Loss of Therapy-Induced Senescence in Myc-Driven Lymphomas Compromises Treatment Outcome In Vivo.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1686-1686
Author(s):  
Jan Dörr ◽  
Yong Yu ◽  
Christoph Loddenkemper ◽  
Ulrich Keller ◽  
Andreas Buck ◽  
...  

Abstract Abstract 1686 Poster Board I-712 Introduction Premature senescence, a terminal cell-cycle arrest condition, reflects an acute cellular stress response upon a variety of cellular insults including oncogene activation and chemotherapeutic DNA damage. Therefore, senescence potentially complements apoptosis as a safeguard program and tumor-suppressive mechanism. Although it is frequently observed in the clinic that patients who only achieve disease stabilization or partial remission in response to chemotherapy may experience lasting freedom from progression, no tumor model has shown that therapy-induced senescence (TIS) significantly contributes to treatment outcome. Here, we employ the Eμ-myc mouse lymphoma model with and without intact alleles of the histone H3 lysine 9 (H3K9) methyltransferase Suv39h1 (controlling senescence) and of p53 (mediating both apoptosis and senescence) to demonstrate a critical role for senescence in cancer therapy in vivo. Methods Lymphoma cells (LCs), retrovirally transduced with bcl2 to block apoptosis, were treated with the DNA damaging anticancer agent adriamycin (ADR) in vitro, or were exposed to the alkylating agent cyclophosphamide upon lymphoma formation in normal immunocompetent mice in vivo. TIS was analyzed by senescence-associated β-galactosidase activity (SA-β-gal), Ki67 staining and BrdU incorporation. Tumor formation, therapy and progression in vivo was monitored by whole body fluorescence and luciferase imaging and 18F-fluoro-deoxyglucose (FDG) and 18F-fluoro-deoxythymidine (FLT) positron emission tomography (PET). Time-course analysis of glucose and oxygen consumption rates and NAD(P)/NAD(P)H ratios over time were used to define a senescence energy consumption profile. Progression-free and overall survival was evaluated using the Kaplan-Meier method. Results Bcl2-protected control (i.e. no further defined genetic defects) LCs, but not Suv39h1- or p53-deficient LCs, enter TIS in vivo as evidenced by uniform SA-β-gal reactivity, high frequency of H3K9-trimethylation-positive cells, and loss of Ki67 staining and BrdU incorporation due to a cell-cycle block in the G1-phase. Notably, Suv39h1- lymphomas recapitulate the proliferation rate and sensitivity to drug-induced apoptosis of control lymphomas, but display significantly shorter progression-free and overall survival after chemotherapy in vivo. Despite their stable growth arrest ADR-senescent control;bcl2 lymphomas exhibited higher glucose metabolism and energy consumption in vitro when compared to their untreated counterparts or senescence-refractory, ADR-treated Suv39h1-;bcl2 lymphomas. Accordingly, TIS LCs can be non-invasively detected by a discordant positive FDG- but negative FLT-PET scan. Discussion: The study demonstrates that Suv39h1 acts as an essential mediator of TIS without compromising apoptosis or altering the proliferative capacity of lymphoma cells. Myc-driven lymphomas senesce in response to DNA damaging anticancer therapies, but display high levels of glucose metabolism. Therefore, they can be detected by FDG-PET despite their resting condition, indicating that a positive post-therapy FDG-PET scan in the clinic does not necessarily reflect a growing tumor lesion. In vivo, Bcl2-protected Suv39h1- lymphoma-bearing mice succumb dramatically fast to their disease after chemotherapy reminiscent of p53null lymphoma-bearing mice. Hence, Suv39h1-controlled TIS is a critical component of anticancer drug therapy in vivo and significantly extends progression-free and overall survival of the host. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2849-2849
Author(s):  
Nicolas Graf ◽  
Zhoulei Li ◽  
Ken Herrmann ◽  
Alexandra Junger ◽  
Daniel Weh ◽  
...  

Abstract Abstract 2849 Purpose: The thymidine analogue [18F]fluorothymidine (FLT) has been shown to reflect proliferation of high-grade lymphoma cells both in preclinical and clinical studies. In this preclinical in vitro and in vivo study we assessed early FLT-uptake as an adequate and robust surrogate marker for response to inhibitors of Nucleophosmin-anaplastic lymphoma kinase (NPM-ALK)-dependent pathways in an anaplastic large cell lymphoma (ALCL) xenotransplant model. Methods: In vitro investigations included viability assessment (MTT assay), cell cycle analysis using propidium iodide staining and western blotting to characterize response of the ALCL cell lines SUDHL-1 and Karpas299 to treatment with heat shock protein 90 (Hsp90) inhibitor NVP-AUY922, the Phosphoinositide 3-kinase (PI3K) inhibitor BGT226 or the mammalian target of rapamycin (mTOR) inhibitor RAD001. Thymidine metabolism in severe combined immunodeficient (SCID) mice bearing SUDHL-1 or Karpas 299 lymphoma xenotransplants was assessed non-invasively prior to and early in the course of therapy (48h to 7 days) by FLT and FDG positron emission tomography (FLT-PET and FDG-PET) using a dedicated small animal PET system. Tumor-to-background ratios (TBR) of FLT-PET were compared to that of PET using the standard radiotracer [18F]fluorodeoxyglucose (FDG). Reference for tumor response was local control of the tumor measured by shifting calliper and histopathological analysis of explanted lymphomas. Results: In vitro, SUDHL-1 cells were sensitive to all three inhibitors (IC50 AUY922= 50 nM; IC50 BGT266= 10 nM; IC50 RAD001= 1 nM). These cells showed a dose-dependent induction of cell-cycle arrest in G1-phase and reduction of S-Phase after 24 to 48 hours and - to a lesser extent - increase of apoptosis. Incubation of SUDHL-1 cells with NVP-AUY922 (50 nM) for 24 hours led to a 70% reduction of ALK level and a abrogation of Akt phosphorylation as determined by western blot analysis. Likewise, no phosphorylation of Akt was detectable after incubation with BGT266 (10 nM) already after 4 hours. RAD001 (0.1-1nM, 24h) completely inhibited phosphorylation of p70 S6K. In contrast, Karpas299 cells were only sensitive to RAD001-induced cell cycle arrest, but insensitive to NVP-AUY922 and BGT266. In vivo, we performed FLT- and FDG-PET scans to monitor inhibition of tumor growth in the course of therapy with NVP-AUY922. Tumor volume in treated animals bearing SUDHL-1 lymphomas showed modest increase within the first week (median increase= + 25%, range -30% to + 80%, n=8) as opposed to a 3.8-fold increase in untreated control animals. After 14 days a clear reduction of tumor mass could be observed (median= - 25%, range -40% to + 30%, n=4). Median TBR of FLT-PET decreased significantly to 40% compared to baseline as earlier as 5 days after initiation of therapy (range 32–67%, n=8, p=0,008). In contrast, the pattern of TBR in FDG-PET did not show any clear tendency (median TBR 79%, range 36%-161%, n=8, p=0,73). We then investigated the ability of FLT-PET to differentiate between sensitive and resistant lymphoma cells. Therefore, mice bearing Karpas299 lymphomas were treated with NVP-AUY922 (resistant in vitro) or RAD001 (sensitive in vitro). According to our in vitro results, no effect was seen during treatment with NVP-AUY299 as indicated by about 3-fold tumor growth on day 7 and increase of median TBR in FLT-PET to 162% (range 106–177%, p=0,008, n=8) on day 2. In contrast, mice receiving RAD001 showed a deceleration of tumor development with doubling of tumor volume within the first week (range -20% to + 320%, n=10) that remained fairly constant over the following weeks. FLT-PET imaging indicated a slight increase of TBR correctly reflecting tumor growth kinetics (median=126%, range 60–129%, no p-value). A larger cohort is currently investigated as well as histopathological analysis of explanted lymphomas. The updated data will be presented at the meeting. Conclusion: In contrast to FDG-PET, FLT-PET is able to predict response to specific inhibitors early in the course of the therapy using a anaplastic large cell lymphoma xenograft model and is able to distinguish between sensitive and resistant lymphoma cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3394-3394
Author(s):  
Kaitlyn Shank ◽  
Yusup Shin ◽  
Carson Wills ◽  
Nicole Cunningham ◽  
Alevtina Domashenko ◽  
...  

Abstract Abstract 3394 Hematopoietic stem cells (HSC) replenish the cellular components of the blood throughout life by a homeostatic process in which the majority of HSCs remain quiescent while a small percentage enter the cell cycle to either self-review or differentiate. During inflammatory responses to infections, Interferons (IFNa, IFNg) perturb HSC homeostasis, presumably in response to the demand for increased numbers of inflammatory cells. Previous studies have highlighted an apparent paradox, i.e. IFNs suppress the proliferation of normally cycling murine hematopoietic progenitor cells (HPCs), yet increase the fraction of normally quiescent Sca+ HSCs that proliferate. To investigate the mechanisms underlying this paradox, we dissected the dynamics of cell surface phenotypes, cell cycle kinetics, pro- and anti-apoptotic pathways within the HSC and HPC compartments in response to pIpC and IFNs both in vivo and in vitro. Forty-eight hours after pIpC injection, bone marrow (BM) cellularity declined by 60%, the proportion of Sca- kit+ HPCs fell from 0.45% to 0.05%, while the proportion of BM cells with the Sca+ kit+ HSC phenotype increased from 0.17 to 0.26%. To determine whether the increase in Sca+kit+ cells was due to proliferation of HSCs or upregulation of Sca-1 on HPCs, we cultured purified CD150+ Sca-Kit+ HPCs and CD150+Sca+kit+ HSCs in vitro with IFNa, IFNg, or PBS. Sca expression was induced on previously Sca- HPCs, and the level of Sca expression on HSCs was also increased. This induction was detectable as early as 6 hours after treatment and accompanied by an increase in Sca mRNA. BrdU incorporation into both HPC and HSC populations decreased from pre-treatment baselines, further indicating that the increase in cells with the HSC phenotype was not due to HSC proliferation, but rather the appearance of cycling HPCs within the HSC staining gate following IFN-induced upregulation of Sca. Staining with FITC-DEVD-FMK identified active cleaved capase-3 in pIpC- or IFN-treated cells, suggesting that the reduced cellularity following IFN reflected a cellular stress that killed Lin+ precursors cells and some HPCs, but spared HSCs. In contrast to lin+kit- precursors, all kit + HPCs and HSCs expressed bcl-2, suggesting that expression of anti-apoptotic proteins may prevent IFN-induced stress from resulting in HSC/HPC apoptosis despite the initial triggering of caspase-3 cleavage. In summary, acute treatment with IFNs has anti-proliferative effects on all hematopoietic cells, including precursors, HPCs and HSCs, with the apparent increase in HSC proliferation the result of HPCs masquerading as Sca+HSCs after exposure to IFN. Unlike precursors, HSCs and some HPCs survive treatment to IFNs despite activation of cleaved caspase-3, possibly due to their expression of bcl-2, and likely related anti-apoptotic regulators. The previously observed increase in HSC proliferation days and weeks following IFN treatment is most likely due to the homeostatic response of HSCs to the depopulation of the precursor and HPCs caused by acute IFN exposure. Disclosures: No relevant conflicts of interest to declare.


Endocrinology ◽  
2015 ◽  
Vol 156 (5) ◽  
pp. 1659-1670 ◽  
Author(s):  
Paloma Alonso-Magdalena ◽  
Marta García-Arévalo ◽  
Iván Quesada ◽  
Ángel Nadal

Evidence now exists supporting the hypothesis that endocrine-disrupting chemicals (EDCs) can harmfully impact glucose metabolism. Thus, EDCs are beginning to be considered important contributors to the increased incidence of diabetes, obesity, or both. The possible effect of exposure to EDCs during pregnancy on glucose homeostasis in mothers later in life is presently unknown. Here we show that several months after delivery, mothers treated with the widespread EDC bisphenol-A (BPA) during gestation, at environmentally relevant doses, exhibit profound glucose intolerance and altered insulin sensitivity as well as increased body weight. These mice presented a decreased insulin secretion both in vivo and in vitro together with reduced pancreatic β-cell mass. The proliferation capacity was decreased in association with a diminished expression of the cell cycle activators: cyclin D2 and cyclin-dependent kinase-4. In addition, the rate of β-cells apoptosis was increased as well as the expression of the cell cycle inhibitors p16 and p53. Conversely, no effects on glucose metabolism or insulin sensitivity were observed when female nonpregnant mice were treated with BPA at the same doses. Taken together, these findings reveal that BPA exposure during gestation has harmful long-term implications in glucose metabolism for the mother. This finding highlights a new window of susceptibility for EDC exposure that may be important for the development of type 2 diabetes.


2021 ◽  
Vol 5 (Supplement_1) ◽  
pp. A982-A983
Author(s):  
Reema S Wahdan-Alaswad ◽  
Ann D Thor

Abstract Background: Breast cancer (BC) and thyroid disease are well-recognized comorbidities. Hyperthyroidism and supraphysiologic thyroid hormone (TH) have been shown to promote BC incidence. We recently reported that thyroid hormone replacement therapy (THRT) was significantly and independently associated with shortened disease-free and overall survival, as well as endocrine resistance only in patients with steroid receptor-positive (SR+) BC (1). TH markedly upregulated estrogen and cell cycle signaling in vivo and in vitro and promoted dedifferentiation to basaloid and pre-stem phenotypes. Metformin (Met) attenuated this shift. Mechanisms of TH-mediated endocrine therapy resistance in ER+ BC are the focus of this report. Design: Two clinical cohorts of early-stage lymph node-negative (LN-) SR+ BC patients (n=820 and n=160) were used to determine the effect of THRT on overall survival using Kaplan-Meier methods. Bi-directional cross-talk between TH and E2 was tested using different BC cell lines, ER+ PDX in vivo models, in vitro methods, and publically available in silico data for modeling. Results: Our results show that E2+TH increases cell proliferation, enhances cell cycle, and hormone-associated oncogenic signaling in SR+/ER+ BC. Given that high expression of THRA is associated with poor prognosis in SR+ BC, knockdown of THRA and ESR1 reduced cell proliferation in ER+ BC cells. ER+ PDX tumors were implanted into NSG mice containing E2 pellet and subsequently treated with TH, Tamoxifen (Tam), Fulvestrant (ICI) or Met. Our data show that TH-mediated endocrine resistance only in the E2+TH+Tam treated tumors (P<0.0001 vs E2+Tam alone). Both ICI and Met provided significant attenuation of tumor growth in vivo. RNAseq analysis of E2+TH+Tam tumors show an increase in pro-oncogenic signaling (Wnt/Fizzled, MMPs, and TCL/LEFT). Our data suggest that the use of Tam did not dampen tumor growth whereas a full ER-antagonist (ICI) or Met attenuated E2-TH mediated cross-talk and tumor growth. Conclusions: These findings suggest that TH+Tam may enhance oncogenic signaling and is associated with a significantly increase in mortality risk in ER+/SR+ BC tumors. Exogenous TH adversely affects SR+ BC and not SR- BC. Understanding the mechanism of cross-talk between TH and E2 allows us to define novel therapeutic strategies that will facilitate rapid clinical application for ER+ BC patients currently taking THRT and anti-estrogen treatments. Reference: (1) Wahdan-Alaswad et. al. Clin Cancer Res October 23 2020 DOI: 10.1158/1078-0432.CCR-20-264.


2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Wen-Xiang Li ◽  
Jia-Jia Zheng ◽  
Gang Zhao ◽  
Chen-Tao LYU ◽  
Wei-Qi Lu

Abstract Objectives Down syndrome critical region 1 (DSCR1) is associated with carcinogenesis and tumor growth in several types of malignancy. However, little is known about the role of DSCR1 in CRC progression. The present study aimed to elucidate the clinicopathological significance, prognostic, and function roles of DSCR1 in CRC. Methods Firstly, we analyzed DSCR1 expression in 58 paired CRC samples and Oncomine database. Then, we analyzed DSCR1 expression in two independent CRC cohorts (test cohort: n = 70; validation cohort: n = 58) and tested its overall survival (OS) by Kaplan-Meier survival analyses. Finally, we overexpressed DSCR1 in two CRC cell lines DLD1 and LoVo and analyzed its effect on cell cycle and senescence. Results DSCR1 expression was significantly decreased in CRC samples and associated with clinicopathologic features of CRC patients, such as tumor size, lymph node metastasis, and TNM stage. CRC patients with low expression of DSCR1 had shorter overall survival (OS). Kaplan-Meier survival analyses showed that the expression of DSCR1 was significant factor for OS in both cohorts. Multiple Cox regression analysis showed that DSCR1 expression was an independent prognostic marker for OS in test cohort. Overexpression of DSCR1 isoform 4 (DSCR1-4) increased p21, p16, p-NFAT1, and p-NFAT2, while decreased CDK2, CDK4, and Cyclin D1 in CRC cells. In addition, overexpression of DSCR1-4 prevented proliferation and colony formation, and induced senescence in vitro. Moreover, overexpression of DSCR1-4 inhibited tumor growth and tumor angiogenesis in vivo. Conclusions Our study found high expression of DSCR1 contributes to favorable prognosis of CRC patients and prevents cell cycle and proliferation of CRC cells, indicating a critical tumor suppressive role in CRC progression.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 345-345 ◽  
Author(s):  
Julia Kase ◽  
Jan Dörr ◽  
Yong Yu ◽  
Maja Milanovic ◽  
Sujuan Ji ◽  
...  

Abstract Introduction Premature senescence, a terminal cell-cycle arrest condition of viable cells in response to acute cellular stresses such as oncogenic activation or DNA-damaging anticancer therapy, is characterized by S-phase entry-controlling histone H3 lysine 9 trimethylation (H3K9me3). Previously, we reported an essential, tumor-suppressive role for the H3K9 histone methyltransferase Suv39h1 in oncogene-induced senescence (OIS) as a barrier to lymphoma development in vivo (Braig-M et al., Nature, 2005). In the current study, we focused, in addition to Suv39h1, on the H3K9-active demethylases LSD1 and JMJD2C in both OIS and therapy-induced senescence (TIS). Methods Human diploid fibroblasts (HDFs) and mouse embryo fibroblasts (MEFs) were stably transfected with H-RasG12V to induce OIS. S-phase-promoting E2F target gene control by H3K9me3 was analyzed by chromatin immunoprecipitation. Transformation was assessed by anchorage-independent colony formation in vitro and tumor development in nude mice. To establish TIS, primary Eµ-myc transgenic mouse lymphoma cells, retrovirally transduced with bcl2 to block apoptosis, were exposed to adriamycin in vitro or cyclophosphamide in vivo. Senescence was analyzed by staining for senescence-associated b-galactosidase activity (SA-b-gal), Ki67 and BrdU incorporation. Lymphoma formation and treatment in vivo were monitored by luciferase and GFP imaging, SA-b-gal/Ki67 staining in situ, and overall survival was assessed by Kaplan Meier analysis. Results H3K9-active demethylases – like overexpression of a dysfunctional H3R9 mutant – blocked cellular senescence, and permitted direct transformation under oncogenic Ras. In Myc-driven lymphomas, either loss of Suv39h1 or overexpression of LSD1 or JMJD2C cancelled TIS in vitro and in vivo. Notably, H3K9me3-impaired lymphomas resembled control lymphomas in their proliferation rate and sensitivity to drug-induced apoptosis, but displayed significantly shorter progression-free and overall survival after chemotherapy. Extended data sets on LSD1 and JMJD2C expression in human diffuse large B-cell lymphoma samples and their correlation to treatment outcome will be presented at the meeting. Conclusion The data underscore the essential albeit dynamic role of the H3K9me3 mark in OIS and TIS, and unveil the oncogenic potential of H3K9 demethylases, thereby providing a mechanistic basis for JMJD2C- or LSD1-targeting strategies in lymphoma therapy. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 733-733 ◽  
Author(s):  
Masashi Numata ◽  
Ramon Klein Geltink ◽  
Gerard Grosveld

Abstract Although ETS-transcription factors play a role in normal and malignant hematopoiesis, their function in hematopoietic stem cells (HSCs) and leukemia initiating cells (LICs) is largely unknown. We originally identified the novel ETS transcription factor ETV7, which is highly homologous to ETV6/TEL, a frequent target of chromosomal translocation in human leukemia. Previously we have shown that ETV7 is a hematopoietic oncoprotein that requires cooperating mutations to induce leukemic transformation. Microarray analysis revealed that ETV7 expression is upregulated in 70% of pediatric ALL and AML samples. This indicates a possible oncogenic function of ETV7 in a variety of leukemias, although the molecular mechanism of ETV7-mediated leukemogenesis remains to be elucidated. ETV7 is widely but not abundantly expressed in various human tissues. Recently we found that overexpression of ETV7 in human cord blood-derived CD34+ cells depletes the number of CD34+CD38- HSCs. In addition, ETV7-transduced cells slightly accerelated cell proliferation. These results suggest that overexpression of ETV7 activates cell proliferation in primary human CD34+cells and depletes the number of HSCs. Here, by using a mouse model, we show that ectopic expression of ETV7 in quiescent HSCs accelerates their cell cycle entry and proliferation, leading to the exhaust of HSCs in mice. The ETV7 gene locus is deleted in part of the rodents including the mouse despite its high level of conservation among vertebrates. To circumvent this limitation, we have generated an ETV7 BAC transgenic mouse that carries a single copy of a human BAC DNA containing the ETV7 gene locus. In flow cytometry (FCM) analysis of wild type (WT) and ETV7 bone marrow (BM)-derived Lin-Sca1+cKit+(LSK) cells, the size and frequency of LT(long term)-HSCs (CD48-CD150+LSK) in ETV7 LSK was 2-fold lower than that in WT LSK, while the frequency of LSK and hematopoietic common progenitor cells in WT and ETV7 BM are similar. As compared with WT-LSK, ETV7-LSK showed a significantly decreased number of myeloid progenitor colonies in both the initial plating (MC1) and replating of MC1 colonies (MC2) in methylcellulose colony formation assay in vitro. To assess the ETV7 HSC function contributing to blood cell generation in vivo, we performed competitive repopulation assays. In agreement with the in vitro results, the repopulation ability of HSC is significantly compromised in ETV7 mice as measured 7 weeks post transplantation. This defect was even more pronounced 16 weeks post transplantation. Since enhanced cell cycle entry is known to cause loss of hematopoietic stem/progenitor cells (HSPCs) through the activation of a tumor suppressor response, we quantified p19ARF, p16INK4a, and p21CDKN1A gene expression in LSK cells by qRT-PCR. At day 6 and day 9 of in vitro culture, ETV7 LSK cells activated the p19ARF, p16INK4a, and p21CDKN1A genes about 2-fold greater than WT LSK cells. To measure the de novo DNA replication of HSPCs in vivo, BrdU-pulse labeled BM cells were harvested and BrdU incorporation was quantified by FCM analysis. ETV7 LSK cells showed elevated BrdU incorporation compared with that of WT. In addition, Hoechst33342/Pyronin Y staining revealed that ETV7 LSK enhanced transition from G0 to the G1 phase of the cell cycle, suggesting that ETV7 forced cell cycle entry of quiescent HSCs. Finally to clarify the involvement of the CDKN2A tumor suppressor in ETV7-associated HSC exhaustion, we examined the frequency of HSPCs in CDKN2A-/- and ETV7+/-CDKN2A-/- LSK cells in vivo by FCM analysis. Loss of CDKN2A but not ARF restored the depletion of ETV7 LT-HSCs. Moreover, loss of CDKN2A rescued the defect of repopulation ability in vivo and self-renewal activity in vitro of ETV7 HSPCs. These results indicate that exhaustion of HSC in ETV7 BM occurred through ETV7-induced activation of cell proliferation and the CDKN2A tumor suppressor pathway in mice. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1666-1666
Author(s):  
Katerina Rejlova ◽  
Miroslava Kardosova ◽  
Martina Slamova ◽  
Marketa Zaliova ◽  
Meritxell Alberich-Jorda ◽  
...  

Abstract The DLX homeodomain genes are part of the Drosophila distal-less family, originally identified in the forebrain of the developing mouse embryo. DLX1 gene is expressed also in the hematopoietic cells. Our previous data showed that patients with FLT3/ITD (internal tandem duplication) mutation representing about 35% of all acute myeloid leukemia (AML) cases have higher expression of DLX1 compared to non-FLT3/ITD AML patients. Further, FLT3 signaling was described to regulate DLX1 gene expression. In the current study we found an association between DLX1 expression level and overall survival using GEP data from TCGA database within the group of FLT3/ITD-positive patients. Worse overall survival was linked to lower expression of DLX1 (p=0.003, n=46) even though generally this group of patients is characterized by their higher DLX1 level. This association was also observed when all AML patients were analyzed (p=0.01, n=197). Since the role of DLX1 in the leukemogenic process is not yet described we aimed to characterize the phenotype of leukemic cells with different expression levels of DLX1 gene. We designed two functional shRNAs (pGhU6-sh1_DLX1 and pGhU6-sh2_DLX1, referred as sh1 and sh2) to downregulate DLX1 expression in MV4;11 (FLT3/ITD positive) leukemic cells, which present high endogenous level of DLX1, and a non-silenced control (pGhU6-NSC). In vitro studies showed that cells with silenced DLX1 were arrested in G0 phase of cell cycle (35%±4.8 (NSC) to 67.5%±2.2 (sh1; p ˂0.01) and 65.5%±2.5 (sh2; p ˂0.01); flow cytometry - Pyronin Y/ Hoechst 33342 staining) and had lower proliferative activity (trypan blue over the period of 10 passages). Moreover, cells with silenced DLX1 were less apoptotic (Annexin V/PI staining). Next, we studied the impact of DLX1 downregulation on leukemic cell infiltration in vivo using sub-lethally irradiated NSG (NOD SCID gamma) mice. We injected 1x106 silenced or non-silenced MV4;11 cells via tail vein (n=20, 6 mice/group, 2 control mice). After two weeks we measured the absolute number of MV4;11 cells by flow cytometry (CD33+/GFP+/DAPI-) in bone marrow (BM) and spleen (SP) of recipient mice. Absolute cell counts of leukemic cells with silenced DLX1 were 1.26x106 (sh1; p=0.015) and 2.89x106 (sh2; p˂0.0001) vs NSC cells 0.52x106 in BM and 1.59x106 (p=0.036) and 14.2x106 (p=0.025) vs 0.34x106 in SP. To ensure that the increased cell numbers were not the result of enhanced homing of DLX1 silenced cells, we performed homing experiments. Control and DLX1 silenced cells were transplanted and the number of cells in BM and SP was determined 16 hours after transplantation. We observed no differences between the studied groups, indicating that DLX1 silencing does not affect the homing ability of MV4;11 cells. Moreover, we analyzed cell cycle in leukemic cells isolated from recipient mice three weeks after transplantation. In agreement with our in vitro results, leukemic cells with silenced DLX1 had higher percentage of cells arrested in G0 phase (48.6%±6.3 (sh1; p ˂0.001) and 80.9%±9.3 (sh2; p ˂0.001) vs 26.4%±4.8 (NSC) in SP). Overall, mice with silenced DLX1 presented worse fitness and bigger splenomegaly. Further, we investigated the signaling pathways which could lead to G0 arrest. Since DLX1 inhibits TGF-β pathway through direct interaction with SMAD4, a key downstream effector of TGF-β/BMP signaling, we studied the changes in target genes in our model cell lines. Expression of CUTL-1 (sh1 - 1.5-fold, sh2 - 2-fold change to NSC), PAI-1 (3.5-fold, 4.2-fold) and CDKN1C (1.3-fold, 2.3-fold) were significantly increased in DLX1 silenced cells. PAI-1 was shown to induce replicative senescence and CDKN1C is an inhibitor of cell cycle progression. While both targets disturb cell cycle and could be responsible for the phenotype we have observed, this hypothesis needs to be elucidated in future experiments. Altogether, our data demonstrate that dysregulation of DLX1 gene in leukemic cells changes the cell phenotype. Lower level of DLX1 gene leads to arrest in G0 phase which in vitro slows down the proliferation whereas in vivo it allows the cells to persist in spleen and BM. We hypothesize that the DLX1 silenced cells become more resistant to external effects which could then be reflected as a reduced survival observed in patients with low levels of DLX1. Supported by P304/12/2214. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Vol 2 (4) ◽  
pp. 83-98 ◽  
Author(s):  
André De Lima Mota ◽  
Bruna Vitorasso Jardim-Perassi ◽  
Tialfi Bergamin De Castro ◽  
Jucimara Colombo ◽  
Nathália Martins Sonehara ◽  
...  

Breast cancer is the most common cancer among women and has a high mortality rate. Adverse conditions in the tumor microenvironment, such as hypoxia and acidosis, may exert selective pressure on the tumor, selecting subpopulations of tumor cells with advantages for survival in this environment. In this context, therapeutic agents that can modify these conditions, and consequently the intratumoral heterogeneity need to be explored. Melatonin, in addition to its physiological effects, exhibits important anti-tumor actions which may associate with modification of hypoxia and Warburg effect. In this study, we have evaluated the action of melatonin on tumor growth and tumor metabolism by different markers of hypoxia and glucose metabolism (HIF-1α, glucose transporters GLUT1 and GLUT3 and carbonic anhydrases CA-IX and CA-XII) in triple negative breast cancer model. In an in vitro study, gene and protein expressions of these markers were evaluated by quantitative real-time PCR and immunocytochemistry, respectively. The effects of melatonin were also tested in a MDA-MB-231 xenograft animal model. Results showed that melatonin treatment reduced the viability of MDA-MB-231 cells and tumor growth in Balb/c nude mice (p <0.05). The treatment significantly decreased HIF-1α gene and protein expression concomitantly with the expression of GLUT1, GLUT3, CA-IX and CA-XII (p <0.05). These results strongly suggest that melatonin down-regulates HIF-1α expression and regulates glucose metabolism in breast tumor cells, therefore, controlling hypoxia and tumor progression. 


2019 ◽  
Vol 20 (1) ◽  
pp. 56-62 ◽  
Author(s):  
Chi Zhang ◽  
Zhichun Gu ◽  
Long Shen ◽  
Xianyan Liu ◽  
Houwen Lin

Background: To deliver drugs to treat Alzheimer’s Disease (AD), nanoparticles should firstly penetrate through blood brain barrier, and then target neurons. Methods: Recently, we developed an Apo A-I and NL4 dual modified nanoparticle (ANNP) to deliver beta-amyloid converting enzyme 1 (BACE1) siRNA. Although promising in vitro results were obtained, the in vivo performance was not clear. Therefore, in this study, we further evaluated the in vivo neuroprotective effect and toxicity of the ANNP/siRNA. The ANNP/siRNA was 80.6 nm with good stability when incubated with serum. In vivo, the treatment with ANNP/siRNA significantly improves the spatial learning and memory of APP/PS1 double transgenic mice, as determined by mean escape latency, times of crossing the platform area during the 60 s swimming and the percentage of the distance in the target quadrant. Results and Conclusion: After the treatment, BACE1 RNA level of ANNP/siRNA group was greatly reduced, which contributed a good AD treatment outcome. Finally, after repeated administration, the ANNP/siRNA did not lead to significant change as observed by HE staining of main organs, suggesting the good biocompatibility of ANNP/siRNA. These results demonstrated that the ANNP was a good candidate for AD targeting siRNA delivery.


Sign in / Sign up

Export Citation Format

Share Document