10 research outputs found

    Analysis of epididymal sperm maturation by MALDI profiling and top-down mass spectrometry

    No full text
    The fertilization ability of male gametes is achieved after their transit through the epididymis where important post-gonadal differentiation occurs in different cellular compartments. Most of these maturational modifications occur at the protein level. The epididymal sperm maturation process was investigated using the ICM-MS (Intact Cell MALDI-TOF MS) approach on boar spermatozoa isolated from four different epididymal regions (immature to mature stage). Differential and quantitative MALDI-TOF profiling for whole cells or sub-cellular fractions was combined with targeted top-down MS in order to identify endogenous biomolecules. Using this approach, 172 m/z peaks ranging between 2 and 20 kDa were found to be modified during maturation of sperm. Using top-down MS, 62 m/z were identified corresponding to peptidoforms/proteoforms with post-translational modifications (MS data are available via ProteomeXchange with identifier PXD001303). Many of the endogenous peptides were characterized as N-, C-terminal sequences or internal fragments of proteins presenting specific cleavages, suggesting the presence of sequential protease activities in the spermatozoa. This is the first time that such proteolytic activities could be evidenced for various sperm proteins through quantification of their proteolytic products. ICM-MS/top-down MS thus proved to be a valid approach for peptidome/degradome studies and provided new contributions to understanding of the maturation process of the male gamete involved in the development of male fertility.Biological significanceThis peptidomic study (i) characterized the peptidome of epididymal spermatozoa from boar (Sus scrofa); (ii) established characteristic molecular phenotypes distinguishing degrees of maturation of spermatozoa during epididymal transit, and (iii) revealed that protease activities were at the origin of numerous peptides from known and unknown proteins involved in sperm maturation and/or fertility processes

    Impact of human and mouse sPLA<sub>2</sub>s on platelet MPs.

    No full text
    <p>(A) MPs from human platelets (stimulated with collagen) labeled with the CMFDA cell tracker were incubated for 1 and 6 hours at 37°c in absence or in presence of indicated concentrations of human recombinant sPLA<sub>2</sub> IIA, V, X, or 1µg/ml of the inactive mutant V H48Q. Fluorochrome-conjugated antibodies directed against CD41 and fluorochrome-conjugated annexin-V were used to assess the quantities of CMFDA<sup>+</sup> MPs (left panel), of CD41<sup>+</sup> MPs (middle panel), of annexin-V<sup>+</sup> MPs (right panel) and were compared to the untreated conditions (dotted line). Data are mean ± SEM of 5 independent experiments presented as % of untreated (control) (B) MPs from mouse platelets (stimulated with collagen), identified using YFP as fluorescent tracker, were incubated 1 and 6 hours at 37°c, in absence or in presence of indicated concentrations of mouse recombinant sPLA<sub>2</sub> IIA, V, X, or 1µg/ml of the inactive mutant X H48Q. Fluorochrome-conjugated antibodies directed against CD41 and fluorochrome-conjugated annexin-V were used to determine the concentrations of YFP<sup>+</sup> MPs (left panel), of CD41<sup>+</sup> MPs (middle panel), of annexin-V<sup>+</sup> MPs (right panel) and then compared to the untreated conditions (dotted line). Data are mean ± SEM of 5 independent experiments presented as % of untreated (control). (C) MPs from human platelets labeled with the CMFDA cell tracker and obtained following stimulation with collagen, thrombin or HA-IgG were incubated 6 hours at 37°c in absence or in presence of indicated concentration of human recombinant sPLA<sub>2</sub> IIA, V and X and 1µg/ml of the inactive mutant sPLA<sub>2</sub> V H48Q. Fluorochrome-conjugated antibodies directed against CD41 and fluorochrome-conjugated annexin-V were used to assess the quantities of CMFDA<sup>+</sup> MPs (left panel), of CD41<sup>+</sup> MPs (middle panel), of annexin-V<sup>+</sup> MPs (right panel) and then compared to the untreated conditions (dotted line). Data are mean ± SEM of 3 independent experiments presented as % of untreated (control). (D) MPs from human platelets (stimulated with collagen) labeled with the CMFDA cell tracker were incubated 6 hours at 37°c in PFP of C57BL6 (supplemented or not with 1µg/ml of recombinant human sPLA<sub>2</sub> IIA) or transgenic mice expressing the human sPLA<sub>2</sub> IIA (Tg). Fluorochrome-conjugated antibodies directed against CD41 and fluorochrome-conjugated annexin-V were used to assess the quantities of CMFDA<sup>+</sup> MPs (left panel), of CMFDA<sup>+</sup> CD41<sup>+</sup> MPs (middle panel) and CMFDA<sup>+</sup> annexin-V<sup>+</sup> MPs (right panel). Data are mean ± SEM of 3 independent experiments. (E) Concentrations of Annexin-V<sup>+</sup> MPs and CD41<sup>+</sup> MPs present in the synovial fluids of RA patients determined by high sensitivity flow cytometry and correlated to the concentration of human sPLA<sub>2</sub> IIA assayed (in the same synovial fluids) by time-resolved immunofluorescence analysis. * P< .05; # P< .01; § P< .001.</p

    Study of swarm detection in high sensitivity flow cytometry.

    No full text
    <p>(A) A mixture of CMFDA<sup>-</sup> and CMFDA<sup>+</sup> platelet MPs (CMFDA<sup>- and +</sup>) and sky blue beads (220 nm in diameter) were analyzed alone (left and middle panel respectively) or mixed (right panel) and their detection resolved on the basis of fluorescence. (B) CMFDA<sup>- and +</sup> platelet MPs and sky blue beads (450 nm in diameter) were analyzed alone (left and middle panel respectively) or mixed (right panel) prior to detection on the basis of fluorescence. (C) CMFDA<sup>+</sup> platelet MPs and RBC MPs labeled with antibodies directed against TER 119 are analyzed alone (left and middle panel respectively) or mixed (right panel). (D, E, F) CMFDA<sup>+</sup> platelet MPs were diluted serially thrice (2-fold dilution) and analyzed by high sensitivity flow cytometry to determine their concentration (D), the CMFDA-height (H) mean of fluorescence (E) and the CMFDA-H median of fluorescence (F) are presented. Data are mean ± SEM of 5 independent experiments. BKD = Background noise.</p

    Impact of sPLA<sub>2</sub>s on MPs from the male reproductive tract.

    No full text
    <p>(A) Human epididymosomes were incubated 6 hours at 37°c in absence or in presence of 1µg/ml of human recombinant sPLA<sub>2</sub> IIA, V, X. Fluorochrome-conjugated annexin-V was used to assess the quantities of annexin-V<sup>+</sup> MPs and were compared to the untreated conditions (dotted line). Data are mean ± SEM of 3 independent experiments presented as % of untreated (control). (B) Mouse epididymosomes were incubated 6 hours at 37°c in absence or in presence of 1µg/ml of mouse recombinant sPLA<sub>2</sub> IIA, V, X. Fluorochrome-conjugated annexin-V was used to assess the quantities of annexin-V<sup>+</sup> MPs and were compared to the untreated conditions (dotted line). Data are mean ± SEM of 4 independent experiments presented as % of untreated (control). (C) Human prostasomes were incubated 6 hours at 37°c in absence or in presence of 1µg/ml of human recombinant sPLA<sub>2</sub> IIA, V, X. Fluorochrome-conjugated antibodies directed against CD13 and fluorochrome-conjugated annexin-V were used to determine the concentrations of CD13<sup>+</sup> MPs (left panel), of annexin-V<sup>+</sup> MPs (right panel) and were compared to the untreated conditions (dotted line). Data are mean ± SEM of 4 independent experiments presented as % of untreated (control) * P< .05; # P< .01; § P< .001.</p

    Impact of human and mouse sPLA<sub>2</sub>s on endothelial cell MPs.

    No full text
    <p>(A) MPs from HUVEC labeled with the CMFDA cell tracker were incubated for 1 and 6 hours at 37°c in absence or in presence of indicated concentrations of human recombinant sPLA<sub>2</sub> IIA, V, X, or 1µg/ml of the inactive mutant V H48Q. Fluorochrome-conjugated antibodies directed against CD31 and fluorochrome-conjugated annexin-V were used to assess the quantities of CMFDA<sup>+</sup> MPs (left panel), of CD31<sup>+</sup> MPs (middle panel), of annexin-V<sup>+</sup> MPs (right panel) and were compared to the untreated conditions (dotted line). Data are mean ± SEM of 5 independent experiments presented as % of untreated (control) (B) MPs from mouse EOMA cells labeled with the CMFDA cell tracker were incubated 1 and 6 hours at 37°c, in absence or in presence of indicated concentrations of mouse recombinant sPLA<sub>2</sub> IIA, V, X, or 1µg/ml of the inactive mutant X H48Q. Fluorochrome-conjugated antibodies directed against CD31 and fluorochrome-conjugated annexin-V were used to determine the concentrations of CMFDA<sup>+</sup> MPs (left panel), of CD31<sup>+</sup> MPs (middle panel), of annexin-V<sup>+</sup> MPs (right panel) and then compared to the untreated conditions (dotted line). Data are mean ± SEM of 5 independent experiments presented as % of untreated (control). # P< .01; § P< .001.</p

    Optimization of flow cytometric methods for the detection of MPs.

    No full text
    <p>(A, B) Acquisition of fluorescent microspheres of 100nm (Blue), 450nm (pink), 840nm (green), 1000nm (red), 3200nm (orange) in diameter on a flow cytometer Canto II modified with a FSC-PMT small particles option. (B) A MP gate including particles from 100 to 1000nm in diameter based on the microsphere sizes (FSC-PMT-H) is presented and used to detect MPs. (C) Portrayal of relative size of human platelets detected with fluorochrome-conjugated antibodies directed against CD41. (D) FSC-PMT/SSC portrayal of platelet MPs detected with annexin-V and fluorochrome-conjugated antibodies directed against CD41 in absence of treatment (control). (E) A known concentration of auto-fluorescent polystyrene microspheres (15 µm in diameter) was added in each tube and a determined number of beads was acquired in the counting bead gate to quantitatively process the data. (F, G) FSC-PMT/SSC portrayal of platelet MPs detected with annexin-V and fluorochrome-conjugated antibodies directed against CD41 and treated with 0.05% triton (F) and 50µM EDTA (G). Total annexin-V<sup>+</sup> events are detected in the pink gate (middle panel) and the quantity of annexin-V<sup>+</sup> MPs is determined in the Annexin-V MP gate (upper panel). Total CD41<sup>+</sup> events are detected in the blue gate (middle panel) and the quantity of CD41<sup>+</sup> MPs is determined in the CD41 MP gate (lower panel). Data are representative of 5 independent experiments. (H) Triton sensitivity of the platelet MPs detected using fluorochrome-conjugated annexin-V (left panel) and fluorochrome-conjugated antibodies directed against CD41 (right panel) is presented as % of untreated (control). (I) EDTA sensitivity of annexin-V (left panel) and CD41 (right panel) labeling is presented as % of untreated (control). Data are representative of 5 independent experiments.</p

    Platelets release mitochondrial antigens in systemic lupus erythematosus

    No full text
    The accumulation of DNA and nuclear components in blood and their recognition by autoantibodies play a central role in the pathophysiology of systemic lupus erythematosus (SLE). Despite the efforts, the sources of circulating autoantigens in SLE are still unclear. Here, we show that in SLE, platelets release mitochondrial DNA, the majority of which is associated with the extracellular mitochondrial organelle. Mitochondrial release in patients with SLE correlates with platelet degranulation. This process requires the stimulation of platelet Fc gamma RIIA, a receptor for immune complexes. Because mice lack Fc gamma RIIA and murine platelets are completely devoid of receptor capable of binding IgG-containing immune complexes, we used transgenic mice expressing Fc gamma RIIA for our in vivo investigations. Fc gamma RIIA expression in lupus-prone mice led to the recruitment of platelets in kidneys and to the release of mitochondria in vivo. Using a reporter mouse with red fluorescent protein targeted to the mitochondrion, we confirmed platelets as a source of extracellular mitochondria driven by Fc gamma RIIA and its cosignaling by the fibrinogen receptor alpha 2b beta 3 in vivo. These findings suggest that platelets might be a key source of mitochondrial antigens in SLE and might be a therapeutic target for treating SLE

    Biogenesis and function of tRNA fragments during sperm maturation and fertilization in mammals

    No full text
    Several recent studies link parental environments to phenotypes in subsequent generations. In this work, we investigate the mechanism by which paternal diet affects offspring metabolism. Protein restriction in mice affects small RNA (sRNA) levels in mature sperm, with decreased let-7 levels and increased amounts of 5\u27 fragments of glycine transfer RNAs (tRNAs). In testicular sperm, tRNA fragments are scarce but increase in abundance as sperm mature in the epididymis. Epididymosomes (vesicles that fuse with sperm during epididymal transit) carry RNA payloads matching those of mature sperm and can deliver RNAs to immature sperm in vitro. Functionally, tRNA-glycine-GCC fragments repress genes associated with the endogenous retroelement MERVL, in both embryonic stem cells and embryos. Our results shed light on sRNA biogenesis and its dietary regulation during posttesticular sperm maturation, and they also link tRNA fragments to regulation of endogenous retroelements active in the preimplantation embryo
    corecore