntos
Fragmentação do DNA/efeitos dos fármacos , Nanopartículas/efeitos adversos , Dióxido de Silício/farmacologia , Espermatozoides/metabolismo , Animais , Masculino , Nanopartículas/química , Polietilenoimina/química , Polietilenoimina/farmacologia , Propilaminas , Silanos/química , Silanos/farmacologia , Dióxido de Silício/química , Espermatozoides/patologia , Suínos
8.
Eur J Cell Biol ; 103(3): 151429, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38905808

RESUMO

Sperm chromatin is distinct from somatic cell chromatin, as a result of extensive remodeling during the final stages of spermatogenesis. In this process, the majority of histones is replaced with protamines. The chromatin is consequently highly condensed and inert, which facilitates protection of the DNA. The sperm epigenomic landscape is shaped by histone retention, histone and protamine modification, DNA methylation, and RNAs. In recent years, sperm chromatin integrity and its epigenetic marks have been increasingly studied, and the constitution of sperm chromatin is steadily being uncovered. This growing body of research prompts assessment of the frequently overlooked involvement of sperm in fertility and embryonic development. Moreover, numerous endogenous and exogenous factors are known to affect sperm chromatin, which may in turn impact the reproductive success. Concerns have been raised about the effects of assisted reproductive technology (ART) on the sperm epigenome, embryonic development and offspring health. This review examines the structure and epigenetic signatures of sperm chromatin in the context of fertility and early embryonic development. Additionally, sperm chromatin evaluation and causes of aberrant integrity are outlined. Building on the knowledge discussed in the current review, future research should aim to elucidate the intricate relationship between all aspects of sperm chromatin and embryo development. This could lead to the uncovering of new targets for treating infertility, as well as the acquisition of much needed insights into the possible reciprocal association between ART and sperm chromatin integrity.

9.
Nanomedicine (Lond) ; 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38686941

RESUMO

Nanoparticles and nanotechnology may present opportunities to revolutionize the prevention, treatment and diagnosis of a range of reproductive health conditions in women. These technologies are also used to improve outcomes of assisted reproductive technology. We highlight a range of these potential clinical uses of nanoparticles for polycystic ovary syndrome, endometriosis, uterine fibroids and sexually transmitted infections, considering in vitro and in vivo studies along with clinical trials. In addition, we discuss applications of nanoparticles in assisted reproductive technology, including sperm loading, gamete and embryo preservation and preventing preterm birth. Finally, we present some of the concerns associated with the medical use of nanoparticles, identifying routes for further exploration before nanoparticles can be applied to women's reproductive health in the clinic.

10.
Biomed Mater ; 19(2)2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38324899

RESUMO

Fertility preservation in prepubertal boys with cancer requires the cryopreservation of immature testicular tissues (ITTs) prior to gonadotoxic treatment. However, the limited number of germ cells in small human ITT biopsies necessitates the development of anin vitroculture system for germ cell expansion using frozen-thawed ITTs. Here, we generated testicular organoids for thein vitromaintenance and expansion of gonocytes from frozen-thawed two-week-old neonatal bovine ITTs. We investigated the effects of different cell-seeding densities, culture serums, seeding methods, and gonadotropin supplementations, on the maintenance and proliferation of enriched gonocytes. Our results demonstrated that enriched gonocytes and testicular cells from frozen-thawed neonatal ITTs could self-assemble into spheroid organoids in three days in an appropriate Matrigel-based culture environment. For the optimal formation of prepubertal testicular organoids, a seeding density of 1 × 106cells/well is recommended over other densities. This strategy results in organoids with a mean diameter of 60.53 ± 12.12 µm; the mean number of organoids was 5.57 ± 1.60/105µm2on day 11. The viability of organoids was maintained at 79.75 ± 2.99% after being frozen and thawed. Supplementing the culture medium with glial cell-derived neurotrophic factor, fibroblast growth factor 2, and leukemia inhibitory factor, increased the proportion of KI67-positive proliferating cells in organoids, elevated the expression ofC-KITbut reduced the expression ofGFRα1at day 28 when compared to those without hormone supplements(p< 0.05). In addition, supplementing the culture medium with follicle-stimulating hormone and testosterone helped to maintain a significantly higher viability (p< 0.05) in ITT organoids at day 28. These organoids could be cryopreserved for storage and thawed as needed. The successful generation of ITT organoids provides a valuable tool for establishingin vitrospermatogenesis, propagating human germ cells, investigating testicular physiology and the origin of germ cell tumors, and testing the toxicity of new drugs in future clinical applications.


Assuntos
Criopreservação , Testículo , Masculino , Animais , Humanos , Bovinos , Testículo/metabolismo , Criopreservação/métodos , Congelamento , Células Germinativas , Organoides
11.
Nat Struct Mol Biol ; 31(6): 964-976, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38789684

RESUMO

The mouse and human embryo gradually loses totipotency before diversifying into the inner cell mass (ICM, future organism) and trophectoderm (TE, future placenta). The transcription factors TFAP2C and TEAD4 with activated RHOA accelerate embryo polarization. Here we show that these factors also accelerate the loss of totipotency. TFAP2C and TEAD4 paradoxically promote and inhibit Hippo signaling before lineage diversification: they drive expression of multiple Hippo regulators while also promoting apical domain formation, which inactivates Hippo. Each factor activates TE specifiers in bipotent cells, while TFAP2C also activates specifiers of the ICM fate. Asymmetric segregation of the apical domain reconciles the opposing regulation of Hippo signaling into Hippo OFF and the TE fate, or Hippo ON and the ICM fate. We propose that the bistable switch established by TFAP2C and TEAD4 is exploited to trigger robust lineage diversification in the developing embryo.


Assuntos
Proteínas de Ligação a DNA , Fatores de Transcrição de Domínio TEA , Fator de Transcrição AP-2 , Fatores de Transcrição , Fator de Transcrição AP-2/metabolismo , Fator de Transcrição AP-2/genética , Animais , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Camundongos , Humanos , Transdução de Sinais , Linhagem da Célula , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/citologia , Via de Sinalização Hippo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Desenvolvimento Embrionário/genética
12.
Mol Reprod Dev ; 80(10): 787-815, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23900730

RESUMO

Fertilization causes mature oocytes or eggs to increase their concentrations of intracellular calcium ions (Ca²âº) in all animals that have been examined, and such Ca²âº elevations, in turn, provide key activating signals that are required for non-parthenogenetic development. Several lines of evidence indicate that the Ca²âº transients produced during fertilization in mammals and other taxa are triggered by soluble factors that sperm deliver into oocytes after gamete fusion. Thus, for a broad-based analysis of Ca²âº dynamics during fertilization in animals, this article begins by summarizing data on soluble sperm factors in non-mammalian species, and subsequently reviews various topics related to a sperm-specific phospholipase C, called PLCζ, which is believed to be the predominant activator of mammalian oocytes. After characterizing initiation processes that involve sperm factors or alternative triggering mechanisms, the spatiotemporal patterns of Ca²âº signals in fertilized oocytes or eggs are compared in a taxon-by-taxon manner, and broadly classified as either a single major transient or a series of repetitive oscillations. Both solitary and oscillatory types of fertilization-induced Ca²âº signals are typically propagated as global waves that depend on Ca²âº release from the endoplasmic reticulum in response to increased concentrations of inositol 1,4,5-trisphosphate (IP3). Thus, for taxa where relevant data are available, upstream pathways that elevate intraoocytic IP3 levels during fertilization are described, while other less-common modes of producing Ca²âº transients are also examined. In addition, the importance of fertilization-induced Ca²âº signals for activating development is underscored by noting some major downstream effects of these signals in various animals.


Assuntos
Sinalização do Cálcio/fisiologia , Fertilização/fisiologia , Espermatozoides/metabolismo , Animais , Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Oócitos/metabolismo , Fosfolipases Tipo C/metabolismo
13.
Theriogenology ; 210: 143-153, 2023 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-37499372

RESUMO

Gonocytes play an important role in early development of spermatogonial stem cells and fertility preservation to acquire more high quality gonocytes in vitro for further germ cell-related research and applications, it is necessarily needed to enrich and in vitro propagate gonocytes from cryopreserved bovine testicular tissues. This study aimed to investigate the isolation, enrichment, and colony formation of gonocytes in vitro for germ cell expansion from cryopreserved neonatal bovine testicular tissues. The effects of several different in vitro culture conditions, including seeding density, temperature, serum replacement and extracellular matrices were investigated for the maintenance, proliferation and formation of gonocyte colonies in vitro. Frozen/thawed two-week-old neonatal bovine testicular tissues were digested and gonocytes were enriched using a Percoll density gradient. Cell viability was accessed by trypan blue staining and cell apoptosis was evaluated by TUNEL assays. Gonocytes were identified and confirmed by immunofluorescence with the PGP9.5 germ cell marker and the OCT4 pluripotency marker while Sertoli cells were stained with vimentin. We found that neonatal bovine gonocytes were efficiently enriched by a 30%-40% Percoll density gradient (p < 0.05). No significant differences were detected between neonatal bovine testicular cells cultured at 34 °C or 37 °C. The formation of gonocyte colonies was observed in culture medium supplemented with knockout serum replacement (KSR), but not fetal bovine serum (FBS), at a seeding density higher than 5.0 × 104 cells/well. A greater number of gonocyte colonies were observed in culture plates coated with laminin (38.00 ± 6.24/well) and Matrigel (38.67 ± 3.78/well) when compared to plates coated with collagen IV and fibronectin (p < 0.05). In conclusion, bovine neonatal gonocytes were able to be efficiently isolated, enriched and maintained in gonocyte colonies in vitro; the development of this protocol provides vital information for the clinical translation of this technology and the future restoration of human fertility.


Assuntos
Células de Sertoli , Testículo , Masculino , Animais , Humanos , Células Germinativas , Proliferação de Células , Espermatogônias
14.
Hum Reprod Update ; 29(4): 395-433, 2023 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-36882116

RESUMO

BACKGROUND: It has long been thought that the factors affecting embryo and foetal development were exclusively maternally derived; hence, if issues regarding fertility and embryo development were to arise, the blame has traditionally been placed solely on the mother. An escalating interest in how paternal factors influence embryo development, however, has begun to prove otherwise. Evidence suggests that both seminal plasma (SP) and sperm contribute multiple factors that shape embryogenesis. This review thus focuses on the role that semen has in driving early embryonic development, and describes how paternal factors, such as SP, sperm centriole, sperm proteins, sperm RNA, sperm DNA, and its integrity, together with epigenetics, may influence the female reproductive tract and post-fertilization events. The important contributions of paternal factors to embryo development highlight the imperative need for further research in this area, which is sure to bring forth breakthroughs leading to improvements in infertility diagnosis and ART as well as reducing the risk of miscarriage. OBJECTIVE AND RATIONALE: This review provides a comprehensive overview of the role of human semen in development of the early embryo, with the aim of providing a better understanding of the influence of SP and sperm on early embryonic divisions, gene and protein expression, miscarriage, and congenital diseases. SEARCH METHODS: PubMed searches were performed using the terms 'sperm structure', 'capacitation', 'acrosome reaction', 'fertilization', 'oocyte activation', 'PLCζ', 'PAWP', 'sperm-borne oocyte activation factor', 'oocyte activation deficiency', 'sperm centriole', 'sperm transport', 'sperm mitochondria', 'seminal plasma', 'sperm epigenetics', 'sperm histone modifications', 'sperm DNA methylation', 'sperm-derived transcripts', 'sperm-derived proteins', 'sperm DNA fragmentation', 'sperm mRNA', 'sperm miRNAs', 'sperm piRNAs', and 'sperm-derived aneuploidy'. The reviewed articles were restricted to those published in English between 1980 and 2022. OUTCOMES: The data suggest that male-derived factors contribute much more than just the male haploid genome to the early embryo. Evidence indicates that semen contributes multiple factors that help shape the fate of embryogenesis. These male-derived factors include contributions from SP, the paternal centriole, RNA and proteins, and DNA integrity. In addition, epigenetic changes have an impact on the female reproductive tract, fertilization, and early stages of embryo development. For example, recent proteomic and transcriptomic studies have identified several sperm-borne markers that play important roles in oocyte fertilization and embryogenesis. WIDER IMPLICATIONS: This review highlights that several male-derived factors are required to work in tandem with female counterparts to allow for correct fertilization and development of the early embryo. A deeper understanding of the contributions of paternal factors that are shuttled over from the sperm cell to the embryo can shed light on how to improve ART from an andrological perspective. Further studies may aid in preventing the passing on of genetic and epigenetic abnormalities of paternal origin, thus decreasing the incidence of male factor infertility. In addition, understanding the exact mechanisms of paternal contribution may assist reproductive scientists and IVF clinicians in determining new causes of recurrent early miscarriage or fertilization failure.


Assuntos
Aborto Espontâneo , Infertilidade Masculina , Sêmen , Feminino , Humanos , Masculino , Gravidez , Infertilidade Masculina/metabolismo , Proteômica , RNA/metabolismo , Espermatozoides/metabolismo
15.
Biol Reprod ; 87(5): 114, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23018181

RESUMO

Assisted reproductive technology (ART) has resulted in more than 5 million births worldwide. However, mainstream ART techniques are not always successful for an estimated 30% of infertile patients in whom gametes are nonviable. Most patients would clearly prefer genetic parenthood, currently possible only via the use of donated gametes or, in future, via the clinical use of artificial gametes (AGs) incorporating parental DNA. Despite much recent progress in the derivation of AGs, significant obstacles remain. Although it is possible to create artificial cells exhibiting some of the molecular and physiological traits of human gametes, they do not yet exhibit the same level of functionality as their in vivo counterparts. Most current effort pays scant attention to confirmation of molecular integrity and clinical applicability of AGs. Here we discuss the various clinical parameters used to assess gamete and embryo viability and discuss markers of gamete function that may be used within future studies attempting to derive AGs. The use of AGs may prove controversial to some members of the general public, and, as such, there is significant need for an appropriate ethical and legal framework governing the clinical use of such cells. However, provided these issues can be successfully overcome, it is highly likely that AGs will represent powerful biological tools for reproductive science, a valuable training resource for embryologists and for potential use in the clinical treatment of human infertility.


Assuntos
Células Germinativas/fisiologia , Técnicas de Reprodução Assistida , Animais , Biomarcadores , Sobrevivência Celular , DNA , Epigênese Genética/genética , Feminino , Humanos , Infertilidade/terapia , Masculino , Doação de Oócitos , Oócitos/fisiologia , Gravidez , Técnicas de Reprodução Assistida/ética , Técnicas de Reprodução Assistida/tendências , Espermatozoides/fisiologia , Doadores de Tecidos
16.
Hum Reprod ; 27(11): 3150-60, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22940771

RESUMO

STUDY QUESTION: Does motile sperm organelle morphology examination (MSOME) affect levels and localization patterns of the oocyte activation factor phospholipase C zeta (PLCζ) in globozoospermic sperm with and without an acrosomal bud? SUMMARY ANSWER: MSOME identified round-headed globozoospermic sperm with increased levels of PLCζ relative to sperm from the same sample that did not undergo MSOME, and identified novel patterns of PLCζ localization in sperm exhibiting an acrosomal bud. WHAT IS KNOWN ALREADY: Absence or reduction in the level of PLCζ in the sperm head, abnormal localization patterning, or defective functional ability as a result of PLCζ gene mutation, have been linked to certain types of human male factor infertility in which oocyte activation is deficient. It has been determined that a subpopulation of sperm (1%) from a patient exhibiting 100% globozoospermia presented with an acrosome bud upon MSOME. A cycle of intracytoplasmic morphologically selected sperm injection, carried out with sperm exhibiting an acrosomal bud led to pregnancy and birth of a healthy baby boy, without the use of assisted oocyte activation (AOA). STUDY DESIGN, SIZE, DURATION: Immunofluorescent analysis of PLCζ in globozoospermic sperm from three patients, before and after MSOME. PARTICIPANTS/MATERIALS, SETTING, METHODS: Quantitative immunofluorescence was used to investigate PLCζ levels and localization patterns in individual sperm (n = 1 patient) identified by MSOME and isolated by micromanipulation, and presenting with and without the acrosomal bud. A secondary aim was to investigate levels and localization patterns of PLCζ in sperm before and after MSOME from two other globozoospermic men. MAIN RESULTS AND THE ROLE OF CHANCE: Non-globozoospermic control sperm exhibited characteristic localization patterns of PLCζ immunofluorescence. Completely round-headed globozoospermic sperm from patients 1-3 were either devoid of PLCζ immunofluorescence, or exhibited an abnormal, punctate, pattern of PLCζ localization. PLCζ immunofluorescence in sperm exhibiting an acrosomal bud was observed in the midpiece with varying fluorescent intensity and was detected in 28.5% of such sperm. The majority of sperm with an acrosomal bud (43.0%) exhibited punctate patterns of PLCζ localization within the sperm head. A further 28.5% of sperm exhibited PLCζ in both the head and the midpiece. Total levels of PLCζ, and the proportions of sperm exhibiting PLCζ immunoreactivity, showed significant variance (P ≤ 0.05) amongst control [45.8 arbitrary units (a.u.) and 95.7%, respectively], non-MSOME-selected (25.9 a.u. and 46.1%, respectively) and MSOME-selected globozoospermic sperm (33.4 a.u. and 65.0%, respectively). Total levels of PLCζ immunofluorescence, and proportions of sperm exhibiting PLCζ immunoreactivity, in control sperm was significantly higher (P≤ 0.05) compared with non-MSOME-selected sperm, but not significantly different from MSOME-selected sperm. LIMITATIONS, REASONS FOR CAUTION: The low numbers of sperm analysed may not be ideal for conclusive statistical analysis. Evaluation of the effects of MSOME on morphologically normal sperm would confirm conclusions. WIDER IMPLICATIONS OF THE FINDINGS: The present findings provide hope for the future treatment of globozoospermia without the need for AOA, and provide further evidence for the clinical application of PLCζ as a therapeutic and prognostic tool. STUDY FUNDING/COMPETING INTEREST(S): The research described herein was funded by the Nuffield Department of Obstetrics and Gynaecology, University of Oxford. The authors report no conflict of interest.


Assuntos
Infertilidade Masculina/patologia , Organelas/patologia , Fosfoinositídeo Fosfolipase C/metabolismo , Análise do Sêmen/efeitos adversos , Cabeça do Espermatozoide/patologia , Acrossomo/metabolismo , Acrossomo/patologia , Adulto , Humanos , Infertilidade Masculina/metabolismo , Masculino , Organelas/metabolismo , Transporte Proteico , Cabeça do Espermatozoide/metabolismo , Peça Intermédia do Espermatozoide/metabolismo , Peça Intermédia do Espermatozoide/patologia
17.
Hum Reprod ; 27(1): 222-31, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22095789

RESUMO

BACKGROUND: Male factor and idiopathic infertility contribute significantly to global infertility, with abnormal testicular gene expression considered to be a major cause. Certain types of male infertility are caused by failure of the sperm to activate the oocyte, a process normally regulated by calcium oscillations, thought to be induced by a sperm-specific phospholipase C, PLCzeta (PLCζ). Previously, we identified a point mutation in an infertile male resulting in the substitution of histidine for proline at position 398 of the protein sequence (PLCζ(H398P)), leading to abnormal PLCζ function and infertility. METHODS AND RESULTS: Here, using a combination of direct-sequencing and mini-sequencing of the PLCζ gene from the patient and his family, we report the identification of a second PLCζ mutation in the same patient resulting in a histidine to leucine substitution at position 233 (PLCζ(H233L)), which is predicted to disrupt local protein interactions in a manner similar to PLCζ(H398P) and was shown to exhibit abnormal calcium oscillatory ability following predictive 3D modelling and cRNA injection in mouse oocytes respectively. We show that PLCζ(H233L) and PLCζ(H398P) exist on distinct parental chromosomes, the former inherited from the patient's mother and the latter from his father. Neither mutation was detected utilizing custom-made single-nucleotide polymorphism assays in 100 fertile males and females, or 8 infertile males with characterized oocyte activation deficiency. CONCLUSIONS: Collectively, our findings provide further evidence regarding the importance of PLCζ at oocyte activation and forms of male infertility where this is deficient. Additionally, we show that the inheritance patterns underlying male infertility are more complex than previously thought and may involve maternal mechanisms.


Assuntos
Infertilidade Masculina/genética , Fosfoinositídeo Fosfolipase C/genética , Fosfoinositídeo Fosfolipase C/metabolismo , Mutação Puntual , Animais , Cálcio/metabolismo , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Camundongos , Mães , Oócitos/citologia , Polimorfismo de Nucleotídeo Único , RNA Complementar/metabolismo , Análise de Sequência de DNA
18.
Cell Commun Signal ; 10(1): 12, 2012 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-22591604

RESUMO

Infertility affects one in seven couples globally and has recently been classified as a disease by the World Health Organisation (WHO). While in-vitro fertilisation (IVF) offers effective treatment for many infertile couples, cases exhibiting severe male infertility (19-57%) often remain difficult, if not impossible to treat. In such cases, intracytoplasmic sperm injection (ICSI), a technique in which a single sperm is microinjected into the oocyte, is implemented. However, 1-5% of ICSI cycles still fail to fertilise, affecting over 1000 couples per year in the UK alone. Pregnancy and delivery rates for IVF and ICSI rarely exceed 30% and 23% respectively. It is therefore imperative that Assisted Reproductive Technology (ART) protocols are constantly modified by associated research programmes, in order to provide patients with the best chances of conception. Prior to fertilisation, mature oocytes are arrested in the metaphase stage of the second meiotic division (MII), which must be alleviated to allow the cell cycle, and subsequent embryogenesis, to proceed. Alleviation occurs through a series of concurrent events, collectively termed 'oocyte activation'. In mammals, oocytes are activated by a series of intracellular calcium (Ca2+) oscillations following gamete fusion. Recent evidence implicates a sperm-specific phospholipase C, PLCzeta (PLCζ), introduced into the oocyte following membrane fusion as the factor responsible. This review summarises our current understanding of oocyte activation failure in human males, and describes recent advances in our knowledge linking certain cases of male infertility with defects in PLCζ expression and activity. Systematic literature searches were performed using PubMed and the ISI-Web of Knowledge. Databases compiled by the United Nations and World Health Organisation databases (UNWHO), and the Human Fertilization and Embryology Authority (HFEA) were also scrutinised. It is clear that PLCζ plays a fundamental role in the activation of mammalian oocytes, and that genetic, molecular, or biochemical perturbation of this key enzyme is strongly linked to human infertility where oocyte activation is deficient. Consequently, there is significant scope for our understanding of PLCζ to be translated to the ART clinic, both as a novel therapeutic agent with which to rescue oocyte activation deficiency (OAD), or as a prognostic/diagnostic biomarker of oocyte activation ability in target sperm samples.

19.
Adv Exp Med Biol ; 740: 1095-121, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22453985

RESUMO

In mammals, gamete fusion initiates a succession of oscillations in the intracellular concentration of calcium within the oocyte, prompting a series of events to occur that are collectively known as "oocyte activation". Such events are a fundamental necessity for the initiation of cell division and subsequent embryogenesis. Compelling evidence now indicates that these calcium oscillations are caused by a testis-specific phospholipase C (PLC) termed PLCzeta (PLCζ), released into the oocyte following gamete fusion. A series of recent studies indicate that abnormal expression or aberrant activity of PLCζ is linked to certain types of human male infertility, where oocyte activation ability is impaired or absent altogether. In this chapter, we discuss the critical role of calcium oscillations in the process of oocyte activation, review the role of PLCs in this fundamental biological reaction, describe how PLCζ has been formally linked to male infertility, and postulate the potential roles for PLCζ in terms of clinical diagnosis and therapy.


Assuntos
Sinalização do Cálcio , Oócitos/fisiologia , Fosfoinositídeo Fosfolipase C/fisiologia , Animais , Fertilização , Humanos , Infertilidade/etiologia , Fosfoinositídeo Fosfolipase C/química
20.
F S Sci ; 3(1): 35-48, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35559994

RESUMO

OBJECTIVE: To investigate the regulatory mechanisms and signaling molecules underlying hatching in mouse embryos. DESIGN: Experimental laboratory study using a mouse embryo model. SETTING: University-based basic scientific research laboratory. ANIMALS: A total of 40 B6C3F1 × B6D2F1 mouse embryos were used in this study. INTERVENTION(S): Frozen/thawed mouse embryos, at the 8-cell stage, were cultured in vitro for 2 days. The resulting hatching and prehatching blastocysts were then used for complementary deoxyribonucleic acid (cDNA) library preparation and ribonucleic acid (RNA) sequencing analysis (n = 8 for each group). Differentially expressed genes were then used for downstream functional analysis. In addition, a list of genes related to developmental progression in humans was used to identify genes that were potentially related to the hatching of human embryos. MAIN OUTCOME MEASURE(S): Differentially expressed genes, enriched Gene Ontology terms and canonical pathways, clustered gene networks, activated upstream regulators, and common genes between a gene list of hatching-related genes in mice and a gene list associated with developmental progression in humans. RESULT(S): A total 275 differentially expressed genes were identified between hatching and prehatching blastocysts: 230 up-regulated and 45 down-regulated genes. Functional enrichment analysis suggested that blastocyst hatching in vitro is an adenosine triphosphate (ATP)-dependent process that involves protein biosynthesis and organization of the cytoskeleton. Furthermore, by regulating cell motility, the RhoA signaling pathway (including Arpc2, Cfl1, Gsn, Pfn1, Tpi1, Grb2, Tmsb10, Enah, and Rnd3 genes) may be a crucial signaling pathway during hatching. We also identified a cluster of genes (Krt8, Krt7, Cldn4, and Aqp3) that exerted functional roles in cell-cell junctions and water homeostasis during hatching. Moreover, some growth factors (angiotensinogen and fibroblast growth factor 2) and endocrine factors (estrogen receptor and prolactin) were predicted to be involved in the regulation of embryo hatching. In addition, we identified 81 potential genes that are potentially involved in the hatching process in human embryos. CONCLUSION(S): Our analysis identified potential genes and molecular regulatory pathways involved in the blastocyst hatching process in mice; we also identified genes that may potentially regulate hatching in human embryos. Our findings enhance our knowledge of embryo development and provide useful information for further exploring the mechanisms underlying embryo hatching.


Assuntos
Blastocisto , RNA , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Blastocisto/metabolismo , Embrião de Mamíferos , Desenvolvimento Embrionário/genética , Feminino , Humanos , Parto , Gravidez , Profilinas/metabolismo , RNA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA