Abstract B84: Decipher the role of IL-33 as an activator of NK cells’ antitumor activity

Author(s):  
Anais Eberhardt ◽  
Elena Blanc ◽  
Emilie Lardenois ◽  
Sarah Renaudineau ◽  
Jennifer Herbulot ◽  
...  
Keyword(s):  
Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 105-105
Author(s):  
Prasad Phatarpekar ◽  
Shiguo Zhu ◽  
Cecele J. Denman ◽  
Nam Nguyen-Jackson ◽  
Stephanie S. Watowich ◽  
...  

Abstract Abstract 105 Background: Signal Transducer and Activator of Transcription 3 (STAT3) activation plays an important role in carcinogenesis, and is recognized as a therapeutic target in many human malignancies. STAT3 was also found to be an important negative regulator in cellular immunity, and in some animal models inhibition of STAT3 has been effective in improving NK antitumor efficacy. However, there has been little direct assessment for the role of STAT3 signaling in human NK cells. We previously described robust ex vivo expansion and activation of NK cells by cocultivation with K562-based artificial antigen presenting cells (aAPC) genetically modified to express membrane bound IL-21 (mIL21). We found that mIL21 results in greater proliferation, longer telomere length, and less senescence than NK cells expanded with mIL15. Since STAT3 is predominantly activated by IL-21 whereas IL-15 predominantly activates STAT5, we hypothesized that activation of STAT3 plays a critical role in NK cell expansion and anti-tumor activity. We also found that the histone deacetylase (HDAC) inhibitor MS275 upregulates NKG2D expression, whereas the HDAC inhibitor valproic acid downregulates NKG2D expression. We found that STAT3 is constitutively phosphorylated in NK cells, and valproic acid, but not MS275, efficiently inhibits STAT3 tyrosine phosphorylation. This led us to hypothesize that constitutive STAT3 phosphorylation is required for NKG2D expression and NK cell antitumor activity. Objective: To assess the role of STAT3 phosphorylation in NK cell proliferation, NKG2D expression, and antitumor activity. Methods: In human NK cells, small molecule STAT3 inhibitors JSI-124 and S3I-201 were applied to block STAT3 phosphorylation. In mice, floxed STAT3 was deleted in hematopoietic cells by Cre expression under the Tie2 promoter. NK cell proliferation was induced by K562 aAPCs expressing mIL21. IL10 and IL21 were applied to induce STAT3 phosphorylation. STAT3 phosphorylation was detected by western blot and phospho-flow. NKG2D expression and NK cell cytoxicity were evaluated by flow cytometry and Calcein release assay. STAT3 binding to DNA upstream of NKG2D was assessed by ChIP assay. Statistical comparison was performed by paired Student's t test using GraphPad Prism. Results: Analysis of phosphorylated STAT3 in NK cells by phospho-flow showed increased levels of activated STAT3 in NK cells stimulated with mIL-21 compared to mIL-15. We found that inhibition of STAT3 strongly suppressed the expansion of NK cells and significantly reduced cytotoxicity of expanded NK cells. We found that NKG2D surface expression on NK cells was significantly downregulated by STAT3 inhibitors, and accordingly, NK cell mediated killing activity was decreased. Conversely, STAT3 activation in primary NK cells by IL10 and IL21 upregulated NKG2D expression, resulting in increased NK cell mediated tumor lysis that could be reversed by small molecule STAT3 inhibition. We then analyzed NKG2D expression on NK cells in mice, and found that NKG2D expression in STAT3 knockouts was significantly lower than on wild type mice. By ChIP assay, we found that phosphorylated STAT3 directly binds upstream of NKG2D. IL10 and IL21 increased this binding, and STAT3 inhibition decreased this binding. Conclusions: Our finding of an immune-activating role for STAT3 in NK cells differs from previous reports, and is for the first to show that NK cell proliferation, NKG2D expression, and cytotoxicity is regulated by STAT3 phosphorylation. This novel role for STAT3 signaling may be important in the application of targeted STAT3 therapies and in improving NK cell therapeutic efficacy for patients with cancer. Work is in progress to delineate downstream signaling of STAT3 in NK cells. Disclosures: No relevant conflicts of interest to declare.


Planta Medica ◽  
2006 ◽  
Vol 72 (15) ◽  
pp. 1415-1417 ◽  
Author(s):  
Hiroko Maruyama ◽  
Hidekazu Tamauchi ◽  
Mariko Iizuka ◽  
Takahisa Nakano

2018 ◽  
Vol 56 (01) ◽  
pp. E2-E89
Author(s):  
C Rennert ◽  
C Tauber ◽  
B Zecher ◽  
A Schuch ◽  
M Hofmann ◽  
...  
Keyword(s):  

Cancers ◽  
2021 ◽  
Vol 13 (8) ◽  
pp. 1802
Author(s):  
Nayoung Kim ◽  
Mi Yeon Kim ◽  
Woo Seon Choi ◽  
Eunbi Yi ◽  
Hyo Jung Lee ◽  
...  

Natural killer (NK) cells are innate cytotoxic lymphocytes that provide early protection against cancer. NK cell cytotoxicity against cancer cells is triggered by multiple activating receptors that recognize specific ligands expressed on target cells. We previously demonstrated that glycogen synthase kinase (GSK)-3β, but not GSK-3α, is a negative regulator of NK cell functions via diverse activating receptors, including NKG2D and NKp30. However, the role of GSK-3 isoforms in the regulation of specific ligands on target cells is poorly understood, which remains a challenge limiting GSK-3 targeting for NK cell-based therapy. Here, we demonstrate that GSK-3α rather than GSK-3β is the primary isoform restraining the expression of NKG2D ligands, particularly ULBP2/5/6, on tumor cells, thereby regulating their susceptibility to NK cells. GSK-3α also regulated the expression of the NKp30 ligand B7-H6, but not the DNAM-1 ligands PVR or nectin-2. This regulation occurred independently of BCR-ABL1 mutation that confers tyrosine kinase inhibitor (TKI) resistance. Mechanistically, an increase in PI3K/Akt signaling in concert with c-Myc was required for ligand upregulation in response to GSK-3α inhibition. Importantly, GSK-3α inhibition improved cancer surveillance by human NK cells in vivo. Collectively, our results highlight the distinct role of GSK-3 isoforms in the regulation of NK cell reactivity against target cells and suggest that GSK-3α modulation could be used to enhance tumor cell susceptibility to NK cells in an NKG2D- and NKp30-dependent manner.


2021 ◽  
Vol 7 (8) ◽  
pp. eabc2331 ◽  
Author(s):  
Jose M. Ayuso ◽  
Shujah Rehman ◽  
Maria Virumbrales-Munoz ◽  
Patrick H. McMinn ◽  
Peter Geiger ◽  
...  

Solid tumors generate a suppressive environment that imposes an overwhelming burden on the immune system. Nutrient depletion, waste product accumulation, hypoxia, and pH acidification severely compromise the capacity of effector immune cells such as T and natural killer (NK) cells to destroy cancer cells. However, the specific molecular mechanisms driving immune suppression, as well as the capacity of immune cells to adapt to the suppressive environment, are not completely understood. Thus, here, we used an in vitro microfluidic tumor-on-a-chip platform to evaluate how NK cells respond to the tumor-induced suppressive environment. The results demonstrated that the suppressive environment created by the tumor gradually eroded NK cell cytotoxic capacity, leading to compromised NK cell surveillance and tumor tolerance. Further, NK cell exhaustion persisted for an extended period of time after removing NK cells from the microfluidic platform. Last, the addition of checkpoint inhibitors and immunomodulatory agents alleviated NK cell exhaustion.


Cancers ◽  
2021 ◽  
Vol 13 (9) ◽  
pp. 2184
Author(s):  
Valentina Cazzetta ◽  
Sara Franzese ◽  
Claudia Carenza ◽  
Silvia Della Bella ◽  
Joanna Mikulak ◽  
...  

Natural killer (NK) and dendritic cells (DCs) are innate immune cells that play a crucial role in anti-tumor immunity. NK cells kill tumor cells through direct cytotoxicity and cytokine secretion. DCs are needed for the activation of adaptive immune responses against tumor cells. Both NK cells and DCs are subdivided in several subsets endowed with specialized effector functions. Crosstalk between NK cells and DCs leads to the reciprocal control of their activation and polarization of immune responses. In this review, we describe the role of NK cells and DCs in liver cancer, focusing on the mechanisms involved in their reciprocal control and activation. In this context, intrahepatic NK cells and DCs present unique immunological features, due to the constant exposure to non-self-circulating antigens. These interactions might play a fundamental role in the pathology of primary liver cancer, namely hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). Additionally, the implications of these immune changes are relevant from the perspective of improving the cancer immunotherapy strategies in HCC and ICC patients.


Author(s):  
Luis Sánchez-del-Campo ◽  
Román Martí-Díaz ◽  
María F. Montenegro ◽  
Rebeca González-Guerrero ◽  
Trinidad Hernández-Caselles ◽  
...  

Abstract Background The application of immune-based therapies has revolutionized cancer treatment. Yet how the immune system responds to phenotypically heterogeneous populations within tumors is poorly understood. In melanoma, one of the major determinants of phenotypic identity is the lineage survival oncogene MITF that integrates diverse microenvironmental cues to coordinate melanoma survival, senescence bypass, differentiation, proliferation, invasion, metabolism and DNA damage repair. Whether MITF also controls the immune response is unknown. Methods By using several mouse melanoma models, we examine the potential role of MITF to modulate the anti-melanoma immune response. ChIP-seq data analysis, ChIP-qPCR, CRISPR-Cas9 genome editing, and luciferase reporter assays were utilized to identify ADAM10 as a direct MITF target gene. Western blotting, confocal microscopy, flow cytometry, and natural killer (NK) cytotoxicity assays were used to determine the underlying mechanisms by which MITF-driven phenotypic plasticity modulates melanoma NK cell-mediated killing. Results Here we show that MITF regulates expression of ADAM10, a key sheddase that cleaves the MICA/B family of ligands for NK cells. By controlling melanoma recognition by NK-cells MITF thereby controls the melanoma response to the innate immune system. Consequently, while melanoma MITFLow cells can be effectively suppressed by NK-mediated killing, MITF-expressing cells escape NK cell surveillance. Conclusion Our results reveal how modulation of MITF activity can impact the anti-melanoma immune response with implications for the application of anti-melanoma immunotherapies.


2021 ◽  
Vol 22 (2) ◽  
pp. 656
Author(s):  
Hantae Jo ◽  
Byungsun Cha ◽  
Haneul Kim ◽  
Sofia Brito ◽  
Byeong Mun Kwak ◽  
...  

Natural killer (NK) cells are lymphocytes that can directly destroy cancer cells. When NK cells are activated, CD56 and CD107a markers are able to recognize cancer cells and release perforin and granzyme B proteins that induce apoptosis in the targeted cells. In this study, we focused on the role of phytoncides in activating NK cells and promoting anticancer effects. We tested the effects of several phytoncide compounds on NK-92mi cells and demonstrated that α-pinene treatment exhibited higher anticancer effects, as observed by the increased levels of perforin, granzyme B, CD56 and CD107a. Furthermore, α-pinene treatment in NK-92mi cells increased NK cell cytotoxicity in two different cell lines, and immunoblot assays revealed that the ERK/AKT pathway is involved in NK cell cytotoxicity in response to phytoncides. Furthermore, CT-26 colon cancer cells were allografted subcutaneously into BALB/c mice, and α-pinene treatment then inhibited allografted tumor growth. Our findings demonstrate that α-pinene activates NK cells and increases NK cell cytotoxicity, suggesting it is a potential compound for cancer immunotherapy.


2021 ◽  
Vol 92 ◽  
pp. 107361
Author(s):  
Khadijeh Dizaji Asl ◽  
Kobra Velaei ◽  
Ali Rafat ◽  
Hamid Tayefi Nasrabadi ◽  
Ali Akbar Movassaghpour ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document