11 research outputs found

    Efficient Human Cytomegalovirus Replication in Primary Endothelial Cells Is SOCS3 Dependent

    Full text link
    Background: In immunocompromised patients, human cytomegalovirus (HCMV) infection is a major cause of morbidity and mortality. Suppressor of cytokine signaling (SOCS) proteins are very potent negative regulators of the janus kinase/signal transducer and activator of transcription (JAK/STAT) pathways. We hypothesized that HCMV exploits SOCS1 and/or SOCS3 to its advantage. Methods: All experiments were carried out with primary human lung-derived microvascular endothelial cells (HMVEC). SOCS1 and SOCS3 were silenced by transfecting the cells with siRNA. HCMV was propagated and titered on human lung-derived fibroblasts MRC5. Real-time PCR and Western blot were used to detect mRNA and protein levels, respectively. Results: The data presented show that an efficient replication of HCMV in HMVEC is dependent on SOCS3 protein. Time course analysis revealed an increase in SOCS3 protein levels in infected cells. Silencing of SOCS3 (siSOCS3) resulted in inhibition of viral immediate early, early, and late antigen production. Consistently, HCMV titers produced by siSOCS3 cultures were significantly decreased when compared to control transfected cultures (siCNTRs). STAT1 and STAT2 phosphorylation was increased in siSOCS3-infected cells when compared to siCNTR-treated cells. Conclusion: These findings indicate the implication of SOCS3 in the mechanism of HCMV-mediated control of cellular immune responses

    PARP inhibitor efficacy depends on CD8+ T cell recruitment via intratumoral STING pathway activation in BRCA-deficient models of triple-negative breast cancer.

    Get PDF
    Combinatorial clinical trials of PARP inhibitors with immunotherapies are ongoing, yet the immunomodulatory effects of PARP inhibition have been incompletely studied. Here, we sought to dissect the mechanisms underlying PARP inhibitor-induced changes in the tumor microenvironment of BRCA1-deficient triple-negative breast cancer (TNBC). We demonstrate that the PARP inhibitor olaparib induces CD8+ T cell infiltration and activation in vivo, and that CD8+ T cell depletion severely compromises anti-tumor efficacy. Olaparib-induced T cell recruitment is mediated through activation of the cGAS/STING pathway in tumor cells with paracrine activation of dendritic cells and is more pronounced in HR-deficient compared to HR-proficient TNBC cells and in vivo models. CRISPR-knockout of STING in cancer cells prevents proinflammatory signaling and is sufficient to abolish olaparib-induced T cell infiltration in vivo. These findings elucidate an additional mechanism of action of PARP inhibitors and provide rationale for combining PARP inhibition with immunotherapies for the treatment of TNBC

    PARP-inhibition reprograms macrophages toward an anti-tumor phenotype

    No full text
    Poly(ADP)ribosylation inhibitors (PARPis) are toxic to cancer cells with homologous recombination (HR) deficiency but not to HR-proficient cells in the tumor microenvironment (TME), including tumor-associated macrophages (TAMs). As TAMs can promote or inhibit tumor growth, we set out to examine the effects of PARP inhibition on TAMs in BRCA1-related breast cancer (BC). The PARPi olaparib causes reprogramming of TAMs toward higher cytotoxicity and phagocytosis. A PARPi-related surge in NAD+ increases glycolysis, blunts oxidative phosphorylation, and induces reverse mitochondrial electron transport (RET) with an increase in reactive oxygen species (ROS) and transcriptional reprogramming. This reprogramming occurs in the absence or presence of PARP1 or PARP2 and is partially recapitulated by addition of NAD derivative methyl-nicotinamide (MNA). In vivo and ex vivo, the effect of olaparib on TAMs contributes to the anti-tumor efficacy of the PARPi. In vivo blockade of the “don’t-eat-me signal” with CD47 antibodies in combination with olaparib improves outcomes in a BRCA1-related BC model.publishedVersio

    Generation and characterization of transgenic mice expressing K14 and K8 reporters.

    No full text
    <p>(A) Flow cytometry analysis for stromal, basal, and luminal compartments of cells from mammary glands of control or K14.tRPT/K8.tGPD double-positive transgenic mouse. For each type of mouse, the first dot plot shows the total population in each cell compartment, whereas the second plot shows only the cells that are positive for the reporters. Gates were set based on the negative control and dots pseudocolored to represent the reporter-positive cells. (B) Fluorescent IHC showing colocalization of tGFP with endogenous K8 in the lung of a K8.tGPD-positive mouse (upper panels). Lower panel shows the control staining on a WT mouse in which no tGFP was detected; scale bar 10 ÎŒm. (C) Positive and negative mice were injected i.p. with either high (“H”; GCV = 100 ÎŒg/g; DT = 50 ng/g) or low (“L”; GCV = 20 ÎŒg/g; DT = 10 ng/g) doses at indicated time points (days). DAPI, 4’,6-diamidino-2-phenylindole; DT, diphtheria toxin; GCV, ganciclovir; IHC, immunohistochemistry; i.p., intraperitoneally; K8.tGPD, keratin-8 promoter followed by turbo green fluorescent protein and diphtheria toxin receptor; K14.tRPT, keratin-14 promoter followed by a turbo red fluorescent protein and herpes simplex virus thymidine kinase; tGFP, turbo green fluorescent protein; WT, wild-type</p

    Characterization of K14.GFP reporter and relationship between K14+ and K14− status and invasive behavior of cells in culture.

    No full text
    <p>(A) Cartoon of the K14 promoter–driven EGFP-P2A-DTR (K14.GFP) reporter construct. (B) IF shows colocalization of endogenous K14 and GFP in K14.GFP+ monolayer, scale bar 20 ÎŒm. (C) Stably transfected K14.GFP reporter cells were sorted by FACS and monitored for changes in percentage of cells expressing GFP by flow cytometry for 54 days. (D) Phase contrast images of K14.GFP+, and K14.GFP− and WT cells grown in 2D, scale bar 100 ÎŒm. (E) Changes in cell number over time. Graph shows the mean ± SEM of 3 independent experiments. (F) EdU labeling and flow cytometry to identify cells in different stages of cell cycle. Shown is the mean ± SD of triplicates. (G) K14.GFP+, and K14.GFP− and WT cells grown in 3D (scale bar 200 ÎŒm) on top of M/Col-I for 4 days. Two days after seeding, cells were treated with DT (5 ng/ml) for 48 hours (lower panels, DT+). (H) Quantification of invasive structures in G, the data shown are means ± SD from independent experiments carried out in triplicates, at least 300 structures/condition were counted; *<i>p</i> < 0.05 by unpaired <i>t</i> test. (I) Phase contrast and GFP overlay images of K14.GFP+ cells grown in 3D in M/Col-I at day 2 and day 4. Black arrow indicates GFP+ cells at the invasive protrusions, white arrows indicate GFP+ in noninvasive 3D structures. DT+ indicates 48-hour exposure to DT, scale bars 100 ÎŒm. 3D, three-dimensional; DT, diphtheria toxin; DTR, diphtheria toxin receptor; EdU, 5-Ethynyl-2ÂŽ-deoxyuridine; EGFP, enhanced green fluorescent protein; FACS, fluorescence-activated cell sorting; GFP, green fluorescent protein; IF, immunofluorescence; K, cytokeratin; M/Col-I, 1:1 mixture of Matrigel/Collagen-I; pA, polyadenylation signal sequence; WT, wild type</p

    K14.GFP+ cells have greater metastatic potential that K14.GFP− cells <i>in vivo</i>.

    No full text
    <p>(A) Primary tumor diameters from mice injected with 4T1 control, or K14.GFP reporter cell lines (K14.GFP+ or K14.GFP−), measured over the course of the experiment. <i>n</i> = 8 (control), 7 (K14.GFP+), and 8 (K14.GFP−); *<i>p</i> < 0.05, **<i>p</i> < 0.001 by one-way ANOVA followed by Newman-Keuls multiple comparisons posttest. (B) Final tumor masses measured after mice were euthanized. (C) Lung metastases were quantified by measuring the average percent area of lung tissue occupied by the tumor in 5 sections (5-ÎŒm thick) cut at 200-ÎŒm intervals. (D) Representative images of H&E-stained lung sections containing metastases from mice injected with K14.GFP+ or K14.GFP− cell lines. Arrows indicate metastasis; scale bar 1 mm. (E) Correlation analysis of primary tumor mass and lung metastasis. Scatter plot of the percent tumor area in lung tissue compared to the primary tumor mass for each mouse analyzed (<i>n</i> = 23). The slope does not significantly differ from zero by linear regression analysis (<i>p</i> = 0.1097). Quantification of percent of positive cells for Ki67 (F) and CC3 (G) in tumors from K14.GFP+ and K14.GFP− cells. Shown are means ± SD of quantifications of whole tumor sections. (H) Final tumor masses measured after mice were euthanized. For DT treatment, the mice were injected i.p. with DT (25 mg/kg) on days 7, 9, 11, and 13. <i>n</i> = 7 (K14.GFP+; no DT), 4 (K14.GFP+; with DT), 8 (K14.GFP−; no DT), and 4 (K14.GFP−; with DT). <i>p</i> = 0.0425 by unpaired <i>t</i> test. (I) Lung metastases were quantified for the mice described in H. Statistical analysis for (B), (C), and (I) was calculated by one-way ANOVA followed by Tukey’s multiple comparisons posttest; *<i>p</i> < 0.05 or n.s. (J) Fluorescent IHC was performed for K14 and GFP on primary tumors generated from K14.GFP+ cell lines either DT− or DT treated (“DT+”) as described in (H); scale bar 40 ÎŒm. (K) Same staining as described in J was carried out on metastatic lung of K14.GFP+-injected mice; scale bar 20 ÎŒm. (L) Immunoblots of lysates from primary tumors were analyzed for GFP expression. Every lane represents a different tumor. Blots were also probed with antibodies for ERK1/2 as loading control. If not otherwise indicated, all graphs show mean ± SEM. DT, diphtheria toxin; ERK1/2, extracellular signal-regulated kinase 1/2; GFP, green fluorescent protein; H&E, hematoxylin and eosin; IHC, immunohistochemistry; i.p., intraperitoneally; K, cytokeratin; n.s., not significant</p

    K14+ cells secrete more Col6a1 and express higher levels of Amigo2 than K14− cells.

    No full text
    <p>(A) Anti-Col6a1 immunoblots of secreted proteins from K14.GFP+ or K14.GFP− reporter cell lines. CM were concentrated and analyzed for levels of secreted Col6a1. ITGB1, also present in the secretome, was used for loading control. (B) RT-PCR for <i>Amigo2</i> mRNA level on K14+ and K14− cells. Results show the mean ± SD of 3 independent experiments, <i>p</i> = 0.0126 by paired <i>t</i> test. (C) Amigo2 protein level was detected in K14+ and K14− cell lysates by western blot. Quantification of 3 independent experiments, <i>p</i> < 0.0001 by paired <i>t</i> test; mean ± SD is shown. (D) Chip for H3K27Ac shows the magnitude of the peaks for K14+ and K14− replicates at the <i>Amigo2</i> locus. (E) Cartoon of self-inactivating lentiviral K14.tRPT (upper cartoon) and K8.tGPD reporters (lower cartoon). (F) RT-PCR analysis of <i>Amigo2</i> mRNA expression in K14+ or K14− human breast cancer cell line HCC1143. Quantification of independent experiments in triplicates <i>p</i> = 0.0148 by paired <i>t</i> test; mean ± SD is shown. (G) Kaplan-Meier plot in TP53 mutant and TP53 WT breast cancer show relationship between <i>Amigo2</i> expression and relapse-free survival. Amigo2, amphoterin-induced protein 2; Chip, chromatin immunoprecipitation; CM, conditioned medium; Col6a1, Collagen VI subunit A; DTR, diphtheria toxin receptor; <i>EF-1α</i>, <i>elongation factor 1α</i>; GFP, green fluorescent protein; H3K27Ac, histone 3 lysine 27; ITGB1, integrin ÎČ-1; K, cytokeratin; K8.tGPD, keratin-8 promoter followed by turbo green fluorescent protein and diphtheria toxin receptor; K14.tRPT, keratin-14 promoter followed by a turbo red fluorescent protein and herpes simplex virus thymidine kinase; LTR, long terminal repeat; RT-PCR, real-time PCR; tBFP, turbo blue fluorescent protein; tGFP, turbo green fluorescent protein; TK, thymidine kinase; tRFP, turbo red fluorescent protein; WT, wild-type.</p
    corecore