Monthly Archives: August 2019

Our results may provide the missing clue to reconcile the seemingly contradictory findings

Its activity was reduced by approximately 50% with an energy load of 30 W, which is 5-fold lower than the energy used for platelet lysis. Using the highenergy Masitinib VEGFR/PDGFR inhibitor sonication protocol employed in previous reported studies, we found that platelet PAI-1 activity was reduced approximately 90%. Taken together, with the activity rates observed in the present study, one would expect sonication to reduce platelet PAI-1 activity to 7�C8%, i.e. to similar levels as reported in previous studies. The magnitude of the reduction in PAI-1 activity was similar when freezing/thawing was used for platelet lysis. However, whereas the reduced activity by sonication was independent of whether tPA was added before or after lysis, the Bortezomib clinical trial underestimation of activity by freezing/thawing could partially be prevented by adding tPA before lysis. Another common procedure for platelet disruption is to use detergents such as Triton X-100. However, it has been shown that Triton X-100 decreases the half-life of active PAI-1 markedly, and 0.2% Triton X-100 decrease the functional half-life of PAI-1 to less than 1 minute at 37uC. Therefore, also with such protocols it is crucial to add tPA before lysis. Since addition of Triton X-100 is not physiological and may facilitate the binding of tPA and PAI-1, we investigated if Triton X-100 affected the results of the Western blot analysis. However, when Triton X-100 was added to the platelets lysed by sonication and freezing/thawing no such enhancement was observed. A potential concern in the present study could have been that the procedure we used in some way could have reactivated PAI-1 although it was in fact inactive in vivo. In vitro PAI-1 can be reactivated by denaturants such as SDS, guanidine HCl, and urea, and it has also been suggested that negatively charged phospholipids exposed on the surface of activated platelets could reactivate PAI-1. On the other hand, it has been reported that SDS may cause dissociation of the tPA-PAI-1 complex. To rule out the possibility that our results were due to reactivation and/or dissociation of the tPA-PAI-1 complex formed, we performed a series of experiments both with and without SDS in the loading buffer before electrophoresis. However, these studies showed no detectable differences in PAI-1 activity whether SDS was present or not. This is in agreement with a previous study reporting that the SDS-activatable form of PAI-1 might not be present in human platelets. How, then, could the activity of PAI-1 be preserved for such a prolonged period of time in the platelet? A potential mechanism has been suggested by Lang and Schleef, who showed that platelets possess a unique mechanism for stabilization of active PAI-1, by packaging together with other large a-granule proteins in a calcium-dependent manner. Active PAI-1 in plasma is stabilized by binding to vitronectin which has also been detected in platelet a-granules. However, some studies have failed to detect the vitronectin-PAI-1 complex in platelets and it is therefore controversial whether vitronectin is the stabilizing factor of PAI-1 in platelets. This issue remains to be evaluated. From a clinical perspective, there is compelling evidence that platelet-derived PAI-1 has an important physiologic and pathophysiologic role in making platelet-rich blood clots resistant to both endogenous and pharmacological thrombolysis. Despite this, most previous studies have reported activity levels of platelet PAI-1 that are probably far too low to explain its putative functional role.

This was demonstrated in several types of tumor cell lines and potentially involved the PI3K/AKT pathway

The mechanisms regulating the inhibitory effects of lovastatin on EGFR function and the synergistic cytotoxicity in combination with gefitinib are currently not known. These findings suggest that mevalonate pathway inhibitors and receptor TKI may represent a novel combinational therapeutic approach in a variety of human cancers. The VEGFR and the EGFR are both members of RTK family that share similar activation, internalization and downstream signaling characteristics. Cycloheximide Therefore, PCI-32765 targeting the mevalonate pathway may have similar inhibitory effects on VEGFR and may also enhance the activity of VEGFR-TKI. VEGFR, particularly VEGFR-2, play important roles in regulating angiogenesis by promoting endothelial cell proliferation, survival and migration. VEGF and VEGFR are also expressed by some tumor cells, like MM, acting in a functional autocrine loop capable of directly stimulating the growth and survival of MM cells. In this study, we have shown lovastatin does indeed inhibit ligand-induced VEGFR-2 activation through inhibition of receptor internalization resulting in diminished AKT activation in HUVEC and MM cells. Lovastatin treatment re-organized the actin cytoskeleton, inhibited proliferation and induced apoptosis of HUVEC at therapeutically relevant doses despite addition of exogenous VEGF. AKT activation, which mediates cell survival, along with its downstream targets S6K1 and 4EBP1 were significantly inhibited by lovastatin treatment. Combining lovastatin with VEGFR-TKIs also induced synergistic cytotoxicity of HUVEC cells. Due to their role in promoting tumor neovascularization, inhibiting the function of VEGF and VEGFR has been the focus of a number of therapeutic approaches. The limited clinical responses associated with these agents have been associated with their ability to promote disease stabilization and rarely induce tumor regression. Thus, agents that can cooperate and enhance the activity of VEGFR-TKI, like lovastatin, may increase their therapeutic activity. MM is a highly aggressive tumor that is rarely curative and median survival is in the range of 10�C17 months, therefore, novel therapies for MM are needed. Elevated levels of circulating and serousal VEGF in MM patients and the expression of VEGF and VEGFR on MM cells that can drive their proliferation and enhance their survival has led to the evaluation of VEGFR targeted therapies. Bevacizumab, a monoclonal antibody against the VEGF, which is approved for the treatment of colon cancer, in combination with chemotherapy, failed to significantly affect outcome to chemotherapy treatment alone. Various VEGFRTKI employed a single agents also failed to demonstrate clinical utility in MM patients. As like HUVEC, MM cells also depend on VEGFR signaling, we also examined the effect of lovastatin alone and in combination with VEGFR-2 TKI on MM cell viability. Combining 5 mM lovastatin treatments with two VEGFR-2 inhibitors in the H28 and H2052 mesothelioma derived cell lines demonstrated synergistic cytotoxicity through the induction of a potent apoptotic response. These results highlight a novel mechanism regulating VEGFR-2 function and a potential novel therapeutic approach for MM. Inhibition of HMG-CoA reductase has been evaluated as an anti-cancer therapeutic approach owing to its ability to inhibit tumor cell proliferation, induce tumor specific apoptosis and inhibit cell motility and metastasis in several tumor models. A number of Phase I Clinical trials evaluating the efficacy of high doses of lovastatin failed to demonstrate significant antitumor activity.

We have documented inhibition of nuclear entry of stress-responsive transcription factors

Since both immune BAY-60-7550 responses depend on intracellular signal transduction by stress responsive transcription factors, we targeted the nuclear import mechanism with a first-in-class, cell-penetrating nuclear import inhibitor. We found that short-term intracellular delivery of this inhibitor afforded long-term protection of the islets from inflammation-driven apoptosis. This long-lasting islet-protecting effect, which arrests diabetes progression without the need for insulin therapy, appears to involve the precipitous reduction of autoreactive lymphocytes through enhancement of activation-induced cell death of T and B lymphocytes. Moreover, this salutary effect of short-term nuclear import targeting is associated with reprogramming of the pro-inflammatory and anti-inflammatory cytokine profile of immune cells isolated from NOD mice. The ultimately fatal outcome of autoimmune diabetes in the widely used and clinically relevant murine NOD model of human T1D depends on progressive and relentless destruction of insulin producing beta cells in pancreatic islets and is inevitable unless insulin-replacement therapy is instituted. We investigated the possibility of protecting islets from autoimmune attack by intracellular delivery of a nuclear import inhibitory peptide. Our results indicate that cSN50 effectively protected islets from immune destruction. This protection is vested in significant reduction of islet-reactive T cells, restoration of the sensitivity of T and B cells to AICD, suppression of islet-toxic proinflammatory cytokine production in primary T and B cells and macrophages isolated from NOD mice, and preservation of a key anti-inflammatory cytokine, IL-10. Thus, a nuclear import inhibitor extinguished autoimmune inflammation-driven islet loss and prevented further progression of diabetes thereby obviating the need for insulin replacement therapy. Remarkably, none of the cSN50 peptide-treated INCB28060 supply animals developed hyperglycemia during the first phase of diabetes onset occurring between days 10�C30 after receiving a bolus of cyclophosphamide, which synchronized the autoimmune diabetes process in NOD mice. In addition to this early protective effect, cSN50 treatment also afforded significant long-term islet protection. While another half of Cy-synchronized control animals progressed to diabetes between days 50 and 100, only two of twenty cSN50treated animals developed diabetes, a finding suggesting that cSN50 treatment resulted in long-term islet protection in NOD mice that are genetically-prone to T1D. This favorable outcome is supported by our demonstration of in vivo elimination of islet-infiltrating and islet-reactive lymphocytes, most likely through enhanced AICD, which was demonstrated ex vivo in cSN50 peptide-treated T and B cells. Our data indicate that at higher levels of stimulation, the sensitivity to AICD is further increased by the nuclear import inhibitory peptide. Thus, chronic activation of isletinfiltrating T and B cells in autoimmune diabetes may favor the AICD-enhancing effect of the nuclear import inhibitor. This hitherto unreported action of cSN50 in a relevant preclinical T1D model adds autoimmune inflammation to the list of conditions in which a nuclear import inhibitor has displayed therapeutic utility. We have previously demonstrated the efficacy of nuclear import inhibitor delivery and its anti-inflammatory and cytoprotective action in acute inflammation models, including lethal challenge with superantigen, staphylococcal enterotoxin B, and lipopolysaccharide, which trigger acute inflammatory lung and liver injury.

shRNA down-regulation of Metnase levels markedly sensitized to adriamycininduced immunoprecipitate with Topo IIa

Together, this provides evidence that Metnase could play a role in the pathogenesis and resistance of metastatic breast cancer to Topo IIa inhibiting therapies. Since Metnase enhances Topo IIa-mediated decatenation, and enhances resistance to ICRF-193 and VP-16 in non-malignant human cells, we hypothesized that Metnase might also promote resistance to the anthracyclines and epididophyllotoxins in MDAMB-231 cells. We first investigated whether reducing Metnase would affect ICRF-193-mediated metaphase arrest. MDA-MB-231 cells were treated with ICRF-193, which inhibits Topo IIa after DNA religation, and therefore does not induce DSBs but does inhibit decatenation, allowing for discrimination between DNA damage and metaphase arrest. The increase in cells arrested at metaphase in the presence of ICRF-193 compared to vehicle controls provides a measure of cells arrested due to failure of decatenation. Using atubulin immunoR428 fluorescence microscopy, we determined the fraction of cells in metaphase after exposure to ICRF-193. Cells with reduced Metnase expression showed a significantly higher percentage of metaphase arrested cells when treated with ICRF193 and cytospun onto slides to retain all cells.After 18 hour treatments with 2 or 10 mM ICRF-193, or 4 hours with 10 mM ICRF-193, cells with reduced Metnase showed 4.9-fold, 2.2-fold, and 2.6-fold increased metaphase arrest, respectively, as compared to vector control and evaluated by student’s t-test. This result suggests that Metnase promotes decatenation in ICRF-193-treated MDA-MB-231 cells, allowing them to proceed through metaphase even in the presence of this Topo IIa specific inhibitor. Prior studies revealed that bladder and lung cancer cells progress through the decatenation checkpoints when Topo IIa is inhibited by high concentrations of ICRF-193. The conclusion from those studies was that these cancer cells failed to arrest because they had inactivated the decatenation checkpoints. While the ability to progress through mitosis even when Topo IIa is inhibited may be a general feature of malignancy, it may be due to the presence of Metnase alone, or Metnase in combination with checkpoint inactivation. Thus, the decatenation checkpoint may be intact in these malignant cells, but Metnase promotes continued Topo IIa function despite the presence of inhibitors, and the decatenation checkpoint is not activated. The Topo IIa inhibitor ICRF-193 does not induce significant DNA damage, and therefore is not relevant in the clinical therapy of breast cancer. To determine whether altering Metnase levels would affect resistance to clinically relevant Topo IIa inhibitors, such as VP-16 and adriamycin, we determined the cytotoxicity of these agents in MDA-MB-231 cell lines that stably under-expressed Metnase using colony formation assays. Decreased Metnase expression increased sensitivity 7.5-fold to VP-16, and 3.5-fold to adriamycin. Together, these results indicate that Metnase expression levels directly correlate with cell survival after exposure to these clinically relevant Topo IIa inhibitors. Adriamycin is an important agent in both adjuvant therapy and in the treatment of metastatic breast adenocarcinoma, so this finding is of relevance for current clinical ICG-001 Wnt/beta-catenin inhibitor regimens. It raises the possibility that treatment efficacy could be improved if the drug was used in combination with a future Metnase inhibitor, or if Metnase levels could be measured and possibly account for variance in responsiveness to adriamycin based chemotherapeutic regimens. Altogether, these results provide further support for the hypothesis that Metnase plays a key role in Topo IIa function. To determine the mechanism for the ability of Metnase to mediate sensitivity to Topo IIa inhibitors, we investigated whether Metnase levels affected the cellular apoptotic response to adriamycin. We exposed MDA-MB-231 cells to adriamycin for 24 hrs and then evaluated annexin-V/FITC fluorescence by flow cytometry.

By protein structure followed by the biological assay validation of the candidate compounds in a cell culture system

These compounds may serve as the basis for the development of new treatments against Dengue virus infection. This procedure may be applied to other viral pathogens or for any other mechanism that involves specific conformational alterations for biological function. Our study also highlights the additional characteristics of certain tetracycline derivatives as effective inhibitors of DV propagation, which will allow further refinement of our screening program and potential medical application. Cell-based screens have been widely used in drug discovery although historically, these assays are conducted using genetically diverse cell lines derived from human tumors. Since the complex intracellular signaling networks that drive cancer cell growth and survival have begun to be elucidated, a more rational approach to drug discovery has become feasible. However, the implementation of target-orientated cell-based screens for anticancer drugs remains a challenge, both because of their reliance on defined genetic changes and because of the lack of proper control cells. To overcome this fundamental problem, we have developed a rational strategy for cell-based drug discovery that is based on the convenienceand flexibility of theBa/F3 cellsystem, animmortalized IL-3-dependent pro-B lymphoblastic cell line. IL-3 supports the growth and survival of Ba/F3 cells through the activation of distinct signaling pathways. Upon AZ 960 binding to its cognate receptor IL-3 activates the Janus kinase signal transduction and transcriptional activation pathways to induce Bcl-xL. Similarly, IL-3 activation of the PI3K/Akt pathway is involved in inhibiting the intrinsic apoptotic machinery in Ba/F3 cells. Overexpression of several SAR131675 constitutively active signaling molecules abrogates the dependence of these cells on IL-3. Hence, we generated isogenic cell lines derived from Ba/F3 in which IL-3 independent survival is sustained by independent signaling events. Each of these isogenic lines was genetically labeled with a fluorescent reporter and thus, the ratio of two spectrally distinct cell populations could be used as primary endpoint of the system to monitor pathway-specific cytotoxicity. Accordingly compounds can be screened in co-cultures of these lines and the change in the relative cell number of the two lines readily and rapidly measured to identify those molecules that specifically interact with one of the signaling pathways. In this instance, BaFiso has been designed as a live-cell system suitable to identify specific inhibitors of Akt signaling. The most frequently used anti-cancer therapies were discovered on the basis of their anti-proliferative activity in functional cell assays but with no pre-existing knowledge of the mechanism of action. As a result none of the current drugs directly targets the molecular lesions responsible for malignant transformation and they are not selective. Indeed this lack of selectivity between cancer cells and normal cells is currently one of the main reasons for the failure of conventional chemotherapy. In recent years, our understanding of the genetics of human cancer has increased rapidly, enabling more rational approaches to drug discovery for anti-cancer therapies to be adopted. Accordingly, the present study set out to develop a rational cell-based drug discovery strategy, an approach that has historically been compromised by the lack of appropriate control cells. With the objective of identifying lead compounds that specifically kill cells with activated Akt signaling and that spare control cells, we have combined the use of co-cultured isogenic cell lines with fluorescent technology. We introduced a myristoylated form of Akt which constitutively localizes to the plasma membrane, bypassing the requirement for PIP3 in Akt activation. This myr-Akt has been shown to constitutively inactivate proapoptotic downstream targets. In order to generate Ba/F3 cells that survive in the absence of IL-3 independent of activated PI3K/Akt signaling.