13 research outputs found

    Elucidating the Niche Microenvironments of Dormant and Metastatic Breast Cancers

    Get PDF
    https://openworks.mdanderson.org/sumexp21/1011/thumbnail.jp

    Semaphorins as Regulators of Phenotypic Plasticity and Functional Reprogramming of Cancer Cells

    No full text
    Semaphorins, initially found as neuronal guidance cues in embryo development, are now appreciated as major regulators of tissue morphogenesis and homeostasis, as well as of cancer progression. In fact, semaphorin signals have a profound impact on cell morphology, which has been commonly associated with the ability to regulate monomeric GTPases, cell-substrate adhesion, and cytoskeletal dynamics. Recently, however, several reports have indicated a novel and additional function of diverse semaphorins in the regulation of gene expression and cell phenotype plasticity. In this review article, we discuss these novel findings, focusing on the role of semaphorin signals in the regulation of bi-directional epithelial-mesenchymal transitions, stem cell properties, and drug resistance, which greatly contribute to the pathogenesis of cancer

    Transmembrane semaphorins: Multimodal signaling cues in development and cancer

    No full text
    Semaphorins constitute a large family of membrane-bound and secreted proteins that provide guidance cues for axon pathfinding and cell migration. Although initially discovered as repelling cues for axons in nervous system, they have been found to regulate cell adhesion and motility, angiogenesis, immune function and tumor progression. Notably, semaphorins are bifunctional cues and for instance can mediate both repulsive and attractive functions in different contexts. While many studies focused so far on the function of secreted family members, class 1 semaphorins in invertebrates and class 4, 5 and 6 in vertebrate species comprise around 14 transmembrane semaphorin molecules with emerging functional relevance. These can signal in juxtacrine, paracrine and autocrine fashion, hence mediating long and short range repulsive and attractive guidance cues which have a profound impact on cellular morphology and functions. Importantly, transmembrane semaphorins are capable of bidirectional signaling, acting both in \u201cforward\u201d mode via plexins (sometimes in association with receptor tyrosine kinases), and in \u201creverse\u201d manner through their cytoplasmic domains. In this review, we will survey known molecular mechanisms underlying the functions of transmembrane semaphorins in development and cancer

    Sema4C/PlexinB2 signaling controls breast cancer cell growth, hormonal dependence and tumorigenic potential

    No full text
    Semaphorin 4C (Sema4C) expression in human breast cancers correlates with poor disease outcome. Surprisingly, upon knock-down of Sema4C or its receptor PlexinB2 in diverse mammary carcinoma cells (but not their normal counterparts), we observed dramatic growth inhibition associated with impairment of G2/M phase transition, cytokinesis defects and the onset of cell senescence. Mechanistically, we demonstrated a Sema4C/PlexinB2/LARG-dependent signaling cascade that is required to maintain critical RhoA-GTP levels in cancer cells. Interestingly, we also found that Sema4C upregulation in luminal-type breast cancer cells drives a dramatic phenotypic change, with disassembly of polarity complexes, mitotic spindle misorientation, cell\u2013cell dissociation and increased migration and invasiveness. We found that this signaling cascade is dependent on the PlexinB2 effectors ErbB2 and RhoA-dependent kinases. Moreover, Sema4C-overexpressing luminal breast cancer cells upregulated the transcription factors Snail, Slug and SOX-2, and formed estrogen-independent metastatic tumors in mice. In sum, our data indicate that Sema4C/PlexinB2 signaling is essential for the growth of breast carcinoma cells, featuring a novel potential therapeutic target. In addition, elevated Sema4C expression enables indolent luminal-type tumors to become resistant to estrogen deprivation, invasive and metastatic in vivo, which could account for its association with a subset of human breast cancers with poor prognosis

    PlexinD1 Is a Novel Transcriptional Target and Effector of Notch Signaling in Cancer Cells

    Get PDF
    <div><p>The secreted semaphorin Sema3E controls cell migration and invasiveness in cancer cells. Sema3E-receptor, PlexinD1, is frequently upregulated in melanoma, breast, colon, ovarian and prostate cancers; however, the mechanisms underlying PlexinD1 upregulation and the downstream events elicited in tumor cells are still unclear. Here we show that the canonical RBPjk-dependent Notch signaling cascade controls PlexinD1 expression in primary endothelial and cancer cells. Transcriptional activation was studied by quantitative PCR and promoter activity reporter assays. We found that Notch ligands and constitutively activated intracellular forms of Notch receptors upregulated PlexinD1 expression; conversely RNAi-based knock-down, or pharmacological inhibition of Notch signaling by gamma-secretase inhibitors, downregulated PlexinD1 levels. Notably, both Notch1 and Notch3 expression positively correlates with PlexinD1 levels in prostate cancer, as well as in other tumor types. In prostate cancer cells, Sema3E-PlexinD1 axis was previously reported to regulate migration; however, implicated mechanisms were not elucidated. Here we show that in these cells PlexinD1 activity induces the expression of the transcription factor Slug, downregulates E-cadherin levels and enhances cell migration. Moreover, our mechanistic data identify PlexinD1 as a pivotal mediator of this signaling axis downstream of Notch in prostate cancer cells. In fact, on one hand, PlexinD1 is required to mediate cell migration and E-cadherin regulation elicited by Notch. On the other hand, PlexinD1 upregulation is sufficient to induce prostate cancer cell migration and metastatic potential in mice, leading to functional rescue in the absence of Notch. In sum, our work identifies PlexinD1 as a novel transcriptional target induced by Notch signaling, and reveals its role promoting prostate cancer cell migration and downregulating E-cadherin levels in Slug-dependent manner. Collectively, these findings suggest that Notch-PlexinD1 signaling axis may be targeted to impair prostate cancer cell invasiveness and metastasis.</p></div

    Notch1, Notch3 and PlexinD1 regulate EMT markers in prostate cancer cells.

    No full text
    <p>(A) PC3 were transiently transfected with GFP, N1-ICD, N3-ICD or PlexinD1, and the mRNA levels of Snail, Slug, Zeb1, Zeb2, Twist1 were analyzed by qPCR (48 hours later). (B) PC3 cells were transfected with GFP, Jag1-Fc, or Sema3E-p61; Slug mRNA levels were analyzed, as above. (C) Slug mRNA levels were assessed in PC3 cells stably expressing pLKO or silenced for Notch1 and PlexinD1. (D) PC3 transiently transfected with pLKO, N1-ICD and PlexinD1 were analyzed for E-cadherin mRNA levels by qPCR. (E) PC3 cells were transiently transfected with GFP, N1-ICD or N3-ICD; after 48 hrs lysates were analyzed by immunoblotting to reveal E-cadherin levels. (F, G) PC3 and DU145 cells transfected with PlexinD1 were analyzed by immunoblotting to reveal E-cadherin levels. (H, I) PC3 and DU145 cells, respectively, stably expressing shScr, shPlexinD1 and shNotch1 were analyzed by immunoblotting to reveal E-cadherin levels. (J) PC3 cells were treated with DMSO, DAPT (25μM) or RO4929097 (25μM) for 72 hrs and protein lysates were analyzed to reveal E-cadherin levels. (K, L) PC3 cells stably expressing shScr or shSema3E were analyzed by western blotting (K) or qPCR (L) to reveal E-cadherin levels. (M) Analysis of E cadherin levels by immunoblotting in PC3 cells stably silenced for Sema3E, and upon re-expression of Sema3E-p61 (S3E-p61). (N) E cadherin and Slug expression levels assessed by qPCR upon overexpression of Sema3E-p61 in control and Slug-depleted cells (by siRNA). (O) The migration of PC3 cells treated as in previous panels was assessed in Boyden Chamber assays. (P) Immunoblotting analysis of E cadherin levels in PC3 cells transfected to overexpress PlexinD1 (or mock), with or without treatment with MAPK inhibitor PD98059 (10μM). Bar graphs display mean values ± SD.</p

    PlexinD1 acts downstream of Notch signaling in down-regulating E-cadherin and promoting cancer cell migration.

    No full text
    <p>(A, B) PC3 cells were transiently transfected with either shScr, N1-ICD, shPlexinD1, or a combination of N1-ICD and shPlexinD1. 72 hrs after transfection, cell migration was analysed by Boyden chamber assays (A). Moreover, cell lysates were analyzed by immunoblotting to reveal E-cadherin, Notch1-ICD and vinculin levels (B); relative E-cadherin band intensity to vinculin levels was quantified and normalized to controls. (C, D) PC3 cells were transiently transfected with either shScr, shNotch1, PlexinD1, or a combination of shNotch1 and PlexinD1. 72 hrs after transfection, cell migration was analysed by Boyden chamber assays (C). Moreover, cell lysates were analyzed by immunoblotting to reveal E-cadherin, N1-ICD, PlexinD1 and vinculin levels (D) and relative E-cadherin band intensity to vinculin levels was quantified and normalized to controls. (E, F) PC3 cells transfected as in C-D were analyzed in vivo by metastatic extravasation assay upon tail vein injection in mice. Representative images are shown (E) along with fluorescence intensity quantification (F). Bar graphs show mean values ± SD (normalized to control).</p

    Notch and PlexinD1 signaling regulate prostate cancer cell migration.

    No full text
    <p>(A) Wound healing assay in PC3 cells treated with gamma secretase inhibitors—DAPT and RO4929097 (as in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0164660#pone.0164660.g003" target="_blank">Fig 3C</a>). Wound closure (24 hours from scratch) was quantified relative to wound width at start time. (B, C) The migration of PC3 cells stably expressing shScr, shNotch1, shPlexinD1 was analyzed in wound healing assays, as above. Panel B shows the quantification of wound sizes at the end of the experiment. (D) The migration of PC3 cells stably expressing shScr, shPlexinD1 and shNotch1 was assayed in overnight Boyden chamber experiments with transwell inserts. Bar graphs indicate mean values ± SD (normalized to controls). (E) PC3 cells overexpressing Dll1-Fc and Jag1-Fc were analyzed in Boyden chamber experiment, as above. (F) PC3 cells transduced to express an autocrine p61-Sema3E circuit were analyzed in Boyden chamber experiments, as above. (G) PC3 cells stably expressing shSema3E compared to shScr were analyzed in Boyden chamber experiments, as above. Mean ± SD is shown in all graphs; values were normalized to respective controls.</p

    Notch signaling upregulates PlexinD1 promoter activity.

    No full text
    <p>(A) Schematic of the reporter construct pEZX-PGO2, containing a 1567 bp region from <i>PLXND1</i> gene promoter (position 1 being transcription start) fused to <i>Gaussia</i> luciferase reporter cassette, and of two mutant constructs with deletions of RBPjk binding site (positioned at -1382 bp upstream transcription start). (B) <i>PLXND1</i> promoter-induced <i>Gaussia</i> luciferase activity was revealed in cell-conditioned media of 293T cells co-transfected with mock plasmid, or with N1-ICD (indicated amounts), in combination with the reporter construct. (C) 293T cells were transiently co-transfected with either mock plasmid, N1-ICD or N3-ICD in combination with PlexinD1 promoter reporter construct. (D) Luciferase reporter activity was tested in 293T cells transiently transfected with N1-ICD (or mock), in association with <i>PLXND1</i> promoter region (Long D1) or with mutated promoter constructs lacking RBPjk binding sites (Mut1_D1 and Mut2_D1, described in panel (A). (E) 293T cells were transiently transfected with the indicated amounts of RBPjk cDNA; cell lysates were analyzed by immunoblotting for PlexinD1 and vinculin; relative band intensity was quantified and normalized to control. (F) 293T cells were transiently co-transfected with wild-type RBPjk or dominant-negative RBPjk (or mock) in combination with <i>PLXND1</i> promoter reporter construct, and luciferase activity was revealed in cell-conditioned media, as in (B). Bar graphs indicate mean values (normalized to controls) ± normalized SD.</p

    PlexinD1 is a putative Notch signaling target.

    No full text
    <p>(A, B) HEK-293T cells were transiently transfected with a constitutively activated Notch-intracellular domain construct (N1-ICD) or with GFP expressing construct. PlexinD1 mRNA levels were measured by qPCR after 72 hrs (A), and protein lysates were analyzed by immunoblotting for PlexinD1 and vinculin (B). (C) HEK-293T cells were transiently transfected with Notch1 ΔE-ICD and cell lysates were immunoblotted for PlexinD1 and vinculin, as above. (D, E) HUVEC endothelial cells were transduced to stably express Notch1-targeted shRNAs (shNotch1); mRNA levels were analyzed by qPCR (D), and protein lysates by immunoblotting (E; relative PlexinD1 band intensity to vinculin levels was quantified and normalized to controls), as above. (F) HUVEC cells were treated with gamma-secretase inhibitor DAPT (25μM) to block Notch signaling, or DMSO (vehicle), for 72 hrs; lysates were analyzed by immunoblotting for PlexinD1 and vinculin (relative PlexinD1 band intensity to vinculin levels was quantified and normalized to controls). (G, H) HEK-293T cells were transiently transfected with activated Notch3 intracellular domain (N3-ICD); after 72hrs, mRNA levels (G) and protein lysates (H) were analyzed, as above. (I, J) PC3 prostate cancer cells transiently transfected with N1-ICD or N3-ICD were analyzed for expression of PlexinD1 by qPCR (I) or western blotting (J). (K) DU145 prostate cancer cells transiently transfected with N1-ICD and N3-ICD were analyzed by immunoblotting for PlexinD1 and vinculin, as above. Mean values ± SD are shown in all graphs; relative gene expression levels were normalized to controls.</p
    corecore