scholarly journals Transcription factor KLF2 regulates homeostatic NK cell proliferation and survival

2016 ◽  
Vol 113 (19) ◽  
pp. 5370-5375 ◽  
Author(s):  
Whitney Rabacal ◽  
Sudheer K. Pabbisetty ◽  
Kristen L. Hoek ◽  
Delphine Cendron ◽  
Yin Guo ◽  
...  

Natural killer (NK) cells are innate lymphocytes that recognize and lyse virally infected or transformed cells. This latter property is being pursued in clinics to treat leukemia with the hope that further breakthroughs in NK cell biology can extend treatments to other cancers. At issue is the ability to expand transferred NK cells and prolong their functionality within the context of a tumor. In terms of NK cell expansion and survival, we now report that Kruppel-like factor 2 (KLF2) is a key transcription factor that underpins both of these events. Excision of Klf2 using gene-targeted mouse models promotes spontaneous proliferation of immature NK cells in peripheral tissues, a phenotype that is replicated under ex vivo conditions. Moreover, KLF2 imprints a homeostatic migration pattern on mature NK cells that allows these cells to access IL-15–rich microenvironments. KLF2 accomplishes this feat within the mature NK cell lineage via regulation of a subset of homing receptors that respond to homeostatic ligands while leaving constitutively expressed receptors that recognize inflammatory cytokines unperturbed. Under steady-state conditions, KLF2-deficient NK cells alter their expression of homeostatic homing receptors and subsequently undergo apoptosis due to IL-15 starvation. This novel mechanism has implications regarding NK cell contraction following the termination of immune responses including the possibility that retention of an IL-15 transpresenting support system is key to extending NK cell activity in a tumor environment.

2021 ◽  
Vol 12 ◽  
Author(s):  
Umut Can Kucuksezer ◽  
Esin Aktas Cetin ◽  
Fehim Esen ◽  
Ilhan Tahrali ◽  
Nilgun Akdeniz ◽  
...  

Natural killer (NK) cells, the large granular lymphocytes differentiated from the common lymphoid progenitors, were discovered in early 1970’s. They are members of innate immunity and were initially defined by their strong cytotoxicity against virus-infected cells and by their important effector functions in anti-tumoral immune responses. Nowadays, NK cells are classified among the recently discovered innate lymphoid cell subsets and have capacity to influence both innate and adaptive immune responses. Therefore, they can be considered as innate immune cells that stands between the innate and adaptive arms of immunity. NK cells don’t express T or B cell receptors and are recognized by absence of CD3. There are two major subgroups of NK cells according to their differential expression of CD16 and CD56. While CD16+CD56dim subset is best-known by their cytotoxic functions, CD16-CD56bright NK cell subset produces a bunch of cytokines comparable to CD4+ T helper cell subsets. Another subset of NK cells with production of interleukin (IL)-10 was named as NK regulatory cells, which has suppressive properties and could take part in immune-regulatory responses. Activation of NK cells is determined by a delicate balance of cell-surface receptors that have either activating or inhibitory properties. On the other hand, a variety of cytokines including IL-2, IL-12, IL-15, and IL-18 influence NK cell activity. NK-derived cytokines and their cytotoxic functions through induction of apoptosis take part in regulation of the immune responses and could contribute to the pathogenesis of many immune mediated diseases including ankylosing spondylitis, Behçet’s disease, multiple sclerosis, rheumatoid arthritis, psoriasis, systemic lupus erythematosus and type-1 diabetes. Dysregulation of NK cells in autoimmune disorders may occur through multiple mechanisms. Thanks to the rapid developments in biotechnology, progressive research in immunology enables better characterization of cells and their delicate roles in the complex network of immunity. As NK cells stand in between innate and adaptive arms of immunity and “bridge” them, their contribution in inflammation and immune regulation deserves intense investigations. Better understanding of NK-cell biology and their contribution in both exacerbation and regulation of inflammatory disorders is a requisite for possible utilization of these multi-faceted cells in novel therapeutic interventions.


Immunity ◽  
2020 ◽  
Vol 52 (6) ◽  
pp. 1075-1087.e8 ◽  
Author(s):  
Jing Ni ◽  
Xi Wang ◽  
Ana Stojanovic ◽  
Qin Zhang ◽  
Marian Wincher ◽  
...  

2019 ◽  
Vol 47 (04) ◽  
pp. 823-839 ◽  
Author(s):  
Dae-Young Lee ◽  
Chan Woong Park ◽  
Sue Jung Lee ◽  
Hye-Ryung Park ◽  
Dae Bang Seo ◽  
...  

Ginseng root has been used in traditional oriental medicine for the enhancement of immune system function. The immunostimulatory effects of ginseng berry polysaccharides, however, remain unclear. Effects of polysaccharides from ginseng berry on the activation of natural killer (NK) cells and inhibition of tumors are reported. A crude polysaccharide was isolated from ginseng berry as a ginseng berry polysaccharide portion (GBPP) and was further fractionated using gel filtration chromatography to obtain the three polysaccharide fractions GBPP-I, -II and -III. GBPP-I consisted of mainly galactose (46.9%) and arabinose (27.5%). GBPP-I showed a high dose-dependent anticomplementary activity. Stimulation of murine peritoneal macrophages by GBPP-I showed the greatest enhancement of interleukin (IL)-6 and IL-12 and tumor necrosis factor (TNF)-[Formula: see text] production. In addition, an ex vivo assay of natural killer (NK) cell activity showed that oral ([Formula: see text] administration of GBPP-I significantly increased NK cell cytotoxicity in YAC-1 tumor cells and production of granzyme B. Prophylactic intravenous ([Formula: see text] and [Formula: see text] administration of GBPP-I significantly and dose-dependently inhibited lung metastatic activity in B16BL6 melanoma cells. Depletion of NK cells after injection of rabbit anti-asialo GM1 partially abolished the inhibitory effect of GBPP-I on lung metastasis, indicating that NK cells play an important role in anticancer effects. GBPP-I exerts a strong immune-enhancing activity and can prevent cancer metastasis through activation of NK cells and other immune-related cells.


2021 ◽  
Vol 8 ◽  
Author(s):  
William C. Kisseberth ◽  
Dean A. Lee

Osteosarcoma is the most common primary bone tumor in both humans and dogs. It is a highly metastatic cancer and therapy has not improved significantly since the inclusion of adjuvant chemotherapy into disease treatment strategies. Osteosarcoma is an immunogenic tumor, and thus development of immunotherapies for its treatment, especially treatment of microscopic pulmonary metastases might improve outcomes. NK cells are lymphocytes of the innate immune system and can recognize a variety of stressed cells, including cancer cells, in the absence of major histocompatibility complex (MHC)-restricted receptor ligand interactions. NK cells have a role in controlling tumor progression and metastasis and are important mediators of different therapeutic interventions. The core hypothesis of adoptive natural killer (NK) cell therapy is there exists a natural defect in innate immunity (a combination of cancer-induced reduction in NK cell numbers and immunosuppressive mechanisms resulting in suppressed function) that can be restored by adoptive transfer of NK cells. Here, we review the rationale for adoptive NK cell immunotherapy, NK cell biology, TGFβ and the immunosuppressive microenvironment in osteosarcoma, manufacturing of ex vivo expanded NK cells for the dog and provide perspective on the present and future clinical applications of adoptive NK cell immunotherapy in spontaneous osteosarcoma and other cancers in the dog.


2019 ◽  
Vol 6 ◽  
pp. 33-42
Author(s):  
Marta Kłopotowska ◽  
Magdalena Winiarska

Immunooncology gained great attention in cancer treatment over the last few years. Adoptive cell therapy is a type of immunotherapy that uses cytotoxic lymphocytes or NK cells. NK cells precisely recognize and kill cancer cells without affecting healthy cells. These properties make NK cells an attractive strategy with potential therapeutic applications. NK cells can be further modified with chimeric antigen receptors (CAR). Currently, several dozen clinical trials with NK cells are registered for cancer treatment, including several CAR-NK cells or NK-92 cell line modified with CAR. Despite the undeniable success of immunotherapy in recent years, many concerns still remain unresolved. One of the challenges for the effective immunotherapy is the immunosuppressive tumor environment. Numerous studies indicate that persistent oxidative stress, an important element associated with cancer, inhibits NK cell activity. However, the exact mechanisms responsible for protecting NK cells against suppressive oxidative stress are not identified. Therefore, in my doctoral dissertation co-financed by the Scientific Polpharma Foundation doctoral scholarship I focused on investigating and identifying mechanisms protecting NK cells against oxidative stress. The obtained results indicate that NK cells are the most sensitive to the oxidative stress, when compared withother immune cells. Moreover, inhibition of PRDX1-TXN1-TXNRD1 system impairs NK cell effector functions, while the increase of PRDX1 significantly improves the survival of NK cells and CAR-NK cells in the presence of H<sub>2</sub>O<sub>2</sub>, resulting in improved effectiveness of NK cells under the oxidative stress conditions.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2536-2536
Author(s):  
Christian M. Capitini ◽  
Joanna L. Meadors ◽  
Monica M. Cho ◽  
Rimas J. Orentas ◽  
Crystal L. Mackall ◽  
...  

Abstract Abstract 2536 Methods to expand natural killer (NK) cells ex vivo for adoptive cell therapy are being explored to improve outcomes after allogeneic blood and marrow transplant (alloBMT). Artificial antigen presenting cells (aAPCs) can present cytokines and/or co-stimulatory molecules that can potentially improve expansion and activity. 4-1BBL (CD137L) has demonstrated mixed results on murine and human NK cells, but the impact on murine NK cell biology after alloBMT has not been explored. NK cells were harvested from either C57BL/6 (B6) or CB6F1 spleens and cultured ex vivo with a recombinant interleukin (IL)-15/IL-15 receptor alpha (Ra) complex in the presence or absence of a CD137L+ aAPC. Because IL-15 is typically presented in trans by IL-15Ra, the complex was utilized to potently increase agonist bioactivity. NK cells cultured with IL-15/IL-15Ra alone showed a peak of 20-fold expansion, but this expansion was decreased with the addition of CD137L+ aAPCs if the ratio of aAPC to NK cells was greater than 1:1. In the presence of IL-15/IL-15Ra, the impact of CD137L+ aAPCs on expression of the inhibitory receptors, Ly49C+I and activating receptor Ly49H was variable and strain dependent, with increased expression in B6 NK cells, but decreased expression in CB6F1 NK cells. The expression of major histocompatibility complex (MHC) class I was not affected in NK cells from either strain by the presence of CD137L+ aAPCs. The production of gamma interferon and tumor necrosis factor-a was robust in NK cells expanded by IL-15/IL-15Ra alone, but attenuated with the addition of CD137L+ aAPCs. Animal experiments showed that administration of NK cells expanded ex vivo with IL-15/IL-15Ra alone was well tolerated after T cell depleted MHC-mismatched alloBMT (CB6F1–>B6), but surprisingly the addition of CD137L+ aAPCs to cultures caused NK cells to induce GVHD-associated weight loss. In summary, IL-15/IL-15Ra expanded murine NK cells demonstrate increased cytokine production and do not cause toxicity when infused after alloBMT. The presence of CD137L+ aAPCs attenuated cytokine production and increased Ly49 receptor expression in NK cells from B6 mice. Remarkably, NK cells expanded by IL-15/IL-15Ra in the presence of CD137L+ aAPCs demonstrate increased propensity to cause GVHD. Ongoing studies are exploring the anti-tumor efficacy of IL-15/IL-15Ra expanded murine NK cells cultured in the presence and absence of CD137L. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5389-5389
Author(s):  
Susann Szmania ◽  
Amy D Greenway ◽  
Joshuah D Lingo ◽  
Katie Stone ◽  
Junaid Khan ◽  
...  

Abstract CS1 is an ideal target for multiple myeloma (MM) therapy as it is highly expressed on MM while having a very limited expression profile in normal tissues.  Elotuzumab (elo), a humanized monoclonal antibody (mAb) targeting CS1, has an acceptable safety profile and clinical activity in relapsed/refractory MM when combined with the immune modulator lenalidomide (len) and low dose dexamethasone (dex).  The primary mechanism of action for elo is NK cell-mediated antibody-dependent cellular cytotoxicity.  Here we report on a patient who was given elo/len via a single patient IND 6 months after receiving therapy with ex vivo activated auto-ENK cells and low dose IL2 as previously described (Szmania et al, Blood ASH Annual Meeting Abstracts 2012;120:1912).  The patient had relapsing GEP70 high-risk MM with cytogenetic abnormalities and had failed multiple lines of prior therapy including 3 auto-PBSC transplants and further salvage treatments including len, bortezomib, pomalidomide and carfilzomib.   Although ENK cell therapy did not induce a response, subsequent disease progression was slow.  IV elo was started 187 days after ENK cell infusion, and given every 14 days at the currently studied dose of 10mg/kg.  Len at 15mg/day was given on days 1-21 of a 28-day cycle.  Dex premedication (p.o. 38mg; IV, 10mg) was added after a grade 2 infusion reaction was observed to elo dose #1. While on the ENK cell protocol this patient had a dramatic increase in circulating NK cell counts peaking 9 days after infusion (6300 NK/µL, a 48-fold increase from baseline). Although still in the high range, NK cell levels at the time of elo treatment had normalized somewhat (539 NK/ml), and the cell surface expression of key activating receptors was consistent with a resting phenotype.  NK cell count remained stable after the first dose of elo (530 NK/ml) but subsequently dipped to 179 NK/µL after elo dose #2.   Since dex has been reported to affect NK cell counts, it is important to note that an additional dose was taken prior to elo dose #2 due to a travel delay (in total 66 mg of dex was taken on this occasion).  Circulating NK cells (collected pre-elo and 11, 25, and 57 days after elo dose #1) had similar low activity against auto-MM collected prior to elo treatment (effector:target ratio 10:1, 0-5% specific lysis) and killing against MM collected after 5 elo doses was only modestly increased (3-12%).  However, the same circulating NK cells exhibited significantly increased cytolytic ability when additional elo (10µg/mL) was added during the in vitro E:T co-incubation (3-11 fold increase in killing over isotype control, p=0.0008) suggesting that the MM targets were not saturated with mAb. Bound mAb may have been reduced in part during target cell isolation and freeze/thaw.  Freshly prepared auto-ENK cells exhibited an activated immunophenotype and induced significantly higher killing of pre-elo MM (45%) compared to non-expanded NK.  ENK killing was higher still against MM collected after 5 doses of elo (61%).  When elo was present during the assay, ENK demonstrated the most effective killing of auto-MM, reaching levels  equivalent to that of the NK sensitive target K562 (85% vs. 82% lysis).  Successful mAb therapy for MM is now moving forward as target antigens with selective, high and homogeneous expression, such as CS1, are identified.  However, the activity of responding effector cells is a critical issue to consider.  Inadequate NK cell count and activity level has been reported in MM and steroids typically given to debulk and preempt mAb-induced infusion reactions may exacerbate this problem.  Immunomodulatory agents given to enhance immune cell activity are not sufficient to reverse the negative effect of steroids.  We have previously shown that large doses of highly activated auto-ENK cells can be safely infused and that these cells expand further after infusion.  In this study we show that ENK cells have significant activity in vitro against auto-MM and that elo further enhances this activity.  Combination therapy incorporating saturating doses of mAb followed by infusion of NK effector cells with optimized activity against auto-MM is an innovative approach that warrants investigation.  Infusing highly activated effector cells after dex/elo may be one way to reap the benefits of combining these modalities while circumventing steroid-induced immune suppression. Disclosures: Barlogie: Celgene: Consultancy, Honoraria, Research Funding; Myeloma Health, LLC: Patents & Royalties.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A718-A718
Author(s):  
Md Faqrul Hasan ◽  
Alicja Copik

BackgroundNatural killer (NK) cells are innate immune cells that directly kill and coordinate responses against cancer prompting interest in using ex vivo expanded NK cells as an adoptive cell therapy for treatment of cancer. NK cells express a set of activating and inhibitory receptors that regulate their activity. Inhibitory receptor TIGIT (T cell Immunoreceptor with Ig and ITIM domain) is upregulated on intratumoral NK cells in some cancers, inhibits NK cell activity and promotes NK cell exhaustion. In this study, the effect of TIGIT blockade on the anti-tumor activities of ex vivo expanded NK cells was evaluated.MethodsNK cells were activated overnight with cytokines or ex vivo expanded with PM21-particles. Their TIGIT expression was determined with qRT-PCR and flow cytometry. Cytotoxicity was assessed by kinetic, imaging-based assay (Incucyte S3) against A549 and NCI-H1299 cells cultured in 3D. Cytotoxicity was calculated based on untreated controls at different time-points. Results from multiple donors were normalized to cytotoxicity of NK cells with isotype for individual donors and was compared to the cytotoxicity of NK cells with anti-TIGIT. Unpaired t test was used to determine statistical significance. K562 cells stably expressing Polio Virus Receptor (PVR), were used to restimulate A549 spheroid-exposed NK cells to measure IFNγ, TNFα and degranulation. Furthermore, phenotypic changes of NK cells upon TIGIT blockade were examined by analyzing a set of activating and inhibitory receptors by flow cytometry.ResultsThe effect of NK cell expansion/activation on TIGIT expression was assessed. TIGIT was upregulated on expanded and cytokine-activated NK cells both on mRNA and protein level. The effect of TIGIT blockade on NK cell cytotoxicity was examined by co-culturing PM21-NK cells with cancer cells in the presence of anti-TIGIT antibodies or respective isotypes. TIGIT blockade significantly increased cytotoxicity of PM21-NK cells against A549 (1.3 fold, P < 0.0001) and NCI-H1299 (1.3 fold, P = 0.0003) spheroids after 48 h. To access exhaustion, NK cells exposed to A549 spheroids for 7 days were restimulated with PVR+ K562 cells. TIGIT blockade prevented NK cell exhaustion resulting in increased expression of IFNγ, TNFα and surface CD107a on restimulated NK cells. TIGIT blockade did not result in changes to the surface phenotype of NK cells.ConclusionsTIGIT was highly expressed on expanded and cytokine-activated NK cells. TIGIT blockade improved anti-tumor activities of PM21-NK cells. Thus, PM21-NK cells and TIGIT antibodies have translational potential as a combination therapy to improve anti-tumor response.


2019 ◽  
Vol 20 (14) ◽  
pp. 3472 ◽  
Author(s):  
Monika Holubova ◽  
Martin Leba ◽  
Hana Gmucova ◽  
Valentina S. Caputo ◽  
Pavel Jindra ◽  
...  

Relapsed acute myeloid leukemia (AML) is a significant post-transplant complication lacking standard treatment and associated with a poor prognosis. Cellular therapy, which is already widely used as a treatment for several hematological malignancies, could be a potential treatment alternative. Natural killer (NK) cells play an important role in relapse control but can be inhibited by the leukemia cells highly positive for HLA class I. In order to restore NK cell activity after their ex vivo activation, NK cells can be combined with conditioning target cells. In this study, we tested NK cell activity against KG1a (AML cell line) with and without two types of pretreatment—Ara-C treatment that induced NKG2D ligands (increased activating signal) and/or blocking of HLA–KIR (killer-immunoglobulin-like receptors) interaction (decreased inhibitory signal). Both treatments improved NK cell killing activity. Compared with target cell killing of NK cells alone (38%), co-culture with Ara-C treated KG1a target cells increased the killing to 80%. Anti-HLA blocking antibody treatment increased the proportion of dead KG1a cells to 53%. Interestingly, the use of the combination treatment improved the killing potential to led to the death of 85% of KG1a cells. The combination of Ara-C and ex vivo activation of NK cells has the potential to be a feasible approach to treat relapsed AML after hematopoietic stem cell transplantation.


Parasitology ◽  
2011 ◽  
Vol 138 (14) ◽  
pp. 1898-1909 ◽  
Author(s):  
THORSTEN LIEKE ◽  
SUSANNE NYLÉN ◽  
LIV EIDSMO ◽  
CHRISTEL SCHMETZ ◽  
LOUISE BERG ◽  
...  

SUMMARYNK cells represent one of the first lines of defence in the immune reaction after invasion ofLeishmaniaparasites. Depletion of mouse natural killer (NK) cells dramatically enhances susceptibility of normally resistant mice. In this study we evaluated the fate of NK cells and parasites after contact formation. The hydrophilic fluorescent dye CMFDA (chloro-methylfluorescin diacetate) that allows analysis of cytotoxicity in flow cytometry and microscopy was used. Furthermore, these findings were confirmed with scanning and transmission electron microscopy. Direct contact points were found betweenLeishmaniapromastigotes and naïve human NK cells. These contacts were associated with transfer of cytosol by membrane bridges and cytotoxicity of NK cells againstLeishmania. However, in contrast to other target cells which allow repeated exocytosis of lytic granules, contact withLeishmaniacauses immediate destruction of NK cells in a non-apoptotic way. Our results give a reasonable explanation forex vivoobservations of reduced NK cell numbers and impaired NK response in patients with acute cutaneous leishmaniasis. Animal models have clearly shown that NK cells play a key role in the induction and direction of the immune response. Thus inhibition of NK cells at the onset of infection would be advantageous for the survival of the parasite.


Sign in / Sign up

Export Citation Format

Share Document