21 research outputs found

    Twist1 Suppresses Senescence Programs and Thereby Accelerates and Maintains Mutant Kras-Induced Lung Tumorigenesis

    Get PDF
    KRAS mutant lung cancers are generally refractory to chemotherapy as well targeted agents. To date, the identification of drugs to therapeutically inhibit K-RAS have been unsuccessful, suggesting that other approaches are required. We demonstrate in both a novel transgenic mutant Kras lung cancer mouse model and in human lung tumors that the inhibition of Twist1 restores a senescence program inducing the loss of a neoplastic phenotype. The Twist1 gene encodes for a transcription factor that is essential during embryogenesis. Twist1 has been suggested to play an important role during tumor progression. However, there is no in vivo evidence that Twist1 plays a role in autochthonous tumorigenesis. Through two novel transgenic mouse models, we show that Twist1 cooperates with KrasG12D to markedly accelerate lung tumorigenesis by abrogating cellular senescence programs and promoting the progression from benign adenomas to adenocarcinomas. Moreover, the suppression of Twist1 to physiological levels is sufficient to cause Kras mutant lung tumors to undergo senescence and lose their neoplastic features. Finally, we analyzed more than 500 human tumors to demonstrate that TWIST1 is frequently overexpressed in primary human lung tumors. The suppression of TWIST1 in human lung cancer cells also induced cellular senescence. Hence, TWIST1 is a critical regulator of cellular senescence programs, and the suppression of TWIST1 in human tumors may be an effective example of pro-senescence therapy

    Macrophage biology in development, homeostasis and disease

    Get PDF
    Macrophages the most plastic cells of the hematopoietic system are found in all tissues and exhibit great functional diversity. They have roles in development, homeostasis, tissue repair, and immunity. While anatomically distinct, resident tissue macrophages exhibit different transcriptional profiles, and functional capabilities, they are all required for the maintenance of homeostasis. However, these reparative and homeostatic functions can be subverted by chronic insults, resulting in a causal association of macrophages with disease states. In this review, we discuss how macrophages regulate normal physiology and development and provide several examples of their pathophysiologic roles in disease. We define the “hallmarks” of macrophages performing particular functions, taking into account novel insights into the diversity of their lineages, identity, and regulation. This diversity is essential to understand because macrophages have emerged as important therapeutic targets in many important human diseases

    <i>TWIST1</i> is overexpressed in human primary lung cancers.

    No full text
    <p>(A) Human non-small cell lung cancer samples (n = 394) compared against normal lung (n = 159) from seven independent microarray datasets for <i>TWIST1</i> expression using Oncomine. The heatmap contains individual studies (see accompanying legend; #2 and #3 are from the same dataset analyzed by adenocarcinoma and squamous cell carcinoma, respectively). The heat map intensity corresponds to percentile overexpression (red) or repression (blue). The median rank across all eight datasets demonstrates <i>TWIST1</i> is overexpressed in human lung cancer, <i>p</i> = 0.04. (B) We validated this microarray analysis by performing qPCR on primary human tumor samples for <i>TWIST1</i>. <i>TWIST1</i> mRNA is overexpressed in human lung cancer (n = 164) compared to normal lung (n = 28), <i>p</i><0.0001 by Mann-Whitney t-test. (C) Analysis of data from (B) broken down by adenocarcinoma (Adeno, n = 73) and squamous cell carcinoma (SCCA, n = 53) histology, <i>p</i><0.0001 using one-way ANOVA.</p

    Inducible <i>Twist1</i> lung model of epithelial mesenchymal transition (EMT).

    No full text
    <p>(A) A mouse line containing the Clara cell secretory protein (CCSP) promoter driving the reverse tetracycline transactivating protein (rtTA) is crossed with a line containing <i>Twist1</i> and <i>Luc</i> under the control of bi-directional tetracycline-responsive elements (<i>tetO<sub>7</sub></i>). In the bitransgenic animal, <i>CCSP-rtTA</i>/<i>Twist1-tetO<sub>7</sub>-luc</i> (CT), absence of doxycycline prevents rtTA protein from binding and activating the tetO operon. Addition of doxycycline (Dox) triggers a conformational change which enables <i>tetO<sub>7</sub></i> binding, activation and <i>Twist1</i> and <i>luc</i> transcription. CT animals express Twist1 and luciferase inducibly in the lungs and trachea of bitransgenic mice as shown by (B) bioluminescence imaging (BLI) on a Xenogen Spectrum and (C) Western blotting for Twist1. BLI was performed on the same CT mouse with time “ON” or “OFF” Dox as indicated. (D) Enrichment plot of an EMT_UP signature following GSEA performed on lung mRNA samples taken from CT mouse lungs Dox ON (n = 2) and wildtype mouse lungs Dox ON (n = 2), NOM <i>p</i>-values, FDR <i>q</i>-values, and FWER <i>p</i>-values were all <0.001. (E) Plot of E-cadherin<sup>low</sup>-Vimentin<sup>high</sup> cells per field of view immunofluorescence (IF) of the lungs from CT animals ON (n = 4) and wildtype (n = 4) animals; <i>p</i><0.01 by Mann-Whitney t-test. d – day; wk – week; and m – month.</p

    <i>Twist1</i> accelerates <i>Kras<sup>G12D</sup></i>-induced lung tumorigenesis and promotes progression to adenocarcinoma.

    No full text
    <p>(A) Kaplan-Meier tumor free survival using serial microCT of <i>CCSP-rtTA</i>/<i>Twist1-tetO<sub>7</sub>-luc</i> (CT), <i>CCSP-rtTA</i>/<i>tetO-Kras<sup>G12D</sup></i> (CR) and <i>CCSP-rtTA</i>/<i>tetO-Kras<sup>G12D</sup></i>/<i>Twist1-tetO<sub>7</sub>-luc</i> (CRT) mice. The double inducible oncogene animals (CRT) developed multiple tumors at a median tumor latency that was significantly shorter than the single CR animals, 15 weeks, by log-rank analysis (<i>p</i><0.0001). A syngenic control cohort consisting of wildtype mice, those with <i>tetO-Kras<sup>G12D</sup></i>/<i>Twist1-tetO<sub>7</sub>-luc</i> (without <i>CCSP-rtTA</i>), <i>CCSP-rtTA</i> alone, or single oncogenes alone (n = 15 total) never developed lung tumors before 12 months of age. (B) Lung tumors from a CRT mouse at necropsy and H&E sections. Black bars equal 200 and 50 µm. H – heart; and L – liver. (C) Immunohistochemical (IHC) phenotyping of CRT tumors using antibodies against CCSP and proSpC. (D) Lung tumor burden is increased at 6 months in CRT <i>versus</i> CR mice qualitatively by microCT and H&E histology. Blue arrowheads denote lung tumors. (Lower panel) Lung tumors were quantified for CR <i>versus</i> CRT mice by microCT (n = 4 mice each). S – spine. Black bar equals 2 mm (E) Ki-67 IHC of CR <i>versus</i> CRT lung tumors (n = 3 mice each). Low - <5%; Med – 5–25%; and High - >25%. Histologic examination of lung tumors for numbers of benign lesions (hyperplasia, atypical adenomatous hyperplasia and adenomas) <i>versus</i> adenocarcinomas (AdenoCA) for CR and CRT mice (n = 2 mice each).</p

    <i>TWIST1</i> knockdown activates senescence in human non-small cell lung cancer cells.

    No full text
    <p>Three different shRNAs were able to knockdown <i>TWIST1</i> mRNA levels and result in decreased TWIST1 protein in the <i>KRAS</i> mutated non-small cell lung cancer (NSCLC) cell line H460 as shown by (A) qPCR and (B) TWIST1 Western blotting on day 9 after the shRNA infection. (C) Representative duplicates of crystal violet staining of serially diluted H460 NSCLC cells demonstrate <i>TWIST1</i> knockdown decreases cellular proliferation. (D) Representative photomicrographs of increased SA-β-gal staining of cells following shRNA mediated <i>TWIST1</i> knockdown using sh-TWIST1-39. (E) Quantification of SA-β-gal stained cells following shRNA mediated <i>TWIST1</i> knockdown. (F) <i>TWIST1</i> knockdown in H460 results in the upregulation of some additional markers of senescence, p21 and p27 as shown by Western blotting on day 9 after the shRNA infection. (G) A549 cells require <i>TWIST1</i> overexpression to form subcutaneous tumors in NOD-SCID mice. A contingency table of A549 cells infected with sh-Scrambled control or sh-TWIST1 shRNA that were implanted into NOD-SCID mice and 4 weeks later scored for tumor development (5/6 <i>versus</i> 0/5, respectively, <i>p</i> = 0.01 by Fisher's exact test).</p

    <i>Twist1</i> accelerates conditional <i>Kras<sup>G12D</sup></i>-induced lung tumorigenesis.

    No full text
    <p>(A) Crosses (CT×LSL) to produce <i>CCSP-rtTA</i>/<i>Twist1-tetO<sub>7</sub>-luc</i>/<i>LSL-Kras<sup>G12D</sup></i> (CT-LSL) mice. CT-LSL mice are infected with intranasal Cre to activate <i>Kras<sup>G12D</sup></i>. Addition of Dox enables <i>Twist1</i> and <i>luc</i> transcription. In contrast to CRT OFF mice, CT-LSL OFF mice have <i>Kras<sup>G12D</sup></i> still active and only <i>Twist1</i> expression is inactivated. (B) Kaplan-Meier tumor free survival by serial microCT of F1 littermates with CT, LSL and CT-LSL genotypes. The double oncogene animals (CT-LSL, n = 18) developed multiple tumors at a median tumor latency that was significantly shorter than the single LSL (n = 14) animals, 5 <i>versus</i> 6 weeks (CT-LSL <i>versus</i> LSL, by log-rank analysis <i>p</i> = 0.0121). CT animals (n = 17) and littermate controls not infected with AdCMVCre (n = 5) never developed lung tumors. (C) Lung tumor burden is increased at 9 weeks post-AdCMVCre in CT-LSL <i>versus</i> LSL mice qualitatively by representative microCT. Blue arrowheads denote lung tumors. H – heart; and S – spine. (D) H&E stained sections of lung tumors from a CT-LSL mouse. Black bars equal 200 and 50 µm. (E) Immunohistochemical (IHC) phenotyping of CT-LSL lung tumors indicate a type II pneumocyte cell of origin using CCSP and proSp-C markers. (F) Ki-67 IHC of LSL <i>versus</i> CT-LSL lung tumors (n = 2). Low - <5%; Med – 5–25%; and High - >25%. (G) pErk1/2 and p19<sup>ARF</sup> IHC staining in serial sections demonstrate overlap. Note the nucleolar staining of p19<sup>ARF</sup>, specific nuclei are denoted by blue arrowheads.</p

    Activation of <i>Kras</i>-induced senescence by down-regulation of <i>Twist1</i> in autochthonous <i>Kras<sup>G12D</sup></i>/<i>Twist1</i>-induced lung tumors.

    No full text
    <p>(A) Verification by qPCR that <i>Twist1</i> mRNA levels are reduced following doxycycline withdrawal in CT-LSL OFF (n = 4) compared to CT-LSL ON (n = 3) lung tumors. (B) CT-LSL OFF lung tumors are static following single <i>Twist1</i> inactivation. Representative serial microCT of CT-LSL lung tumor moribund mouse just before, CT-LSL ON, and 4 weeks following doxycycline removal from the drinking water, CT-LSL OFF, resulting in de-induction of <i>Twist1</i>only (n = 13 tumors quantified from 4 mice). For comparison, <i>LSL-Kras<sup>G12D</sup></i> (LSL) mouse lung tumors grow despite withdrawal of doxycycline, LSL OFF (n = 11 tumors quantified from 3 mice). Percent tumor volume growth was quantified and calculated showing CT-LSL OFF tumor stasis after 4 weeks compared to LSL OFF (<i>p</i><0.0001). H – heart; and S – spine. CT-LSL OFF lung tumors demonstrate markers consistent with an increase in the number of senescent cells, such as (C) reduction in proliferation by Ki-67 IHC, (D) increased lung tumors positive for SA-β-gal staining, increased levels of (E) p21 and (F) p16 by IHC (n = 3 mice per staining). (G) pErk1/2 levels reduced moderately following <i>Twist1</i> inactivation in CT-LSL OFF tumors. (H–L) Quantification of (C–G) as described in previous figures for Ki67 (see <a href="http://www.plosgenetics.org/article/info:doi/10.1371/journal.pgen.1002650#pgen-1002650-g002" target="_blank">Figure 2</a>) and SA-β-gal (see <a href="http://www.plosgenetics.org/article/info:doi/10.1371/journal.pgen.1002650#pgen-1002650-g003" target="_blank">Figure 3</a>) staining; and 21, p16 and pERk1/2 as follows - Low - <10%; Med – 10–25%; and High - >25%. All animals in these experiments were taken off doxycycline (“OFF”) continuously for 2–5 weeks and then sacrificed.</p
    corecore