Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Hum Reprod ; 36(9): 2514-2528, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34333622

ABSTRACT

STUDY QUESTION: Does LH protect mouse oocytes and female fertility from alkylating chemotherapy? SUMMARY ANSWER: LH treatment before and during chemotherapy prevents detrimental effects on follicles and reproductive lifespan. WHAT IS KNOWN ALREADY: Chemotherapies can damage the ovary, resulting in premature ovarian failure and reduced fertility in cancer survivors. LH was recently suggested to protect prepubertal mouse follicles from chemotoxic effects of cisplatin treatment. STUDY DESIGN, SIZE, DURATION: This experimental study investigated LH effects on primordial follicles exposed to chemotherapy. Seven-week-old CD-1 female mice were randomly allocated to four experimental groups: Control (n = 13), chemotherapy (ChT, n = 15), ChT+LH-1x (n = 15), and ChT+LH-5x (n = 8). To induce primary ovarian insufficiency (POI), animals in the ChT and ChT+LH groups were intraperitoneally injected with 120 mg/kg of cyclophosphamide and 12 mg/kg of busulfan, while control mice received vehicle. For LH treatment, the ChT+LH-1x and ChT+LH-5x animals received a 1 or 5 IU LH dose, respectively, before chemotherapy, then a second LH injection administered with chemotherapy 24 h later. Then, two animals/group were euthanized at 12 and 24 h to investigate the early ovarian response to LH, while remaining mice were housed for 30 days to evaluate short- and long-term reproductive outcomes. The effects of LH and chemotherapy on growing-stage follicles were analyzed in a parallel experiment. Seven-week-old NOD-SCID female mice were allocated to control (n = 5), ChT (n = 5), and ChT+LH-1x (n = 6) groups. Animals were treated as described above, but maintained for 7 days before reproductive assessment. PARTICIPANTS/MATERIALS, SETTING, METHODS: In the first experiment, follicular damage (phosphorylated H2AX histone (γH2AX) staining and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay), apoptotic biomarkers (western blot), and DNA repair pathways (western blot and RT-qPCR) were assessed in ovaries collected at 12 and 24 h to determine early ovarian responses to LH. Thirty days after treatments, remaining mice were stimulated (10 IU of pregnant mare serum gonadotropin (PMSG) and 10 IU of hCG) and mated to collect ovaries, oocytes, and embryos. Histological analysis was performed on ovarian samples to investigate follicular populations and stromal status, and meiotic spindle and chromosome alignment was measured in oocytes by confocal microscopy. Long-term effects were monitored by assessing pregnancy rate and litter size during six consecutive breeding attempts. In the second experiment, mice were stimulated and mated 7 days after treatments and ovaries, oocytes, and embryos were collected. Follicular numbers, follicular protection (DNA damage and apoptosis by H2AX staining and TUNEL assay, respectively), and ovarian stroma were assessed. Oocyte quality was determined by confocal analysis. MAIN RESULTS AND THE ROLE OF CHANCE: LH treatment was sufficient to preserve ovarian reserve and follicular development, avoid atresia, and restore ovulation and meiotic spindle configuration in mature oocytes exposed at the primordial stage. LH improved the cumulative pregnancy rate and litter size in six consecutive breeding rounds, confirming the potential of LH treatment to preserve fertility. This protective effect appeared to be mediated by an enhanced early DNA repair response, via homologous recombination, and generation of anti-apoptotic signals in the ovary a few hours after injury with chemotherapy. This response ameliorated the chemotherapy-induced increase in DNA-damaged oocytes and apoptotic granulosa cells. LH treatment also protected growing follicles from chemotherapy. LH reversed the chemotherapy-induced depletion of primordial and primary follicular subpopulations, reduced oocyte DNA damage and granulosa cell apoptosis, restored mature oocyte cohort size, and improved meiotic spindle properties. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: This was a preliminary study performed with mouse ovarian samples. Therefore, preclinical research with human samples is required for validation. WIDER IMPLICATIONS OF THE FINDINGS: The current study tested if LH could protect the adult mouse ovarian reserve and reproductive lifespan from alkylating chemotherapy. These findings highlight the therapeutic potential of LH as a complementary non-surgical strategy for preserving fertility in female cancer patients. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by grants from the Regional Valencian Ministry of Education (PROMETEO/2018/137), the Spanish Ministry of Science and Innovation (CP19/00141), and the Spanish Ministry of Education, Culture and Sports (FPU16/05264). The authors declare no conflict of interest.


Subject(s)
Ovarian Reserve , Alkylating Agents/toxicity , Animals , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Ovarian Follicle , Pregnancy
2.
J Eur Acad Dermatol Venereol ; 34(4): 691-708, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31541557

ABSTRACT

BACKGROUND: The incidence of cutaneous melanoma (CM), the deadliest form of skin cancer, has gradually increased in the last decades among populations of European origin. Epidemiological studies suggested that farmers and agricultural workers are at an increased risk of CM because they were exposed to pesticides. However, little is known about the relationship between pesticides and CM. OBJECTIVES: To investigate the association between exposure to pesticides and CM by systematically reviewing the literature. Secondary aim was to determine the categories of pesticides mainly involved in CM development. METHODS: A systematic review of the literature was performed up to September 2018 using MEDLINE, Embase and Web of Science. Studies assessing CM risk in licensed pesticide applicators were considered. Strict criteria were established to select independent studies and risk estimates; random effect models, taking into account heterogeneity, were applied. A pooled risk estimate for CM was calculated for the use of each type of pesticide and type of exposure. Between-study and estimate heterogeneity was assessed and publication bias investigated. RESULTS: A total of nine studies (two case-controls and seven cohorts) comprising 184 389 unique subjects were included. The summary relative risks for the categories 'herbicides - ever exposure', 'insecticides - ever exposure', 'any pesticide - ever exposure' and 'any pesticide - high exposure' resulted 1.85 [95% confidence interval (CI): 1.01, 3.36], 1.57 (95% CI: 0.58, 4.25), 1.31 (95% CI: 0.85, 2.04) and 2.17 (95% CI: 0.45, 10.36), respectively. Herbicides and insecticides had no between-study heterogeneity (I2  = 0%), while a significant heterogeneity (I2  > 50%) was detected for the high exposure to any pesticide. No indication for publication bias was found. CONCLUSIONS: Individuals exposed to herbicides are at an increased risk of CM. Future properly designed observational studies are required to confirm this finding.


Subject(s)
Melanoma/chemically induced , Occupational Diseases/chemically induced , Occupational Exposure/adverse effects , Pesticides/toxicity , Skin Neoplasms/chemically induced , Humans , Melanoma, Cutaneous Malignant
3.
Hum Reprod Update ; 25(6): 673-693, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31600388

ABSTRACT

BACKGROUND: Anti-cancer therapy is often a cause of premature ovarian insufficiency and infertility since the ovarian follicle reserve is extremely sensitive to the effects of chemotherapy and radiotherapy. While oocyte, embryo and ovarian cortex cryopreservation can help some women with cancer-induced infertility achieve pregnancy, the development of effective methods to protect ovarian function during chemotherapy would be a significant advantage. OBJECTIVE AND RATIONALE: This paper critically discusses the different damaging effects of the most common chemotherapeutic compounds on the ovary, in particular, the ovarian follicles and the molecular pathways that lead to that damage. The mechanisms through which fertility-protective agents might prevent chemotherapy drug-induced follicle loss are then reviewed. SEARCH METHODS: Articles published in English were searched on PubMed up to March 2019 using the following terms: ovary, fertility preservation, chemotherapy, follicle death, adjuvant therapy, cyclophosphamide, cisplatin, doxorubicin. Inclusion and exclusion criteria were applied to the analysis of the protective agents. OUTCOMES: Recent studies reveal how chemotherapeutic drugs can affect the different cellular components of the ovary, causing rapid depletion of the ovarian follicular reserve. The three most commonly used drugs, cyclophosphamide, cisplatin and doxorubicin, cause premature ovarian insufficiency by inducing death and/or accelerated activation of primordial follicles and increased atresia of growing follicles. They also cause an increase in damage to blood vessels and the stromal compartment and increment inflammation. In the past 20 years, many compounds have been investigated as potential protective agents to counteract these adverse effects. The interactions of recently described fertility-protective agents with these damage pathways are discussed. WIDER IMPLICATIONS: Understanding the mechanisms underlying the action of chemotherapy compounds on the various components of the ovary is essential for the development of efficient and targeted pharmacological therapies that could protect and prolong female fertility. While there are increasing preclinical investigations of potential fertility preserving adjuvants, there remains a lack of approaches that are being developed and tested clinically.


Subject(s)
Antineoplastic Agents/adverse effects , Fertility Preservation/methods , Infertility, Female/chemically induced , Ovarian Follicle/pathology , Ovarian Reserve/drug effects , Primary Ovarian Insufficiency/chemically induced , Cisplatin/adverse effects , Cryopreservation , Cyclophosphamide/adverse effects , Doxorubicin/adverse effects , Female , Fertility/physiology , Humans , Oocytes/physiology , Pregnancy
4.
Cell Death Dis ; 6: e1906, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26469955

ABSTRACT

Stem cells are unique cell types capable to proliferate, some of them indefinitely, while maintaining the ability to differentiate into a few or any cell lineages. In 2003, a group headed by Hans R. Schöler reported that oocyte-like cells could be produced from mouse embryonic stem (ES) cells in vitro. After more than 10 years, where have these researches reached? Which are the major successes achieved and the problems still remaining to be solved? Although during the last years, many reviews have been published about these topics, in the present work, we will focus on an aspect that has been little considered so far, namely a strict comparison between the in vitro and in vivo developmental capabilities of the primordial germ cells (PGCs) isolated from the embryo and the PGC-like cells (PGC-LCs) produced in vitro from different types of stem cells in the mouse, the species in which most investigation has been carried out. Actually, the formation and differentiation of PGCs are crucial for both male and female gametogenesis, and the faithful production of PGCs in vitro represents the basis for obtaining functional germ cells.


Subject(s)
Gametogenesis , Germ Cells/physiology , Stem Cells/physiology , Animals , Cell Differentiation , Cells, Cultured , Humans
5.
Cell Cycle ; 14(7): 1036-45, 2015.
Article in English | MEDLINE | ID: mdl-25603532

ABSTRACT

As the name implies, Stimulated by Retinoic Acid 8 is an early retinoic acid (RA) responsive gene pivotal for the beginning of meiosis in female and male germ cells. Its expression is strictly time-dependent and cell-specific (pre-meiotic germ cells) and likely requires a complex mechanism of regulation. In this study, we demonstrate a direct negative control of SOHLH1 and SOHLH2, 2 germ cell specific bHLH transcription factors, on Stra8 expression. We observed a negative correlation between STRA8 and SOHLH1 expression in prepuberal differentiating mouse KIT(+) spermatogonia and found that SOHLH1 and SOHLH2 were able to directly and cooperatively repress STRA8 expression in cell lines in vitro through binding to its promoter. We also identified 2 canonical E-Box motives in the Stra8 promoter that mediated the negative regulation of SOHLH1 and SOHLH2 on these gene both in the cell lines and KIT(+) spermatogonia. We hypothesize that this novel negative activity of SOHLH1 and SOHLH2 in male cooperates with that of other transcription factors to coordinate spermatogonia differentiation and the RA-induced meiosis and in female ensures STRA8 down-regulation at mid-end stages of meiotic prophase I.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Basic Helix-Loop-Helix Transcription Factors/physiology , Adaptor Proteins, Signal Transducing/genetics , Base Sequence , Cell Differentiation , Down-Regulation , Gene Expression , Gene Silencing , HEK293 Cells , Humans , Male , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding , Spermatogonia/physiology
6.
Andrology ; 3(2): 265-70, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25598385

ABSTRACT

Measurement of reactive oxygen species (ROS) producing leukocytes in semen has been a standard component of the semen analysis, but its true significance remains still unknown. In this study, we have correlated the number of seminal leukocytes to various semen parameters. We found a negative correlation between the leukocyte number and sperm concentration (rs  = -0.22; p = 0.01) and motility (rs  = -0.20; p = 0.02). In contrast, a positive correlation between the number of leukocytes and both seminal ROS (rs  = 0.70, p < 0.001; n = 125) and the number of spermatozoa with DNA fragmentation (rs  = 0.43, p = 0.032; n = 25) was found. However, only a trend of positive correlation between ROS and the number of spermatozoa with TUNEL-detected DNA fragmentation was observed. Moreover, this latter was not correlated with loss of sperm mitochondrial membrane potential (MMP) (10% vs 35%, rs  = 0.25, p = 0.08; n = 50). Overall these results indicate that the presence of high number of leukocytes in the ejaculate negatively affects key semen parameters, as sperm concentration and motility, associated with infertility conditions. Moreover, they suggest that leukocytes are the major source of the seminal ROS and cause of sperm DNA fragmentation. However, the absence of a clear correlation between ROS and sperm DNA fragmentation, and spermatozoa with damaged DNA and MMP loss, suggest that ROS produced by leukocytes might be not the only cause of DNA damage in spermatozoa and that intrinsic mitochondrial-dependent apoptotic pathways might not have a major impact on sperm DNA fragmentation.


Subject(s)
DNA Damage , Leukocytes/cytology , Membrane Potential, Mitochondrial , Reactive Oxygen Species/metabolism , Semen/cytology , Spermatozoa/cytology , Humans , Male
7.
Ann N Y Acad Sci ; 1163: 475-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19456391

ABSTRACT

Here we show that genistein, through an estrogen receptor-mediated action, modulates gene expression in the mouse testis throughout development. Genistein passed from the lactating mother to the suckling offspring at levels sufficient to activate gene expression in the testis of the pups. Testis are already responsive to genistein as well as to estradiol at day 14.5 of fetal development. Activation of luciferase correlates with an activation of cell proliferation. In conclusion, our results show that genistein affects reproductive organs of male mice at all developmental ages.


Subject(s)
Genistein/pharmacology , Receptors, Estrogen/metabolism , Testis/drug effects , Testis/metabolism , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Animals , Male , Mice , Mice, Transgenic , Receptors, Estrogen/genetics , Tissue Culture Techniques
8.
Phys Med Biol ; 53(20): 5675-88, 2008 Oct 21.
Article in English | MEDLINE | ID: mdl-18812651

ABSTRACT

Myelin is a multi-lamellar membrane surrounding neuronal axons and increasing their conduction velocity. When investigated by small-angle x-ray scattering (SAXS), the lamellar quasi-periodical arrangement of the myelin sheath gives rise to distinct peaks, which allow the determination of its molecular organization and the dimensions of its substructures. In this study we report on the myelin sheath structural determination carried out on a set of human brain tissue samples coming from surgical biopsies of two patients: a man around 60 and a woman nearly 90 years old. The samples were extracted either from white or grey cerebral matter and did not undergo any manipulation or chemical-physical treatment, which could possibly have altered their structure, except dipping them into a formalin solution for their conservation. Analysis of the scattered intensity from white matter of intact human cerebral tissue allowed the evaluation not only of the myelin sheath periodicity but also of its electronic charge density profile. In particular, the thicknesses of the cytoplasm and extracellular regions were established, as well as those of the hydrophilic polar heads and hydrophobic tails of the lipid bilayer. SAXS patterns were measured at several locations on each sample in order to establish the statistical variations of the structural parameters within a single sample and among different samples. This work demonstrates that a detailed structural analysis of the myelin sheath can also be carried out in randomly oriented samples of intact human white matter, which is of importance for studying the aetiology and evolution of the central nervous system pathologies inducing myelin degeneration.


Subject(s)
Brain/diagnostic imaging , Brain/ultrastructure , Myelin Sheath/diagnostic imaging , Myelin Sheath/ultrastructure , Scattering, Small Angle , X-Ray Diffraction/methods , Humans , Molecular Conformation , Radiography
9.
Reproduction ; 134(2): 241-52, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17660234

ABSTRACT

We report a short-term culture system that allows to define novel characteristic of programmed cell death (PCD) in fetal oocytes and to underscore new aspects of this process. Mouse fetal oocytes cultured in conditions allowing meiotic prophase I progression underwent apoptotic degeneration waves as revealed by TUNEL staining. TEM observations revealed recurrent atypical apoptotic morphologies characterized by the absence of chromatin margination and nuclear fragmentation; oocytes with autophagic and necrotic features were also observed. Further characterization of oocyte death evidenced DNA ladder, Annexin V binding, PARP cleavage, and usually caspase activation (namely caspase-2). In the aim to modulate the oocyte death process, we found that the addition to the culture medium of the pan-caspase inhibitors Z-VAD or caspase-2-specific inhibitor Z-VDVAD resulted in a partial and transient prevention of this process. Oocyte death was significantly reduced by the antioxidant agent NAC and partly prevented by KL and IGF-I growth factors. Finally, oocyte apoptosis was reduced by calpain inhibitor I and increased by rapamycin after prolonged culture. These results support the notion that fetal oocytes undergo degeneration mostly by apoptosis. This process is, however, often morphologically atypical and encompasses other forms of cell death including caspase-independent apoptosis and autophagia. The observation that oocyte death occurs mainly at certain stages of meiosis and can only be attenuated by typical anti-apoptotic treatments favors the notion that it is controlled at least in part by stage-specific oocyte-autonomous meiotic checkpoints and when activated is little amenable to inhibition being the oocyte able to switch back and forth among different death pathways.


Subject(s)
Apoptosis/physiology , Fetus/physiology , Meiotic Prophase I/physiology , Oocytes/cytology , Ovary/cytology , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Autophagy , Caspase Inhibitors , Cell Culture Techniques , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , DNA Fragmentation , Female , Glycoproteins/pharmacology , Immunosuppressive Agents/pharmacology , In Situ Nick-End Labeling , Insulin-Like Growth Factor I/pharmacology , Mice , Mice, Inbred Strains , Microscopy, Electron, Transmission , Necrosis , Oligopeptides/pharmacology , Oocytes/drug effects , Oocytes/ultrastructure , Sirolimus/pharmacology , Stem Cell Factor/pharmacology
10.
Hum Reprod ; 21(12): 3185-92, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16905672

ABSTRACT

BACKGROUND: There is much evidence involving the KIT tyrosine kinase receptor and its ligand KITLG in the survival and proliferation of germ cells. Animal models and functional studies in humans suggest that this signalling pathway plays a role in male infertility. METHODS: We studied three and two single-nucleotide polymorphisms (SNPs) (rs3819392, rs3134885, rs2237012, rs10506957 and rs995030) located within the genomic region of the KIT and KITLG genes, respectively. A total of 167 idiopathic infertile men (sperm counts <5 million spz/ml) and 465 unrelated healthy controls from the same geographical region were genotyped for these SNPs. RESULTS: We found a statistically significant association of the rs3819392 polymorphism, which is located within the KIT gene, with idiopathic male infertility. In addition, a deviation from the Hardy-Weinberg equilibrium (HWE) law was observed for rs10506957 polymorphism within the KITLG gene only in the infertile group. CONCLUSIONS: Our data indicate that the KIT/KITLG system may be involved in a low sperm count trait in humans.


Subject(s)
Genetic Markers/genetics , Infertility, Male/genetics , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-kit/genetics , Stem Cell Factor/genetics , Chromosome Mapping , Gene Frequency , Haplotypes , Humans , Infertility, Male/etiology , Male
11.
Med Phys ; 32(8): 2455-63, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16193774

ABSTRACT

We present EGS4 Monte Carlo calculations of the spatial distribution of the dose deposited by a single x-ray pencil beam, a planar microbeam, and an array of parallel planar microbeams as used in radiation therapy research. The profiles of the absorbed dose distribution in a phantom, including the peak-to-valley ratio of the dose distribution from microbeam arrays, were calculated at micrometer resolution. We determined the dependence of the findings on the main parameters of photon and electron transport. The results illustrate the dependence of the electron range and the deposited in-beam dose on the cut-off energy, of the electron transport, as well as the effects on the dose profiles of the beam energy, the array size, and the beam spacing. The effect of beam polarization also was studied for a single pencil beam and for an array of parallel planar microbeams. The results show that although the polarization effect on the dose distribution from a 3 cm x 3 cm microbeam array inside a water phantom is large enough to be measured at the outer side of the array (16% difference of the deposited dose for x-ray beams of 200 keV), it is not detectable at the array's center, thus being irrelevant for the radiation therapy purposes. Finally we show that to properly compare the dose profiles determined with a metal oxide semiconductor field emission transistor detector with the computational method predictions, it is important to simulate adequately the size and the material of the device's Si active element.


Subject(s)
Models, Biological , Radiometry/methods , Radiotherapy Planning, Computer-Assisted/methods , Software , Body Burden , Computer Simulation , Models, Statistical , Monte Carlo Method , Radiotherapy Dosage , Relative Biological Effectiveness , Scattering, Radiation , X-Ray Therapy/methods , X-Rays
12.
Hum Reprod Update ; 10(3): 197-206, 2004.
Article in English | MEDLINE | ID: mdl-15140867

ABSTRACT

New information regarding primordial germ cell (PGC's) segregation and proliferation over the last decade is reviewed. Advances have been obtained in the mouse but current knowledge of human PGC's remains scant. Questions still fully or partially unresolved about the emergence of the germline in mammals are addressed. (i) When and where is the germ line set aside in the embryo? (ii) How is the germ line segregated from the somatic lineages? (iii) Which factors guide PGC's to the gonadal ridges? (iv) Which factors regulate PGC's proliferation? The main purpose of this review is to outline the information obtained using mainly in vitro culture systems about two aspects of these processes namely the segregation of PGC's and their proliferation.


Subject(s)
Germ Cells/cytology , Gonads/cytology , Gonads/embryology , Animals , Cell Division/physiology , Cell Lineage/physiology , Humans
13.
Genomics ; 82(2): 230-7, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12837272

ABSTRACT

Repetitive DNA elements account for a substantial fraction of the mammalian genome. Many are subject to DNA methylation, which is known to undergo dynamic change during mouse germ cell development. We found that repeat sequences of three different classes retain high levels of methylation at E12.5, when methylation is erased from many single-copy genes. Maximal demethylation of repeats was seen later in development and at different times in male and female germ cells. At none of the time points examined (E12.5, E15.5, and E17.5) did we see complete demethylation, suggesting that methylation patterns on repeats may be passed on from one generation to the next. In male germ cells, we observed a de novo methylation event resulting in complete methylation of all the repeats in the interval between E15.5 and E17.5, which was not seen in females. These results suggest that repeat sequences undergo coordinate changes in methylation during germ cell development and give further insights into germ cell reprogramming in mice.


Subject(s)
DNA Methylation , Germ Cells/chemistry , Mice/genetics , Repetitive Sequences, Nucleic Acid/genetics , Animals , Blotting, Southern , Female , Histological Techniques , Male
14.
Cell Biol Toxicol ; 18(2): 131-45, 2002.
Article in English | MEDLINE | ID: mdl-12046691

ABSTRACT

Several strategies for the assessment of reproductive toxicity of chemical compounds has have been proposed. In the present work, we devised experimental in vitro assays to test the effect of potential toxicants on proliferating primordial germ cells (PGCs) in vitro using recently developed methods for isolation and culture of mouse PGCs. Primordial germ cells are the embryonic precursors of gametes of the adult that carry the genome from generation to generation. Any damage or mutations caused to these cells by potential toxicants might impair normal reproduction and be transmitted to next generation. Three representative compounds, N-ethyl-N-nitrosourea (ENU), adriamycin (ADR), and mono-(2-ethylhexyl)phthalate (MEHP), toxic to different targets and known to affect germ cell development and impair fertility, were tested on PGCs in culture using three different experimental protocols. Survival and growth of PGCs and their ability to adhere to cell monolayers, were taken as endpoints for drug effects. For each compound, sublethal and acute toxicity doses were determined. In addition, information about the mechanisms of action of these compounds on PGCs was obtained. Whereas the effects of ENU and ADR on PGCs were attributable to growth inhibition and apoptosis induction, MEHP affected PGC adhesion to somatic cells without significantly altering their growth and survival. The results of our in vitro tests were not always exactly predictive of the effects of the tested compounds on PGCs in vivo, determined in parallel experiments in which pregnant mice were exposed to the same compounds. Nevertheless, they can provide information on the sensitivity of PGCs to the direct action of drugs or the mechanisms of action of such agents.


Subject(s)
Diethylhexyl Phthalate/analogs & derivatives , Diethylhexyl Phthalate/toxicity , Doxorubicin/toxicity , Ethylnitrosourea/toxicity , Germ Cells/drug effects , Teratogens/toxicity , Animals , Cell Line , Female , In Situ Nick-End Labeling , Mice , Pregnancy
15.
Zygote ; 9(3): 201-10, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11508739

ABSTRACT

In this study the solubility to alpha-chymotrypsin of the zona pellucida (ZP) of human oocytes and polyploid embryos obtained during various clinical procedures of assisted fertilisation (IVF, ICSI, cyropreservation) was evaluated. The aim of the study was to determine whether changes in ZP solubility occur during such procedures and whether abnormal solubility could be likened to fertilisation failure. Correlation between ZP solubility and cortical granule (CG) density was also studied. The results showed that ZP solubility varied considerably among germinal vesicle or metaphase oocytes obtained from different subjects, but was essentially identical for the oocyte cohort obtained from individual women. On the basis of ZP solubility metaphase oocytes were subdivided into two classes: class I, average ZP dissolution time +/- SE = 24.1+/-0.9 min, n = 28; and class II, 46.7+/-2.0 min, n = 13. Prolonged ZP dissolution times of metaphase oocytes were significantly correlated with a low in vitro fertilisation rate in sibling oocytes. The zonae of fertilised eggs (polyploid embryos) showed long solubilisation times (IVF: 45.3+/-3.4 min, n = 18; ICSI: 48.9+/-2.7 min, n = 19). ZP solubility of oocytes that failed to fertilise was intermediate between that of class I metaphase oocytes and embryos (unfertilised IVF: 33.0+/-2.7 min, n = 13; unfertilised ICSI: 43.0+/-2.4 min, n = 9). A moderate spontaneous ZP hardening occurred when metaphase oocytes were cultured for 24 h. Finally, cryopreservation of unfertilised oocytes caused hardening of their ZP, with dissolution times that were comparable to those found in fertilised eggs (49.5+/-2.3 min, n = 10). In most cases, an inverse correlation was found between ZP dissolution time and CG density (longer solubilisation times corresponding to lower CG density). ZP hardening caused by cryopreservation, however, was not associated with a significant reduction in CG density in most of the oocytes examined.


Subject(s)
Oocytes/metabolism , Oocytes/ultrastructure , Zona Pellucida/metabolism , Adult , Chymotrypsin/pharmacology , Cryopreservation , Exocytosis , Female , Fertilization , Fertilization in Vitro/methods , Humans , Metaphase , Oocytes/physiology , Sperm Injections, Intracytoplasmic/methods , Time Factors , Zona Pellucida/drug effects , Zona Pellucida/physiology
16.
Int J Dev Biol ; 45(3): 519-22, 2001.
Article in English | MEDLINE | ID: mdl-11417894

ABSTRACT

Just twenty years ago I was preparing a research project centred on establishing methods for the isolation and culture of mouse primordial germ cells (PGCs). The project had been suggested to me by Anne McLaren and was to be developed at the Medical Research Council (MRC) "Mammalian Development Unit" in London under the direction of Anne herself. At that time I was a young postdoctoral researcher at the Institute of Histology and Embryology of the University of Rome "La Sapienza" and did not imagine that my decision to be involved in this project would signal a profound switch in my scientific life. From then on my research would mostly concentrate on primordial germ cell biology. I feel like saying that the modern history of mammalian primordial germ cells began twenty years ago at the MRC Mammalian Development Unit under Anne McLaren's impulse. It is not surprising that among the most active researchers in the last twenty years in studying mammalian primordial germ cells, three, namely Chris Wylie, Peter Donovan and myself, began their studies under Anne McLaren's guidance. Over the years, Anne's suggestions and encouragement were always precious for my studies and her presence marked my most important findings on PGC biology. She often invited me to present the results obtained in my laboratory to workshops and congresses. In the present article some of these results particularly influenced by Anne's teaching and suggestions will be briefly reviewed.


Subject(s)
Embryology/history , Germ Cells , Animals , Cell Differentiation , Female , Germ Cells/cytology , History, 20th Century , Italy , Male , Meiosis , Mice , Pregnancy , Research/history , United Kingdom
17.
J Mol Biol ; 304(4): 529-40, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-11099378

ABSTRACT

Oct-4 is a transcription factor that is specifically expressed in mouse embryonic stem cells and in cell lines derived thereof. In these cells, Oct-4 activates transcription from remote binding sites due to as of yet unknown co-activators. Expression of Oct-4 in differentiated cells is not sufficient to activate transcription from a distance, rather it requires the co-expression of co-activators such as the adenoviral oncoprotein E1A. In this paper, we used phage display to identify Oct-4-interacting proteins. We first analyzed the interaction between Oct-4 and E1A in order to optimize the biochemical conditions that enable Oct-4-specific interactions with other interacting proteins. A panning approach was used to enrich Oct-4 interacting phages from a pool of excess unspecific phages. The biochemical conditions established in our interaction assays were then used to screen a P19 EC cell cDNA expression library in M13 filamentous phage. A number of phage clones displaying portions of unknown and known transcription factors were obtained, from which the HMG-1 transcription factor was identified. HMG-1, and the closely related factor HMG-2, interact with Oct-4 when co-expressed in mammalian cells. In addition, HMG-1 was found to cooperate with Oct-4 in P19 EC cells. These results provide the first evidence of a non-viral factor that enhances Oct-4 distance-dependent transactivation in stem cells.


Subject(s)
DNA-Binding Proteins/metabolism , Germ Cells/metabolism , Peptide Library , Transcription Factors , Adenovirus E1A Proteins/chemistry , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Animals , Bacteriophage M13/genetics , COS Cells , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , High Mobility Group Proteins/chemistry , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Mice , Octamer Transcription Factor-3 , Organ Specificity , Precipitin Tests , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Transfection , Tumor Cells, Cultured
19.
Int J Dev Biol ; 44(6): 575-80, 2000.
Article in English | MEDLINE | ID: mdl-11061420

ABSTRACT

Primordial germ cells (PGCs) are the founders of the gametes. They arise at the earliest stages of embryonic development and migrate to the gonadal ridges, where they differentiate into oogonia/oocytes in the ovary, and prospermatogonia in the testis. The present article is a review of the main studies undertaken by the author with the aim of clarifying the mechanisms underlying the development of primordial germ cells. Methods for the isolation and purification of migratory and post-migratory mouse PGCs devised in the author's laboratory are first briefly reviewed. Such methods, together with the primary culture of PGCs onto suitable cell feeder layers, have allowed the analysis of important aspects of the control of their development, concerning in particular survival, proliferation and migration of mouse PGCs. Compounds and growth factors affecting PGC numbers in culture have been identified. These include survival anti-apoptotic factors (SCF, LIF) and positive regulators of proliferation (cAMP, PACAPs, RA). Evidence has been provided that the motility of migrating PGCs relies on integrated signals from extracellular matrix molecules and the surrounding somatic cells. Moreover, homotypic PGC-PGC interaction has been evidenced that might play a role in PGC migration and in regulating their development. Several molecules (i.e. integrins, specific types of oligosaccharides, E-cadherin, the tyrosine kinase receptor c-kit) have been found to be expressed on the surface of PGCs and to mediate adhesive interactions of PGCs with the extracellular matrix, somatic cells and neighbouring PGCs.


Subject(s)
Cell Culture Techniques/methods , Germ Cells/growth & development , Germ Cells/metabolism , Animals , Cell Adhesion , Cell Movement , Cell Survival , Coculture Techniques , Growth Substances/metabolism , Mice
20.
Dev Biol ; 226(2): 209-19, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11023681

ABSTRACT

In this study we show that mouse primordial germ cells and fetal germ cells at certain stages of differentiation express E-cadherin and alpha and beta catenins. Moreover, we demonstrate that the formation of germ cell aggregates that rapidly occurs when monodispersed germ cell populations are released from embryonic gonads in culture is E-cadherin mediated, developmentally regulated, and dependent on the sex of the germ cells. Immunoblotting analyses indicate that the lower ability to form aggregates of primordial germ cells in comparison to fetal germ cells is not due to gross changes in E-cadherin expression, altered association with beta catenin, or changes in beta catenin phosphorylation. Investigating possible functions of E-cadherin-mediated adhesion in primordial germ cell development, we found that E-cadherin-mediated adhesion may stimulate the motility of primordial germ cells. Moreover, treatment of primordial germ cells cultured on STO cell monolayers with an anti-E-cadherin antibody caused a significant decrease in their number and markedly reduced their ability to form colonies in vitro. The same in vitro treatment of explanted undifferentiated gonadal ridges cultured for 4 days results in decreased numbers and altered localization of the germ cell inside the gonads. Taken together these results suggest that E-cadherin plays an important role in primordial germ cell migration and homing and may act as a modulator of primordial germ cell development.


Subject(s)
Cadherins/physiology , Germ Cells/metabolism , Trans-Activators , Animals , Antibodies, Monoclonal/pharmacology , Cadherins/biosynthesis , Cadherins/genetics , Cadherins/immunology , Calcium/pharmacology , Cell Adhesion , Cell Aggregation/drug effects , Cell Differentiation , Cell Movement , Cells, Cultured , Colony-Forming Units Assay , Cytochalasin B/pharmacology , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , Female , Gene Expression Regulation, Developmental , Germ Cells/cytology , Germ Cells/drug effects , Gonads/cytology , Gonads/embryology , Male , Mice , Peptide Fragments/pharmacology , Phosphorylation , Protein Processing, Post-Translational , Temperature , Trypsin/pharmacology , alpha Catenin , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...