16 research outputs found

    Notch1-expressing cells contribute to alveologenesis at pregnancy and expand in response to hormonal stimulation.

    No full text
    <p><b>(A)</b> N1Cre<sup>ERT2</sup>R26<sup>mTmG</sup> females were induced with a low dose of tamoxifen (1µg/g of mouse body weight) at 4 wk of age and analyzed as virgins (left) or at mid-pregnancy (14.5 d post coitum, dpc) (right panel). Representative pictures of whole mount mammary gland ducts are shown, where red denotes tomato fluorescence, while green indicates Notch1-derived GFP<sup>pos</sup> cells. <i>n</i> = 3. <b>(B)</b> Quantification by flow cytometry of the percentage of GFP<sup>pos</sup> luminal cells (Lin<sup>-</sup>CD24<sup>+</sup>CD29<sup>low</sup>) from N1Cre<sup>ERT2</sup>R26<sup>mTmG</sup> females induced at 4 wk of age (0.1mg/g of mouse body weight) and analyzed at virgin or pregnant state (14.5 dpc). An extensive increase in the amount of GFP<sup>pos</sup> cells is evident at pregnancy in A and B. <i>n</i> = 7. (**) <i>p</i> = 0.001 with <i>t</i> test. <b>(C)</b> Left, representative mammary section of N1Cre<sup>ERT2</sup>R26<sup>mTmG</sup> females injected with tamoxifen at mid-pregnancy (14.5 dpc) and analyzed 24 h later (pulse). Right, mammary section of N1Cre<sup>ERT2</sup>R26<sup>mTmG</sup> female injected at 4 wk of age and analyzed at mid-pregnancy (chase). <b>(D)</b> Quantification by flow cytometry of the percentage of GFP<sup>pos</sup> cells luminal cells (Lin<sup>-</sup>CD24<sup>+</sup>CD29<sup>low</sup>) from N1Cre<sup>ERT2</sup>R26<sup>mTmG</sup> females injected at mid-pregnancy (pulse: 69.75 ±3.55%) compared to females injected at 4 wk of age and analyzed at mid-pregnancy (chase: 86.1 ±2.01%). Values are shown as percentages ±s.e.m. of <i>n</i> = 5. <b>(E)</b> 3-D organotypic cultures from adult N1Cre<sup>ERT2</sup>R26<sup>mTmG</sup> females were induced with 4-hydroxytamoxifen (4-OHT) in vitro and treated for 10 d with different hormones, as indicated. Red denotes tomato fluorescence and green indicates Notch1-marked cells at Day 0 and progeny at Day 10; <i>n</i> = 4. <b>(F)</b> Quantification of the clonal expansion of GFP<sup>pos</sup> cells in 3-D organoids, classified as single cells (one cell, in light green) or clones (two or more cells, in dark green) indicates a significant expansion only in response to estradiol (β-2E bars). (***) <i>p</i> < 0.001 with <i>t</i> test. Scale bars correspond to 40 µm in A and E and 20 µm in C.</p

    Notch1 is expressed in multipotent embryonic stem cells.

    No full text
    <p><b>(A)</b> Schematic representation of the Notch1-CreERT2<sup>SAT</sup> knock-in mice (referred to as N1Cre<sup>ERT2</sup>) and Rosa26<sup>mTomato/mGFP</sup> reporter mice (called R26<sup>mTmG</sup>) used in this study. Pregnant females were induced with tamoxifen to label their embryos at embryonic day E15.5 and double transgenic N1Cre<sup>ERT2</sup>R26<sup>mTmG</sup> littermates were analyzed 24 h later <b>(B–D)</b> or 5 wk after birth (<b>E–G</b>). <b>(B–D)</b> Representative embryonic mammary bud sections show that Notch1<sup>pos</sup> cells (marked by GFP in green) express both myoepithelial (K5, in red in B) and luminal markers (K8, in red in C) and they are negative for ERα (in red in D), <i>n</i> = 2. (<b>E–G</b>) Representative pubertal mammary gland sections show that Notch1-derived clones (in green) contain myoepithelial (K5<sup>pos</sup> in red in E) and luminal cells (K8<sup>pos</sup> in red in F) as well as ERα<sup>pos</sup> and ERα<sup>neg</sup> cells (ERα in red in G), <i>n</i> = 3. 4',6-diamidino-2-phenylindole (DAPI) stains DNA in blue. Scale bars correspond to 20 µm in B–G, and 10 µm in the magnifications of the insets.</p

    The transcriptional signature of ERα<sup>neg</sup> mammary luminal progenitors is conserved in their derived lineages.

    No full text
    <p><b>(A)</b> Genome-wide heat map showing the pattern of GFP<sup>neg</sup> and GFP<sup>pos</sup> luminal cells (Lin<sup>-</sup>CD24<sup>+</sup>CD29<sup>low</sup>) from 10-wk-old N1Cre<sup>ERT2</sup>R26<sup>mTmG</sup> females induced for 24 h. Biological triplicates for each group were performed by pooling two females per experiment and are represented by different columns in the unsupervised clustering. <b>(B)</b> GSEA analysis showing the enrichment plots for GFP<sup>neg</sup> and GFP<sup>pos</sup> transcriptome profiles in four published gene signatures (Luminal_Mature_Up, Luminal_Mature_Down, Luminal_Progenitor_Up, Luminal_ Progenitor_Down). Sorted GFP<sup>neg</sup> cells correlate with luminal mature cells, while sorted GFP<sup>pos</sup> cells correspond to luminal progenitors. ES: Enrichment Score. <b>(C)</b> qRT-PCR analysis, in adult N1Cre<sup>ERT2</sup>R26<sup>mTmG</sup> females induced for 24 h, of the top-ten ranked genes in the transcriptomic analysis (first row: up-regulated in GFP<sup>pos</sup> and second row: down-regulated in GFP<sup>pos</sup>) confirms the differential expression of each of these genes detected in the microarray experiments. The expression levels of each gene in sorted myoepithelial cells are also shown (Myo). Relative mRNA expression was normalized to the housekeeping gene 18S. (*) <i>p</i> < 0.05, (**) <i>p</i> < 0.01, (***) <i>p</i> < 0.001 with <i>t</i> test. <i>n</i> = 2. <b>(D)</b> FACS analysis of sorted cells from N1Cre<sup>ERT2</sup>R26<sup>mTmG</sup> females induced at 4 wk of age and analyzed 10 wk later (<i>n</i> = 8) shows that GFP<sup>pos</sup> cells (Notch1-derived progeny, in green) overlap with ER<sup>neg</sup> luminal progenitor markers (CD49b<sup>pos</sup>, Sca1<sup>neg</sup> and CD133<sup>neg</sup> in blue) gated as Lin<sup>-</sup>CD24<sup>+</sup>CD29<sup>low</sup> luminal cells, showing an identical profile as Notch1-expressing cells 24 h post-induction (see <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002069#pbio.1002069.g004" target="_blank">Fig. 4D</a>).</p

    Notch1-expressing cells reveal a high regenerative capacity in transplantation assays.

    No full text
    <p><b>(A)</b> 10,000 GFP<sup>pos</sup> luminal cells were sorted by FACS within the CD24<sup>+</sup>/CD29<sup>low</sup> gate, mixed 1:1 with Tomato epithelial cells (Lin<sup>neg</sup>), and injected in cleared fat pads of host mice. All transplanted animals showed outgrowths (<i>n</i> = 2). GFP<sup>pos</sup> cells generated luminal cells but not myoepithelial cells, stained with anti-K5 antibody (in red in A). (<b>B–C</b>) Whole-mounts of GFP<sup>pos</sup> outgrowths obtained in virgin host mice (<i>n</i> = 1 out of 7 transplanted, in B) and pregnant host females (<i>n</i> = 7 out of 11 transplanted, in C). On the right of each panel, graphic representations of the extent to which each transplant filled the fat pad (in black). <b>(D–F)</b> 10,000 sorted luminal GFP<sup>pos</sup> (Notch1-expressing cells) cells were injected per fat pad, in the absence of myoepithelial cells. Immunofluorescence of representative sections of obtained outgrowths show that GFP<sup>pos</sup> cells generate all mammary lineages, both in virgin (D–E) and in pregnant recipient mice (F). Immunofluorescence labeling of myoepithelial cells (anti-K5 in red in D), and ERα<sup>pos</sup> (in red in E and F) show that GFP-derived clones contain K5<sup>pos</sup> myoepithelial cells, ERα<sup>pos</sup> and ERα<sup>neg</sup> luminal cells. Scale bars correspond to 10 µm in A, 2 mm in B–C, and 20 µm in D–F.</p

    Proposed model for luminal cell hierarchy during mammary gland development.

    No full text
    <p><b>(A)</b> Lineage tracing from embryos reveals that Notch1-expressing (N1<sup>pos</sup>) multipotent stem cells exist only during embryonic mammary development, when they co-express myoepithelial cytokeratin (K5) and luminal cytokeratin (K8). These multipotent stem cells can generate all mammary lineages (labeled in green postnatally). <b>(B)</b> After birth, luminal stem cells resolve in two distinct luminal progenitors: ERα<sup>pos</sup> and ERα<sup>neg</sup> cells, which maintain exclusively their own lineage throughout adulthood. Unipotent progenitors lacking ERα expression (ERα<sup>neg</sup>) marked by Notch1 are responsible for generating milk-producing alveoli at pregnancy and define self-renewing luminal cells able to survive mammary gland involution.</p

    Notch1-expressing cells define highly clonogenic ERα<sup>neg</sup> luminal progenitors.

    No full text
    <p><b>(A)</b> N1Cre<sup>ERT2</sup>R26<sup>mTmG</sup> females induced with tamoxifen at puberty (6 wk of age) and analyzed 24 h later present a high proportion of proliferative GFP<sup>pos</sup> cells (73.45 ±3.72%), as indicated by Ki67 expression (in red); DAPI stains DNA in blue, <i>n</i> = 5. <b>(B)</b> Left, histogram of the cell cycle profile of total luminal cells (Luminal, in blue) compared to Notch1-expressing cells (GFP<sup>pos</sup>, in green) sorted by FACS and stained with Hoechst-33342 to evaluate their DNA content. Right, quantification of cycling (S/G2/M) and non-cycling (G<sub>0</sub>/G<sub>1</sub>) cells in total luminal cells (blue, S/G2/M = 26.9 ±5.5%) and in Notch1-expressing cells (green, S/G2/M = 47.0 ±5.2%). Data are represented as a mean ± s.e.m. of <i>n</i> = 5 animals, (*) <i>p</i> < 0.05 with <i>t</i> test. <b>(C)</b> N1Cre<sup>ERT2</sup>R26<sup>mTmG</sup> females were induced with tamoxifen at 10 wk of age and analyzed 24 h later. GFP<sup>neg</sup> and GFP<sup>pos</sup> cells were sorted as Lin<sup>-</sup>CD24<sup>+</sup>CD29<sup>low</sup> luminal cells by FACS, seeded on a feeder layer of irradiated fibroblast and their clonogenic capacity was evaluated after 7 d in culture. Top, quantification of the number of colonies generated by GFP<sup>neg</sup> and GFP<sup>pos</sup> cells. <i>n</i> = 3 experiments with two mice each, (***) <i>p</i> < 0.001 with <i>t</i> test. Bottom, representative pictures of counted colonies stained with Hematoxylin are shown below each bar. <b>(D)</b> Flow cytometry analysis indicates that GFP<sup>pos</sup> cells (in green) are found within progenitor cell populations (CD49b<sup>pos</sup>, Sca1<sup>neg</sup> and CD133<sup>neg</sup>) gated in the luminal population (Lin<sup>-</sup>CD24<sup>+</sup>CD29<sup>low</sup>) (in blue). GFP<sup>pos</sup> cells represent 82.02 ±3.28% of CD49b<sup>pos</sup> cells, 96.9 ±0.6% of Sca1<sup>neg</sup> cells, and 93.7 ±1.0% of CD133<sup>neg</sup> cells. <i>n</i> = 10. Scale bars correspond to 20 µm in A and 10 µm in the insets in A.</p

    ERα<sup>neg</sup> luminal cells represent self-sustained mammary progenitors that clonally expand and self-renew in vivo.

    No full text
    <p><b>(A–B)</b> N1Cre<sup>ERT2</sup>R26<sup>mTmG</sup> females induced with a low dose of tamoxifen (1µg/g of mouse body weight) at 4 wk of age and analyzed at different time points: 24 h (pulse), 2 wk (2 wk chase), 6 wk (6 wk chase), or 10 wk later (10 wk chase). <b>(A)</b> Representative images of whole mount digestion of mammary ducts labeling single cells (one cell), small clones (two to five cells), and large clones (six or more cells). Notch1-labelled cells are in green and Tomato fluorescence labels non-targeted cells. <i>n</i> = 11 mice. <b>(B)</b> Quantification of the in vivo clonal expansion of GFP-marked cells, at the indicated time points; <i>n</i> = 11. <b>(C–E)</b> N1Cre<sup>ERT2</sup>R26<sup>mTmG</sup> females were induced with a single dose of tamoxifen (0.1mg/g of mouse body weight) at 4 wk of age and analyzed 10 wk later. The clonal progeny of Notch1-expressing cells (marked in green by GFP) is still luminal (K5<sup>neg</sup> in red in C), and ERα<sup>neg</sup> (in red in D) and PR<sup>neg</sup> (in red in E), as the cells from which it derives. DAPI stains DNA in blue, <i>n</i> = 4. <b>(F)</b> FACS plots of N1Cre<sup>ERT2</sup>R26<sup>mTmG</sup> females induced at puberty and analyzed after three cycles of pregnancy, lactation, and involution. Mammary epithelial cells (MEC) from females in the third week of the last involution were dissociated and analyzed by flow cytometry. GFP<sup>pos</sup> cells represent 28.9 ±1.4% of the luminal population (CD24<sup>+</sup>and CD29<sup>low</sup>), <i>n</i> = 2. (<b>G–H</b>) Representative sections of mammary ducts from N1Cre<sup>ERT2</sup>R26<sup>mTmG</sup> females induced at 4 wk of age and analyzed after three serial pregnancies. GFP-expressing cells (in green) remain invariably negative for ERα (in red in G) and PR expression (in red in H). DAPI stains DNA in blue, <i>n</i> = 2. Scale bars correspond to 40 µm in A; 20 µm in C–E, G and H; and 10 µm in the insets in C–E and G–H.</p

    Depletion of xlPTP4A3 inhibits both migration of NCC and uveal melanoma cells.

    No full text
    <p>A) Isotopic and isochronic grafts of xlPTP4A3-depleted NC explants into host embryos showed a delayed NCC migration (b-b’) when compared to the control histone 2b-GFP NC explants (a-a’). B) Timelapse videomicroscopy of stage 17-derived NCC treated with a pharmacological inhibitor of PTP4A3, a rhodanine derivative (PRL-3 inhibitor I), shows that inhibition of PTP4A3 increases the average of pausing NC cells and strongly decreases the migration velocity of the treated cells (relative to that of vehicle treated cells). The result is representative of three independent experiments and the average of counted cells is 60 cells per condition (***p<0,001). C) Timelapse videomicroscopy of uveal melanoma OCM1 cells stably expressing EGFP-PTP4A3, EGFP-PTP4A3(C104S) or EGFP on collagen I shows that inhibition of PTP4A3, using PRL-3 Inhibitor I, decreases specifically the migration velocity of the PTP4A3 expressing cells (relative to the velocity of untreated and control cells). The result is representative of three independent experiments and the average of counted cells is 50 cells per condition (***p<0,001).</p

    xlPTP4A3 gain of function induces strengthening of the anterior NC migration streams <i>in</i><i>vivo</i>.

    No full text
    <p>xlPTP4A3, mmPTP4A3 or the mutant form mmPTP4A3(C104S) were injected at the four-cell stage in the animal pole region, and embryos were cultured until stage 22. A) Overexpression of xlPTP4A3 induces a strengthening (b, d <i>vs</i>. d’) and/or lengthening (a, c) of the anterior NC migration streams as showed by whole-mount <i>in </i><i>situ</i> hybridization on xlTWIST. B) xlPTP4A3 corresponding phenotype quantification was determined as the area of the NC migratory streams showing that the migratory streams surface is greater in the injected side than in the uninjected side (n=15) (**p<0,01). C) Overexpression of mmPTP4A3 causes a strengthening and/or lengthening (e <i>vs</i>.e’, f) of the anterior NC migration streams while overexpression the mutant form mmPTP4A3(C104S) does not seem to significantly affect the migration of the NC (g <i>vs</i>.g’, h). D) mmPTP4A3 or mmPTP4A3(C104S) corresponding phenotype quantification was determined as the area of the NC migratory streams showing that mmPTP4A3 injected embryos display a greater anterior NC area in the injected side than in the uninjected side while no difference in the anterior NC area could be observed in the mmPTP4A3(C104S) injected embryos between the two sides (n=15) (**p<0,01). E) Immunodetection of phosphoHistone 3 (Ser10) on xlPTP4A3 gain of function injected embryos. Injection of xlPTP4A3 does not seem to affect cell proliferation in the injected side relative to that of the uninjected side (a <i>vs</i>.b). </p

    Depletion of xlPTP4A3 reduces NCC territory.

    No full text
    <p>Control 5MM-MO or xlPTP4A3-MO were injected at the four-cell stage in the animal pole region and embryos were cultured until stage 22. A) Morpholino sequence targeting xlPTP4A3. B) xlPTP4A3-MO specifically knocked down the translation of xlPTP4A3 <i>in </i><i>vitro</i> using the TNT® Coupled Reticulocyte Lysate System. C) xlPTP4A3 depleted embryos showed blockade of NCC territory (a-a’) and can be rescued by co-injection of mouse mPTP4A3 (c-c’), as shown by whole-mount <i>in </i><i>situ</i> hybridization on xlTWIST. The injected side was monitored using the co-injection of nuclear-targeted β-galactosidase (red dotted signal). Injection of the control 5MM-MO does not affect the NCCs territory, as in the control noninjected embryos (b-b’). D) Quantitative results of relative phenotype (**p<0,01). E) The phenotype quantification was determined as the length of the NC migratory streams showing that the migratory streams are shorter in the injected side than in the uninjected side (n=15) (***p<0,001). F) Immunodetection of cleaved caspase 3 on xlPTP4A3-MO injected embryos. Two phenotypes are observed: one of weaker apoptosis in the injected side compared to the uninjected side (a-a’) and one of no differential apoptosis between the two sides (b-b’).</p
    corecore