Assessment of NK Cell Metabolism

Author(s):  
Molly P. Keppel ◽  
Megan A. Cooper
Keyword(s):  
Nk Cell ◽  
2019 ◽  
Vol 10 ◽  
Author(s):  
Iñigo Terrén ◽  
Ane Orrantia ◽  
Joana Vitallé ◽  
Olatz Zenarruzabeitia ◽  
Francisco Borrego

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Chloe Choi ◽  
David K. Finlay

AbstractImmunotherapy has ushered in an exciting new era for cancer treatment. The recent discovery and success of immune checkpoint blockade and chimeric antigen receptor (CAR) T cell adoptive cell transfer has raised interest in using other immune cells, including Natural Killer (NK) cells, which might overcome some limitations with CAR T cell therapy. In this review article, we discuss the evidence that cellular metabolism is crucial for NK cell effector function. Additionally, potential strategies to optimise the metabolism of therapeutic NK cells for improved function within the metabolically adverse tumour microenvironment will be explored.


eLife ◽  
2020 ◽  
Vol 9 ◽  
Author(s):  
Jessica F Walls ◽  
Jeff J Subleski ◽  
Erika M Palmieri ◽  
Marieli Gonzalez-Cotto ◽  
Clair M Gardiner ◽  
...  

Natural Killer (NK) cells have an important role in immune responses to viruses and tumours. Integrating changes in signal transduction pathways and cellular metabolism is essential for effective NK cells responses. The glycolytic enzyme Pyruvate Kinase Muscle 2 (PKM2) has described roles in regulating glycolytic flux and signal transduction, particularly gene transcription. While PKM2 expression is robustly induced in activated NK cells, mice lacking PKM2 in NK cells showed no defect in NK cell metabolism, transcription or antiviral responses to MCMV infection. NK cell metabolism was maintained due to compensatory PKM1 expression in PKM2-null NK cells. To further investigate the role of PKM2, we used TEPP-46, which increases PKM2 catalytic activity while inhibiting any PKM2 signalling functions. NK cells activated with TEPP-46 had reduced effector function due to TEPP-46-induced increases in oxidative stress. Overall, PKM2-regulated glycolytic metabolism and redox status, not transcriptional control, facilitate optimal NK cells responses.


2021 ◽  
Vol 12 ◽  
Author(s):  
Abrar Ul Haq Khan ◽  
Saeedah Musaed Almutairi ◽  
Alaa Kassim Ali ◽  
Rosalba Salcedo ◽  
C. Andrew Stewart ◽  
...  

Natural killer (NK) cells are the predominant innate lymphocytes that provide early defense against infections. In the inflammatory milieu, NK cells modify their metabolism to support high energy demands required for their proliferation, activation, and functional plasticity. This metabolic reprogramming is usually accompanied by the upregulation of nutrient transporter expression on the cell surface, leading to increased nutrient uptake required for intense proliferation. The interleukin-1 family members of inflammatory cytokines are critical in activating NK cells during infection; however, their underlying mechanism in NK cell metabolism is not fully elucidated. Previously, we have shown that IL-18 upregulates the expression of solute carrier transmembrane proteins and thereby induces a robust metabolic boost in NK cells. Unexpectedly, we found that IL-18 signaling is dispensable during viral infection in vivo, while the upregulation of nutrient transporters is primarily MyD88-dependent. NK cells from Myd88-/- mice displayed significantly reduced surface expression of nutrient receptors and mTOR activity during MCMV infection. We also identified that IL-33, another cytokine employing MyD88 signaling, induces the expression of nutrient transporters but requires a pre-exposure to IL-12. Moreover, signaling through the NK cell activating receptor, Ly49H, can also promote the expression of nutrient transporters. Collectively, our findings revealed multiple pathways that can induce the expression of nutrient transporters on NK cells while highlighting the imperative role of MyD88 in NK cell metabolism during infection.


2015 ◽  
Author(s):  
Guanqiao Li ◽  
Philip J. Vernon ◽  
Bennett van Houten ◽  
Xiaoyan Liang ◽  
Michael T. Lotze

2020 ◽  
Vol 14 (Supplement_1) ◽  
pp. S187-S188
Author(s):  
V Zaiatz Bittencourt ◽  
E Ryan ◽  
F Jones ◽  
G Doherty

Abstract Background Cell metabolism, in addition to providing energy and biochemical building blocks, also regulates immune cell function. We have increased our understanding of how natural killer cells (NK cell) metabolism impacts human NK cell function; however, little is known about NK cell role and metabolic changes in autoimmune diseases. The dysfunctional NK cell phenotype observed in pathological settings linked to defective cell metabolism will give insights into the development of new therapies for inflammatory diseases. IBD is an autoimmune disease of unknown origin characterised by severe gut inflammation and increased risk of colorectal cancer. NK cells can augment autoimmune responses through their activation of adaptive immune response by interaction with macrophages, dendritic cells and T cells; therefore, it is essential to decipher NK cells function in IBD to propose novel therapeutic targets. In this study, we defined the phenotype and metabolic profile of NK cells isolated from peripheral blood of IBD patients. Methods Blood samples were obtained from healthy donors and IBD patients from whom written consent had been obtained. PBMC were isolated by Lymphoprep (Axis-Shield) gradient. PBMC were stained with a combination of cell surface receptors, intracellular cytokines, proteins and analysed by flow cytometry. Cells were either stained ex vivo or after overnight stimulation with cytokine combination IL-12 + IL-15 and in the presence or absence of mTORC1 inhibitor rapamycin. Changes in OxPhos or glycolysis were detected by XFp extracellular flux analyser (Agilent Technologies). Mitochondria mass and polarisation were measured by flow cytometry utilising MitoTracker Green and MitoTracker Red, respectively (ThermoFisher). Results NK cells from IBD patients have impaired mTORC1 activity (a key signalling pathway regulating cell metabolism), which is associated with increased expression of pro-inflammatory cytokines IL-17 and TNF-α. Patients NK cells have dysfunctional mitochondria characterised by reduced mitochondria mass, low membrane potential and limited OXPHOS and glycolysis. Furthermore, patients NK cells have limited IFNγ production, reduced TRAIL expression and defective capability of killing cancer cells. Conclusion Taken together, our results strongly imply that NK cells from IBD patients have a distinct bioenergetic programme that favours the increased production of pro-inflammatory cytokines. This might be an important unknown defect of NK cells in the regulation of inflammation in IBD that can be used for the development of new treatments. Further studies need to be conducted to identify the reason for the observed defects and determination of other key metabolic pathways involved in NK-cell function during IBD.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A182-A182
Author(s):  
Tram Dao ◽  
Sandro Matosevic

BackgroundNatural killer (NK) cells have emerged as a viable alternative to T cells in adoptive cell transfer for cancer treatment. NK cell activity is driven by the balance between inhibitory and activating receptors, many of which remain elusive. In addition, NK cell metabolism is also a driver of NK cell fitness in tumor settings, where changes in NK metabolic states with the tumor microenvironment in vivo, or with stimulants ex vivo, further confounds the NK cells’ cytotoxic function in cancer settings. One receptor that lies at the intersection between NK cell function and metabolism is TIM-3, with its expression having consequences on NK cell cytokine production and glucose metabolism. However, the contribution of TIM-3 to NK cell anti-tumor immunity is unclear and its role in driving NK cell function so far not fully defined.MethodsNK cells were isolated from healthy adult peripheral blood and expanded in feeder-cell media. NK cell metabolism and function were evaluated by different flow cytometric assays to measure glucose uptake, cytotoxicity, degranulation, and cytokine production. TIM-3 knock-out cells were generated using the CRISPR-Cas9 system. Patient samples, including whole blood and tumor, were also processed and phenotyped to compare expression level with healthy donor samples.ResultsPreviously, we discovered that TIM-3 downregulation was associated with decreased cytokine production and target cytotoxicity, and that maintenance of expression above a certain threshold was needed for NK cell function. As cytokine production reflects immune cell metabolic state, we hypothesized that TIM-3 participates in regulation of ex vivo-activated NK cell metabolism, which in turn affect the production of the cytokine IFN- γ to sensitize cancer targets to NK cell-mediated lysis. Here, we report the consequences of glucose starvation on TIM-3 expression, and how knock-out of TIM-3 on human NK cells affects NK cell metabolism and functionalities against glioblastoma targets. We also cross-reference TIM-3 expression level with glioblastoma patient samples, which provide clinical context for microenvironmental cues and nutrient deprivation.ConclusionsOur findings suggest that TIM-3 expression is associated with both ex vivo-activated NK cell glucose metabolism and cytotoxic function against glioblastoma. As ex vivo-activated NK cells are considered to be highly glycolytic, and as such associated with higher cytotoxicity, TIM-3’s involvement with glucose uptake could prove crucial in sustaining NK cytotoxic phenotype in the tumor microenvironment. This information is shedding further light on the immunomodulatory roles of TIM-3, and aiding in leveraging this receptor usage in future NK cell-based immunotherapies.Ethics ApprovalAll procedures performed in studies involving human participants were approved by Purdue University’s Institutional Review Board (IRB) in August 2018 (#1804020540). All institutional safety and biosecurity procedures were adhered to.


2018 ◽  
Vol 200 (12) ◽  
pp. 3934-3941 ◽  
Author(s):  
Vanessa Zaiatz-Bittencourt ◽  
David K. Finlay ◽  
Clair M. Gardiner

Author(s):  
Nidhi Kedia-Mehta ◽  
Laura Tobin ◽  
Vanessa Zaiatz-Bittencourt ◽  
Marta Pisarska ◽  
Conor De Barra ◽  
...  

Natural killer (NK) cells are a population of innate immune cells which can rapidily kill cancer cells and produce cytokines such as interferon gamma (IFN-gamma). A key feature of NK cells is their ability to respond without prior sensitation, however it is now well established that NK cells can possess memory-like features. After activation with cytokines, NK cells demonstrate enhanced effector functions upon restimulation days or weeks later. This demonstrates that NK cells may be "trained" to be more effective killers and harnessed as more potent cancer immunotherapy agents. We have previously demonstrated that cellular metabolism is essential for NK cell responses, with NK cells upregulating both glycolysis and oxidative phosphorylation upon cytokine stimulation. Limiting NK cell metabolism results in reduced cytotoxicity and cytokine production. We have also demonstrated that defective NK cell responses in obesity are linked to defective cellular metabolism. In the current study we investigated if cellular metabolism is required during the initial period of NK cell cytokine training, and if NK cells from people with obesity (PWO) can be effectively trained. We show that increased flux through glycolysis and OXPHOS during the initial cytokine activation period is essential for NK cell training, as is the metabolic signalling factor Srepb. We show that NK cells from PWO, which are metabolically defective, display impaired NK cell training, which may have implications for immunotherapy in this particularly vulnerable group.


Sign in / Sign up

Export Citation Format

Share Document