Photodynamic Therapy (PDT)-Mediated Inhibition of the Transcription Factor Yin Yang 1 (YY1) That Regulates Resistance In Lymphoma

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 5113-5113
Author(s):  
Valentina Rapozzi ◽  
Sara Huerta-Yepez ◽  
Abhijeet Joshi ◽  
Mario I. Vega ◽  
Stavroula Baritaki ◽  
...  

Abstract Abstract 5113 Photodynamic therapy (PDT) is a cancer therapeutic treatment that uses a compound called the “photosensitizer” and a particular type of visible light. When photosensitizers are exposed to a specific wavelength of light (600-800 nm), cytotoxic oxygen species are generated that kill cells (Dougherty, TJ et al., JNCI 90:889, 1998). Several clinical trials are currently underway to evaluate the use of PDT for a variety of cancers. A phase II study has been completed with photodynamic therapy in the treatment of patients with lymphoma or chronic lymphocytic leukemia. (NCT00054171). Recently, we have focused our attention about the properties of the photosensitizer Pheophorbide a (Pba), a chlorine, and its effects on different types of solid tumor cells (Rapozzi, V et al., Cancer Biol Ther 14:1318, 2009). The objective of the present study is to investigate the biochemical and molecular mechanisms by which PDT signals the B-NHL Raji lymphoma cell line (as model) and rendering the cells susceptible to both the cytotoxic mechanism of the tumor microenvironment in vivo or to the response to cytotoxic agents in vitro. We hypothesized that treatment of Raji cells with Pba/PDT in our in vitro system may result in the inhibition of resistance factors that regulate tumor cell responses to both chemotherapeutic and immunotherapeutic drugs. Our recent findings demonstrated that the constitutively overexpressed transcription factor Yin Yang 1 (YY1) regulates, in part, tumor cell resistance in lymphoma (Vega, MI et al., J Immun 175:2174, 2005). Accordingly, we examined whether treatment of Raji lymphoma cells with Pba/PDT will also result in the downregulation of YY1 expression and reverse resistance. The Raji cells were seeded at a cell density of 2×105/ml in Petri dishes. When the cells reached a 70% confluency, they were treated with different concentration (80-160-240 nM) of Pba for three hours in the dark and were then irradiated by an LED light source (640 nm at 12,7 mW for 9 min; 6.7 J/cm2). Following the light treatment, the cells were harvested at different times of incubation (18-36h) to assess apoptosis by the activation of caspase 3 using flow cytometry. In addition, different aliquots of cells were used to prepare slides for immunohistochemistry analyses. The results demonstrate that, indeed, treatment with Pba/PDT resulted in the inhibition of YY1 protein expression in Raji cells. By immunohistochemistry, PDT inhibited the basal nuclear and cytoplasmic expression of YY1 and resulted in weak cytoplasmic YY1 expression. The mechanism of YY1 inhibition might have been the result of PDT-mediated inhibition of NF-κB activity (Karmakar, S. et al., Neurosci lett 415: 242, 2007) since YY1 is transcriptionally regulated by NF-κB (Wang, H et al., Mol Cell Biol 67:4374, 2007). In addition, our preliminary findings demonstrate that treatment of drug-resistant tumor cells with PDT sensitizes the cells to drug-induced apoptosis. Overall, the data suggest that YY1 may be considered as a novel therapeutic target in PDT. Based on the findings here, we are currently examining the role of PDT in the dysregulation of the NF-κB/YY1/Snail/RKIP loop (Wu, K and Bonavida, B. Crit Rev Immun 29:241, 2009) that regulates cell survival and proliferation and resistance in lymphoma. (We acknowledge Doctors Oscar Stafsudd and Romaine Saxton for their assistance.) Disclosures: No relevant conflicts of interest to declare.

BMC Cancer ◽  
2020 ◽  
Vol 20 (1) ◽  
Author(s):  
Ethan P. Metz ◽  
Erin L. Wuebben ◽  
Phillip J. Wilder ◽  
Jesse L. Cox ◽  
Kaustubh Datta ◽  
...  

Abstract Background Quiescent tumor cells pose a major clinical challenge due to their ability to resist conventional chemotherapies and to drive tumor recurrence. Understanding the molecular mechanisms that promote quiescence of tumor cells could help identify therapies to eliminate these cells. Significantly, recent studies have determined that the function of SOX2 in cancer cells is highly dose dependent. Specifically, SOX2 levels in tumor cells are optimized to promote tumor growth: knocking down or elevating SOX2 inhibits proliferation. Furthermore, recent studies have shown that quiescent tumor cells express higher levels of SOX2 compared to adjacent proliferating cells. Currently, the mechanisms through which elevated levels of SOX2 restrict tumor cell proliferation have not been characterized. Methods To understand how elevated levels of SOX2 restrict the proliferation of tumor cells, we engineered diverse types of tumor cells for inducible overexpression of SOX2. Using these cells, we examined the effects of elevating SOX2 on their proliferation, both in vitro and in vivo. In addition, we examined how elevating SOX2 influences their expression of cyclins, cyclin-dependent kinases (CDKs), and p27Kip1. Results Elevating SOX2 in diverse tumor cell types led to growth inhibition in vitro. Significantly, elevating SOX2 in vivo in pancreatic ductal adenocarcinoma, medulloblastoma, and prostate cancer cells induced a reversible state of tumor growth arrest. In all three tumor types, elevation of SOX2 in vivo quickly halted tumor growth. Remarkably, tumor growth resumed rapidly when SOX2 returned to endogenous levels. We also determined that elevation of SOX2 in six tumor cell lines decreased the levels of cyclins and CDKs that control each phase of the cell cycle, while upregulating p27Kip1. Conclusions Our findings indicate that elevating SOX2 above endogenous levels in a diverse set of tumor cell types leads to growth inhibition both in vitro and in vivo. Moreover, our findings indicate that SOX2 can function as a master regulator by controlling the expression of a broad spectrum of cell cycle machinery. Importantly, our SOX2-inducible tumor studies provide a novel model system for investigating the molecular mechanisms by which elevated levels of SOX2 restrict cell proliferation and tumor growth.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2388-2388 ◽  
Author(s):  
Laura G. Corral ◽  
Dan Zhu ◽  
Yuedi Wang ◽  
Bernd Stein

Abstract IMiDs® immunomodulatory drugs are thalidomide analogues that have been developed for improved anti-cancer and anti-inflammatory properties and decreased side effects. Many IMiDs® immunomodulatory drugs have been shown to have activities in hematologic cancers and solid malignancies, as well as having profound effects on the bone marrow microenvironment. Specifically in NHL, it was shown that addition of Revlimid® or CC-4047 to Rituxan enhances anti-tumor activity in a SCID mouse lymphoma model. Here we tested the direct effects of Revlimid® and CC-4047 on NHL tumor cells by treating Raji cells with each drug alone or with each drug in combination with anti-CD20 antibodies B1 and Rituxan. CC-4047 alone caused up to 40% inhibition of proliferation at 10 μM in Raji cells, which corresponded to G1 arrest. In combination with B1, CC-4047 showed a small additive effect at 10 μM while Revlimid® effects were minimal up to 10 μM. In combination with Rituxan, CC-4047 showed a slight additive effect at 10 μM and Revlimid® at 50 μM. We have also developed a co-culture assay of PBMC and NHL tumor cells as an in vitro model of tumor-host immune system interaction, to further explore the anti-tumor potential of the drugs in NHL. This assay is non-radioactive and flow cytometry based. In this co-culture system using Raji and PBMC, we have shown that pre-treatment of PBMC with Revlimid® or CC-4047 can enhance the PBMC activity in inducing Raji cell apoptosis in a dose dependent manner. In addition, our data indicate that pre-treatment of Raji cells with Rituxan can further enhance the apoptosis induced by PBMC pre-treated with Revlimid® or CC-4047. Since minimal additive effect between each drug and Rituxan was observed in the Raji single tumor cell model, these studies suggest that the co-culture system is a more appropriate cellular model to assess the anti-tumor activities of certain IMiDs® immunomodulatory drugs. This system can reveal the effects of certain IMiD® immunomodulatory drugs not observable with single tumor cell proliferation models. In summary, our data clearly demonstrate that Revlimid® and CC-4047 directly induce NHL tumor cell growth arrest and effectively enhance tumor cell apoptosis induced by PBMC. These results support clinical evaluation of Revlimid® and certain IMiDs® immunomodulatory drugs in relapsed B-cell NHL in combination with Rituxan.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2887-2887
Author(s):  
Melisa Martinez-Paniagua ◽  
Mario I. Vega ◽  
Sara Huerta-Yepez ◽  
Hari Hariharan ◽  
Haiming Chen ◽  
...  

Abstract Abstract 2887 Galiximab (anti-CD80 mAb) is a primatized mAb (human IgG1 constant region and Cynomogous macaque variable region) that binds CD80 on lymphoma cells. It has been shown in vitro that Galiximab inhibits tumor cell proliferation and mediates ADCC. Galiximab is currently in clinical trials for a variety of cancers. Our preliminary findings demonstrated that Galiximab treatment of B-NHL cell lines, like Raji, triggers the cells and inhibits the constitutively activated NF-κB pathway. We hypothesized that Galiximab-induced inhibition of NF-κB may result in the inhibition downstream of several anti-apoptotic gene products and sensitizes cells to drug-induced apoptosis. Raji cells were treated with Galiximab (20-100 μg/ml) for 18h and followed by treatment with the chemotherapeutic drug CDDP (5-10 μg/ml) for 24h and apoptosis was determined by flow for activation of caspase 3. The findings demonstrated that the cells treated with Galiximab were sensitized to CDDP-induced apoptosis. Analysis of the apoptotic pathway following treatment with Galiximab revealed the inhibition of anti-apoptotic gene products such as Bcl-2 and Bclxl. We have also found that Galiximab, like rituximab, inhibits the Fas and DR5 transcription repressor Yin Yang 1 (YY1) and the direct inhibition of YY1 resulted in tumor cell sensitization to both Fas-L and TRAIL. We examined whether inhibition of YY1 by Galiximab was also involved in the sensitization to CDDP apoptosis. Raji cells were treated with YY1 siRNA and, unlike control siRNA or non-treated siRNA cells, the tumor cells were sensitized to CDDP apoptosis. The inhibition of YY1 by siRNA correlated with the inhibition of Bcl-2 and Bclxl. The direct role of Bcl-2 and Bclxl in the regulation of resistance was corroborated by treatment of cells with the Bcl-2 family inhibitor, 2MMA3, and such cells mimicked Galiximab and were sensitive to CDDP-induced apoptosis. The mechanism by which treatment with YY1 siRNA resulted in the inhibition of Bcl-2 and Bclxl and the reversal of resistance is not clear. We suggest that YY1 inhibition, following Galiximab-induced inhibition of NF-κB, will result in the inhibition of Snail transcription (Palmer, MB et al., Mol cancer Res 7:221, 2009). Inhibition of the RKIP (Raf kinase inhibitor protein) repressor Snail will result in the induction of RKIP (Wu, K and Bonavida, B Crit Rev immu 29:241, 2009) and, in turn, RKIP will inhibit NF-κB and resulting downstream in the inhibition of Bcl-2 and Bclxl. In addition, it has been reported that YY1 negatively regulates p53 (Sui, G et al., Cell 117:889, 2004) and YY1 inhibition by Galiximab will upregulate p53 and which will result in the inhibition of Bcl-2 and Bclxl (see scheme below). The present findings demonstrate that Galiximab sensitizes drug-resistant B-NHL cells to drug-induced apoptosis via modulation of the NF-κB/YY1/Snail/RKIP/p53 loop. Current studies are validating the present findings with freshly-derived B-NHL cells and also examining the molecular mechanism by which YY1 regulates Bcl-2/Bclxl expression and the reversal of resistance. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Gautier Follain ◽  
Naël Osmani ◽  
Valentin Gensbittel ◽  
Nandini Asokan ◽  
Annabel Larnicol ◽  
...  

AbstractTumor progression and metastatic dissemination are driven by cell-intrinsic and biomechanical cues that favor the growth of life-threatening secondary tumors. We recently identified pro-metastatic vascular regions with blood flow profiles that are permissive for the arrest of circulating tumor cells. We have further established that such flow profiles also control endothelial remodeling, which favors extravasation of arrested CTCs. Yet, how shear forces control endothelial remodeling is unknown. In the present work, we aimed at dissecting the cellular and molecular mechanisms driving blood flow-dependent endothelial remodeling. Transcriptomic analysis of endothelial cells revealed that blood flow enhanced VEGFR signaling, among others. Using a combination of in vitro microfluidics and intravital imaging in zebrafish embryos, we now demonstrate that the early flow-driven endothelial response can be prevented upon specific inhibition of VEGFR tyrosine kinase and subsequent signaling. Inhibitory targeting of VEGFRs reduced endothelial remodeling and subsequent metastatic extravasation. These results confirm the importance of VEGFR-dependent endothelial remodeling as a driving force of CTC extravasation and metastatic dissemination. Furthermore, the present work suggests that therapies targeting endothelial remodeling might be a relevant clinical strategy in order to impede metastatic progression.


2020 ◽  
Vol 22 (Supplement_3) ◽  
pp. iii278-iii278
Author(s):  
Viktoria Melcher ◽  
Monika Graf ◽  
Marta Interlandi ◽  
Natalia Moreno ◽  
Flavia W de Faria ◽  
...  

Abstract Atypical teratoid/rhabdoid tumors (ATRT) are pediatric brain neoplasms that are known for their heterogeneity concerning pathophysiology and outcome. The three genetically rather uniform but epigenetically distinct molecular subgroups of ATRT alone do not sufficiently explain the clinical heterogeneity. Therefore, we examined the tumor microenvironment (TME) in the context of tumor diversity. By using multiplex-immunofluorescent staining and single-cell RNA sequencing (scRNA-seq) we unveiled the pan-macrophage marker CD68 as a subgroup-independent negative prognostic marker for survival of ATRT patients. ScRNA-seq analysis of murine ATRT-SHH, ATRT-MYC and extracranial RT (eRT) provide a delineation of the TME, which is predominantly infiltrated by myeloid cells: more specifically a microglia-enriched niche in ATRT-SHH and a bone marrow-derived macrophage infiltration in ATRT-MYC and eRT. Exploring the cell-cell communication of tumor cells with tumor-associated immune cells, we found that Cd68+ tumor-associated macrophages (TAMs) are central to intercellular communication with tumor cells. Moreover, we uncovered distinct tumor phenotypes in murine ATRT-MYC that share genetic traits with TAMs. These intermediary cells considerably increase the intratumoral heterogeneity of ATRT-MYC tumors. In vitro co-culture experiments recapitulated the capability of ATRT-MYC cells to interchange cell material with macrophages extensively, in contrast to ATRT-SHH cells. We found that microglia are less involved in the exchange of information with ATRT cells and that direct contact is a prerequisite for incorporation. A relapse xenograft model implied that intermediary cells are involved in the acquisition of chemotherapy resistance. We show evidence that TAM-tumor cell interaction is one mechanism of chemotherapy resistance and relapse in ATRT.


1992 ◽  
Vol 23 (4) ◽  
pp. 891-897 ◽  
Author(s):  
Amato J. Giaccia ◽  
Elizabeth A. Auger ◽  
Albert Koong ◽  
David J. Terris ◽  
Andrew I. Minchinton ◽  
...  

2010 ◽  
Vol 107 (12) ◽  
pp. 1490-1497 ◽  
Author(s):  
Konstanze Beck ◽  
Ben J. Wu ◽  
Jun Ni ◽  
Fernando S. Santiago ◽  
Kristine P. Malabanan ◽  
...  

1976 ◽  
Vol 143 (3) ◽  
pp. 601-614 ◽  
Author(s):  
J W Schrader ◽  
G M Edelman

Cytotoxic T lymphocytes were generated in vitro against H-2 compatible or syngeneic tumor cells. In vitro cytotoxic activity was inhibited by specific anti-H2 sera, suggesting that H-2 antigens are involved in cell lysis. Two observations directly demonstrated the participation of the H-2 antigens on the tumor cells in their lysis by H-2-compatible T cells. First, coating of the H-2 antigens on the target tumor cell reduced the number of cells lysed on subsequent exposure to cytotoxic T cells. Second, when cytotoxic T cells were activated against an H-2 compatible tumor and assayed against an H-2-incompatible tumor, anti-H-2 serum that could bind to the target cell, but not to the cytotoxic lymphocyte, inhibited lysis. H-2 antigens were also shown to be present on the cytotoxic lymphocytes. Specific antisera reacting with these H-2 antigens, but not those of the target cell, failed to inhibit lysis when small numbers of effector cells were assayed against H-2-incompatible target cells or when effector cells of F1-hybrid origin and bearing two H-2 haplotypes were assayed against a tumor cell of one of the parental strains. These findings suggest that it is the H-2 antigens on the tumor cell and not those on the cytotoxic lymphocytes that are important in cell-mediated lysis of H-2-compatible tumor cells.


2017 ◽  
Vol 2 (1) ◽  
Author(s):  
Dalia Martinez-Marin ◽  
Courtney Jarvis ◽  
Thomas Nelius ◽  
Stéphanie Filleur

Abstract Macrophages have been recognized as the main inflammatory component of the tumor microenvironment. Although often considered as beneficial for tumor growth and disease progression, tumor-associated macrophages have also been shown to be detrimental to the tumor depending on the tumor microenvironment. Therefore, understanding the molecular interactions between macrophages and tumor cells in relation to macrophages functional activities such as phagocytosis is critical for a better comprehension of their tumor-modulating action. Still, the characterization of these molecular mechanisms in vivo remains complicated due to the extraordinary complexity of the tumor microenvironment and the broad range of tumor-associated macrophage functions. Thus, there is an increasing demand for in vitro methodologies to study the role of cell–cell interactions in the tumor microenvironment. In the present study, we have developed live co-cultures of macrophages and human prostate tumor cells to assess the phagocytic activity of macrophages using a combination of Confocal and Nomarski Microscopy. Using this model, we have emphasized that this is a sensitive, measurable, and highly reproducible functional assay. We have also highlighted that this assay can be applied to multiple cancer cell types and used as a selection tool for a variety of different types of phagocytosis agonists. Finally, combining with other studies such as gain/loss of function or signaling studies remains possible. A better understanding of the interactions between tumor cells and macrophages may lead to the identification of new therapeutic targets against cancer.


Sign in / Sign up

Export Citation Format

Share Document