fas l
Recently Published Documents


TOTAL DOCUMENTS

176
(FIVE YEARS 20)

H-INDEX

35
(FIVE YEARS 2)

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3492-3492
Author(s):  
Martina Antoniolli ◽  
Maria Solovey ◽  
Deepak Bararia ◽  
Carolin Dorothea Strobl ◽  
William David Keay ◽  
...  

Abstract Follicular lymphoma (FL) is a clinically and genetically heterogeneous disease. Somatic gene mutations contribute to the heterogeneous clinical course of FL. ARID1A, which encodes for a subunit of the SWI/SNF chromatin remodeling complex, is among the most commonly mutated genes in FL (up to 15% of cases). These mutations are mostly disruptive and are predicted to result in protein haplodeficiency. While we have previously shown that ARID1A mutations are predictive of treatment outcome (Pastore, 2015), the underlying biology of ARID1A loss in FL is unclear. A functional genome-wide in vitro screen showed that ARID1A loss rescued a number of cancer cell lines from FAS-L induced apoptosis (Luo, 2008). FAS-L induced apoptosis plays a critical role in normal B-cell development and homeostasis. Thus, FAS/FAS-L deficiency could contribute to FL development and disease biology. Therefore, we studied the role of ARID1A loss in FAS expression and regulation. We first tested FAS-L induced apoptosis in established lymphoma cell lines that harbor the FL-hallmark translocation t(14;18)[BCL2/IGH] plus ARID1A mutations (Karpas422, WSU-FSCCL) or no ARID1A mutations (OCI-Ly1, OCI-Ly8, SU-DHL16). ARID1A mutant (mut) cells were indeed markedly less sensitive to FAS-L (300 ng/mL/24 hrs) compared to ARID1A wild type (WT) cells (98% vs 52% mean viability by Annexin-V). FAS receptor expression on mutant cells was reduced by almost half compared to WT cells by FACS analysis (N=3, P=0.0004). To test if reduced FAS expression was directly linked to ARID1A loss, we generated single-cell derived clones (from OCI-Ly1 and OCI-Ly8) with either heterozygous (het) loss or complete knock-out (KO) of ARID1A by CRISPR/Cas9. ARID1A loss was validated by Sanger sequencing and Western blot. We consistently observed significantly reduced FAS-L induced apoptosis in het and KO clones (exemplary shown for OCI-Ly8 in Fig A). Remarkably, re-expressed of ARID1A in het cells (het+ARID1A) rescued sensitivity to FAS-L induced apoptosis (Fig A). We confirmed reduced FAS expression on mutant clones by FACS, while re-expression of ARID1A rescued its expression (Fig B). Furthermore, FAS mRNA expression was significantly reduced by qPCR in mut vs WT clones (N=4, P<0.05), while FAS mRNA levels were rescued to WT levels in het+ARID1A cells. To understand the molecular mechanism that links ARID1A loss and reduced FAS expression, we performed ATAC sequencing (Seq) and RNA Seq on 15 single-cell derived clones (9 mut and 6 WT from OCI-Ly1 and OCI-Ly8). RNA Seq confirmed significantly lower ARID1A and FAS mRNA levels (adj p<0.001 each) in the mut clones. We first hypothesized that ARID1A loss could directly affect chromatin accessibility at the FAS promoter. However, we did not observe different chromatin accessibility at the FAS promoter. Next, we searched our data for all known FAS-regulating transcription factors (TFs) (https://dorothea.opentargets.io/#/), but could not identify candidates that were both differentially accessible and differentially expressed. Finally, we searched our data for transcriptional networks, i.e. hubs of all recognized FAS-regulating TFs and their known and predicted interacting partners (https://string-db.org/). Through this, we identified RUNX3, a predicted Co-TF of ETS1, to be both less accessible ("closed chromatin") and less expressed upon ARID1A loss (Fig C), suggesting a novel ARID1A-dependent FAS-regulatory network. To functionally validate our model, we first confirmed reduced RUNX3 expression in ARID1A mutant clones by qPCR and Western blot, and showed that ETS1 levels were unaffected by ARID1A loss. Then, we stably overexpressed RUNX3 in ARID1A mutant clones by lentiviral transduction and could indeed show rescue of FAS surface levels by FACS (Fig D). Lastly, we wanted to validate our findings in primary patients samples. We quantified FAS expression in FL biopsies with known ARID1A mutation status by nCounter gene expression profiling (GEP; N=51, 12 mut vs 39 WT) and quantitative multispectral imaging (QMI; N=44, 10 mut vs 34 WT) (Fig E). Both approaches showed significantly reduced FAS expression in ARID1A mutant FL (P<0.05 for GEP, P<0.0001 for QMI; Fig E). In summary, we show that ARID1A loss is directly linked to reduced FAS expression via a novel RUNX3/ETS1 transcriptional network, potentially opening avenues for therapeutic targeting of this clinically relevant perturbation. Figure 1 Figure 1. Disclosures Subklewe: Pfizer: Consultancy, Speakers Bureau; Takeda: Speakers Bureau; Klinikum der Universität München: Current Employment; Janssen: Consultancy; Seattle Genetics: Consultancy, Research Funding; Roche: Research Funding; Novartis: Consultancy, Research Funding, Speakers Bureau; MorphoSys: Research Funding; Miltenyi: Research Funding; Gilead: Consultancy, Research Funding, Speakers Bureau; Amgen: Consultancy, Research Funding, Speakers Bureau; BMS/Celgene: Consultancy, Research Funding, Speakers Bureau. von Bergwelt: Kite/Gilead: Honoraria, Research Funding, Speakers Bureau; Roche: Honoraria, Research Funding, Speakers Bureau; Novartis: Honoraria, Research Funding, Speakers Bureau; Astellas: Honoraria, Research Funding, Speakers Bureau; Miltenyi: Honoraria, Research Funding, Speakers Bureau; BMS: Honoraria, Research Funding, Speakers Bureau; Mologen: Honoraria, Research Funding, Speakers Bureau; MSD Sharpe & Dohme: Honoraria, Research Funding, Speakers Bureau. Weigert: Janssen: Speakers Bureau; Epizyme: Membership on an entity's Board of Directors or advisory committees; Roche: Research Funding.


2021 ◽  
pp. 103443
Author(s):  
Mohammad-Sadegh Razeghinia ◽  
Maryam Sahebazzamani ◽  
Saeed Soleymani ◽  
Ali-Asghar Ketabchi ◽  
Mohammad Moqaddasi-Amiri ◽  
...  
Keyword(s):  

Author(s):  
Sawsan Ahmed Nasr ◽  
Amel Abd El-Moneim Saad

Abstract Background Promoting cancer cells apoptosis is one of the effective methods to treat cancer. Human hepatocellular carcinoma (HepG2) and colorectal cancer (HCT-116) cell lines were used in the present study to evaluate the cytotoxic and anticancer properties of Nepeta septemcrenata Polysaccharide (NSP). Result Treatment of the two examined cells with NSP displayed a significant cytotoxicity towards HepG2 in a dose-dependent manner; meanwhile, its effect on HCT-116 was obtained under the influence of low doses. The quantitative real- time PCR (QRT-PCR) investigation revealed that NSP significantly up-regulated the expression levels of p53, p16, Fas, Fas-L, Bax, caspases-3, caspase-9, and TNF-α in association with down-regulation of cyclin D1, TERT, and BCL2. These findings declare the apoptotic characteristic of NSP.NSP, can also inhibit the development of cancer cells through the down-regulation of TGF-β and VEGF. Conclusions Our results suggested that the polysaccharides isolated from N. septemcrenata possess anticancer properties that could be explored for the development of novel anticancer agents.


2021 ◽  
Vol 19 ◽  
Author(s):  
John Paul Demosthenes ◽  
GnanaduraiJohn Fletcher ◽  
Uday George Zachariah ◽  
George Mannil Varghese ◽  
Susanne Alexander Pulimood ◽  
...  

Background : Chronic immune activation is one of the most widely recognized hallmarks of HIV infection. T-cells that express CD38+ and HLA-DR+ show poor proliferative potential, signal transduction, and increased apoptotic potential. This affects HIV pathogenesis and its outcome and further complicates with a coinfection like HBV. Method: Study Design: Cross-sectional. Blood samples were collected and analyzed for virological markers using ELISA for HBeAg and RT-PCR for HIV&HBV Viral load. Chronic immune activation markers of CD8+ and CD4+ T cells were measured by Flow cytometry for both HIV and HBV Results: There was a significant increase in HBV replication shown by higher HBV DNA (p=0.002), a higher proportion of HBeAg (p=0.0049), and lower CD4 counts (p=0.04) among HIV/HBV coinfected individuals, compared to the monoinfected groups. The frequencies of CD4+ CD38+ HLA-DR+ and CD8+ CD38+ HLA-DR+ in the HIV/HBV coinfection were significantly higher than HBV monoinfected group (P< 0.0001) and in the HIV monoinfected group (P < 0.0001). The Liver fibrosis score APRI and FIB-4, were higher in the coinfected group compared with HBV monoinfected group (0.67 vs 0.25, p = 0.0085; 3.48 vs 0.98, p = 0.0026), respectively. The cytokine levels of IL-17, Fas-L, TNF -α, IL-10, IL-2and Granzyme B were also measured and compared among the study groups. Conclusion: Our data suggest that HIV probably influences the immune activation of CD4+ and CD8+ T cells, and this may play a significant role in accelerating the disease outcome among HIV/HBV coinfected individuals.


2021 ◽  
Vol 11 ◽  
Author(s):  
Laurence Booth ◽  
Cameron West ◽  
Daniel Von Hoff ◽  
John M. Kirkwood ◽  
Paul Dent

We defined the lethal interaction between the novel therapeutic GZ17-6.02 and the standard of care combination of the MEK1/2 inhibitor trametinib and the B-RAF inhibitor dabrafenib in PDX isolates of cutaneous melanoma expressing a mutant B-RAF V600E protein. GZ17-6.02 interacted with trametinib/dabrafenib in an additive fashion to kill melanoma cells. Regardless of prior vemurafenib resistance, the drugs when combined interacted to prolong ATM S1981/AMPK T172 and eIF2α S51 phosphorylation and prolong the reduced phosphorylation of JAK2 Y1007, STAT3 Y705 and STAT5 Y694. In vemurafenib-resistant cells GZ17-6.02 caused a prolonged reduction in mTORC1 S2448, mTORC2 S2481 and ULK1 S757 phosphorylation; regardless of vemurafenib resistance, GZ17-6.02 caused a prolonged elevation in CD95 and FAS-L expression. Knock down of eIF2α, Beclin1, ATG5, ATM, AMPKα, CD95 or FADD significantly reduced the ability of GZ17-6.02 to kill as a single agent or when combined with the kinase inhibitors. Expression of activated mTOR, activated STAT3, activated MEK1 or activated AKT significantly reduced the ability of GZ17-6.02 to kill as a single agent or when combined with kinase inhibitors; protective effects that were significantly less pronounced in cells treated with trametinib/dabrafenib. Regardless of vemurafenib resistance, the drugs alone or in combination all reduced the expression of PD-L1 and increased the levels of MHCA, which was linked to degradation of multiple HDAC proteins. Our findings support the use of GZ17-6.02 in combination with trametinib/dabrafenib in the treatment of melanomas expressing mutant B-RAF V600E proteins.


2021 ◽  
Vol 11 ◽  
Author(s):  
Iva Arato ◽  
Domenico Milardi ◽  
Stefano Giovagnoli ◽  
Giuseppe Grande ◽  
Catia Bellucci ◽  
...  

Sertoli cells (SC) are immune privileged cells with the capacity of modulating the immune response by expressing several immune-regulatory factors. SC have the capacity to respond to external stimuli through innate phagocytic and antibacterial activities. This evidence evoked a potential role of SC as drug carriers and therapeutic agents. Such stimuli drive SC towards a still unknown evolution, the clinical relevance of which as yet remains undisclosed. This study sought to investigate the effects of external stimuli in the form of polymeric microparticles (MP) and bacteria derived endotoxins, such as lipopolysaccharides (LPS), in order to identify the pathways potentially involved in cell phenotype modifications. Compared to single stimulation, when combined, MP and LPS provoked a significant increase in the gene expression of IDO, PD-L1, FAS-L, TLR-3, TLR-4, MHC-II, ICAM-1, TFGβ1, BDF123, BDF129, BDF3 and pEP2C. Western Blotting analysis demonstrated up-regulation of the ERK 1–2 and NF-kB p65 phosphorylation ratios. Our study, showing the exponential increase of these mediators upon combined MP and LPS stimulation, suggests a “switch” of SC function from typical cells of the blood-testicular barrier to nonprofessional tolerogenic antigen-presenting cells. Further studies should target the clinical and technological implications of such stimuli-induced SC transformation.


Author(s):  
Hanqing Tang ◽  
Shitian Zhang ◽  
Cenhan Huang ◽  
Keming Li ◽  
Qiuhua Zhao ◽  
...  
Keyword(s):  

2020 ◽  
Vol 21 (1) ◽  
pp. 84-90
Author(s):  
Emine Erdag ◽  
Eda Becer ◽  
Yusuf Mulazim ◽  
Hafize Seda Vatansever ◽  
Hilal Kabadayı ◽  
...  

Background: 2(3H)-Benzoxazolone derivatives are preferential structural blocks in pharmacological probe designing with the possibility of modifications at various positions on the core structure. Benzoxazolones showed various biological activities such as analgesics, anti-inflammatory and anti-cancer. Objective: In the present work, we have prepared new Mannich bases of 2(3H)-benzoxazolone derivatives and evaluated their cytotoxicities and proapoptotic properties in MCF-7 breast cancer cell line. Methods: The structures of these compounds were characterized by FT-IR, elemental analysis, 1H and 13C NMR. Cytotoxicities of all the target compounds were investigated by MTT assay. Apoptotic properties of compounds were evaluated by immunocytochemistry using antibodies against caspase-3, cytochrome-c, FasL, and also TUNEL assay. Results: These two novel compounds, 1 and 2, both have the same piperazine substituent on the nitrogen atom of benzoxazolone and the main difference in the structures of these compounds is the presence of Cl substituent at the 5- position of the benzoxazolone ring. MTT results showed that compounds 1 and 2 were effective in terms of reduction of cell viability at 100μM and 50μM concentration for 48h, respectively. As a result of immunohistochemical staining, Fas L and caspase-3 immunoreactivities were significantly increased in MCF-7 cells after treatment with compound 1. Additionally, caspase-3 and cytochrome-c immunoreactivities were also increased significantly in MCF-7 cells after treatment with compound 2. The number of TUNEL positive cells was significantly higher in MCF-7 cells when compared with the control group after treatment with both compounds 1 and 2. Conclusion: It could be concluded that N-substituted benzoxazolone derivatives increase potential anti-cancer effects and they could be promising novel therapeutic agents for chemotherapy.


2020 ◽  
Author(s):  
Chen Wang ◽  
Rongrong Zhang ◽  
Qi Zhang ◽  
Huixiang Jin ◽  
Chenghua Wei ◽  
...  

Purpose: The purpose of our study was to investigate the profiles of inflammatory cytokines and the macrophage polarization gene in a choroidal neovascularization (CNV) mouse model before and after intravitreal aflibercept treatment. Methods: The CNV mouse model was conducted by laser photocoagulation. A total of 58 cytokines were measured by multiplex mouse cytokine antibody array. The macrophage polarization genes were tested by reverse transcription polymerase chain reaction. The relationship between the cytokines and the CNV lesion area was analyzed by correlation. Results: MIP-1a on day 3 after laser photocoagulation, MCP-5 and Fas-L on day 7, and IL-15 and IL-7 on day 14 were significantly upregulated (p< 0.001, fold change > 10.0). After the intravitreal aflibercept treatment, GM-CSF and MCP-1 on day 3 and TIMP-1 on days 7 and 14 were the most significantly upregulated cytokines (p< 0.001, fold change > 10.0). MIP-1 on day 3, IL-13 and Fas-L on day 7, and Fas-L on day 14 were the most significantly downregulated cytokines after intravitreal aflibercept treatment (p< 0.001, fold change > 5.0). M2 polarization and VEGFA genes were significantly increased in the CNV formation, whereas aflibercept suppressed M2 polarization and VEGFA genes. IL-7 was negatively related to the CNV lesion area on day 14 after intravitreal aflibercept treatment (r = −0.938, p = 0.006). Conclusion: The inflammatory cytokines and the M1/M2 macrophage genes significantly changed in the CNV mouse model. This result suggests that inflammatory cytokines and macrophages play a critical role in the physiopathology of CNV.


Sign in / Sign up

Export Citation Format

Share Document