cytotoxic function
Recently Published Documents


TOTAL DOCUMENTS

157
(FIVE YEARS 28)

H-INDEX

34
(FIVE YEARS 4)

Author(s):  
Haohao Zhang ◽  
Yiming Hu ◽  
Dandan Liu ◽  
Zhi Liu ◽  
Ningxia Xie ◽  
...  

AbstractIntestinal intraepithelial lymphocytes (IELs) are distributed along the length of the intestine and are considered the frontline of immune surveillance. The precise molecular mechanisms, especially epigenetic regulation, of their development and function are poorly understood. The trimethylation of histone 3 at lysine 27 (H3K27Me3) is a kind of histone modifications and associated with gene repression. Kdm6b is an epigenetic enzyme responsible for the demethylation of H3K27Me3 and thus promotes gene expression. Here we identified Kdm6b as an important intracellular regulator of small intestinal IELs. Mice genetically deficient for Kdm6b showed greatly reduced numbers of TCRαβ+CD8αα+ IELs. In the absence of Kdm6b, TCRαβ+CD8αα+ IELs exhibited increased apoptosis, disturbed maturation and a compromised capability to lyse target cells. Both IL-15 and Kdm6b-mediated demethylation of histone 3 at lysine 27 are responsible for the maturation of TCRαβ+CD8αα+ IELs through upregulating the expression of Gzmb and Fasl. In addition, Kdm6b also regulates the expression of the gut-homing molecule CCR9 by controlling H3K27Me3 level at its promoter. However, Kdm6b is dispensable for the reactivity of thymic precursors of TCRαβ+CD8αα+ IELs (IELPs) to IL-15 and TGF-β. In conclusion, we showed that Kdm6b plays critical roles in the maturation and cytotoxic function of small intestinal TCRαβ+CD8αα+ IELs.


Cancers ◽  
2021 ◽  
Vol 13 (23) ◽  
pp. 5967
Author(s):  
Nicolas Jacquelot ◽  
Maryam Ghaedi ◽  
Kathrin Warner ◽  
Douglas C. Chung ◽  
Sarah Q. Crome ◽  
...  

Immune checkpoints (IC) are broadly characterized as inhibitory pathways that tightly regulate the activation of the immune system. These molecular “brakes” are centrally involved in the maintenance of immune self-tolerance and represent a key mechanism in avoiding autoimmunity and tissue destruction. Antibody-based therapies target these inhibitory molecules on T cells to improve their cytotoxic function, with unprecedented clinical efficacies for a number of malignancies. Many of these ICs are also expressed on innate lymphoid cells (ILC), drawing interest from the field to understand their function, impact for anti-tumor immunity and potential for immunotherapy. In this review, we highlight ILC specificities at different tissue sites and their migration potential upon inflammatory challenge. We further summarize the current understanding of IC molecules on ILC and discuss potential strategies for ILC modulation as part of a greater anti-cancer armamentarium.


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.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A218-A218
Author(s):  
Daniel Corey ◽  
Sunil Thomas ◽  
Brandon Cieniewicz ◽  
Linh Nguyen ◽  
Jared Clever ◽  
...  

BackgroundActivated T cells have limited antigen presenting capability due to inefficient capture.1 This process can be enhanced through novel chimeric engulfment receptors (CERs) expressing a human Tim-4 phagocyte receptor that recognizes phosphatidylserine (Ptd-Ser)2 fused to T cell and macrophage/dendritic cell-derived signaling domains. CERs can facilitate antigen capture, processing, and presentation, and impart target-dependent cytotoxic function when expressed in T cells. This combined function is hypothesized to improve tumor clearance and durability of response, making CER T cell products ideal clinical candidates.MethodsWe generated Tim-4 receptors fused to toll-like receptor (TLR)-2 or -8, CD28 or CD3 zeta and tested phagocytic, antigen presentation and cytotoxic function in healthy donor T cells. To assess phagocytosis, target cells treated with a small molecule to induce Ptd-Ser externalization were labeled with pH-Rodo followed by co-culture with CER T cells. Activated CER T cells were evaluated by transmission electron microscopy (TEM) or flow cytometry (FC) for lysosomal uptake of cell fragments. Antigen capture and presentation were characterized by FC for the capacity of human papilloma virus 16 (HPV 16) E7 peptide-pulsed CER T cells to activate and induce proliferation of autologous HPV 16 E7-TCR transduced T cells. Cytotoxic function was evaluated in co-culture assays of CER T cells in the presence of subtherapeutic doses of BTKi (ibrutinib)-treated JeKo-1 lymphoma cells.ResultsTEM imaging demonstrated that CER T cells engulfed target cell fragments, illustrated by multi-vesicular bodies containing tumor fragments (some measuring >0.5 uM) and pseudo-pod like formations around apoptotic target cell blebs. RNA analysis revealed upregulation of TLR, myeloid differentiation, and antigen presentation pathways. In the HPV 16 E7 co-culture model, T-cell surface activation markers CD25 and CD69 were upregulated 41% and 23%, respectively, on E7-TCR-T cells relative to controls. In addition, the percentage of dividing E7-TCR-T cells was increased (44% vs 8%) after 6 days in co-culture. Addition of CER T cells to JeKo- 1 target cells in the presence of BTKi at low effector: target ratios enhanced cytotoxicity by over 99%, demonstrating synergy with a targeted small molecule to fully eliminate lymphoma cells.ConclusionsNovel Tim-4/TLR containing CERs can capture tumor cell fragments and present soluble antigen, a function previously demonstrated to be a barrier to effective antigen presentation in T cells. Enhanced T-cell antigen capture and presentation capability alongside inducible and target-specific cytotoxic function in single T cells represents a significant advancement in the potential for chimeric receptor-based therapies.ReferencesLanzavecchia A, Roosnek E, Gregory T, Berman P, Abrignani S. T cells can present antigens such as HIV gp120 targeted to their own surface molecules. Nature 1988 Aug 11;334(6182):530–2.Caronni N, Piperno GM, Simoncello F, Romano O, Vodret S, Yanagihashi Y, et al. TIM4 expression by dendritic cells mediates uptake of tumor-associated antigens and anti-tumor responses. Nat Commun 2021 Apr 14;12(1):2237.


2021 ◽  
Author(s):  
Amera L Martinez ◽  
Justin T Gunesch ◽  
Emily M Mace

CD56 is the main identifying cell surface molecule of NK cells and has been recently identified as a regulator of cytotoxic function in NK cell lines. Despite its newly defined role in lytic granule polarization and exocytosis, biological questions remain involving the localization and function of CD56 at the immunological synapse. Here we use confocal and structured illumination microscopy to demonstrate recruitment of CD56 to the pSMAC of the immunological synapse of lytic effector cells. We provide additional data demonstrating that cell lines that are less dependent on CD56 for function are not utilizing alternative pathways of cytotoxicity, and that those that are dependent on CD56 have normal expression of activating and adhesion receptors. Finally, we use actin reporter (LifeAct) expressing NK92 cell lines and live cell confocal microscopy to visualize live cell killing events with WT and CD56-KO cells. This work further characterizes the novel role for CD56 in cytotoxic function of NK cells and provides deeper insight into the role of CD56 at the NK cell immunological synapse.


2021 ◽  
Vol 12 ◽  
Author(s):  
Yorleny Vicioso ◽  
Derek P. Wong ◽  
Nand K. Roy ◽  
Nayanika Das ◽  
Keman Zhang ◽  
...  

Natural Killer (NK) cells are cytotoxic lymphocytes critical to the innate immune system. We found that germline deficiency of NF-κB c-Rel results in a marked decrease in cytotoxic function of NK cells, both in vitro and in vivo, with no significant differences in the stages of NK cell development. We found that c-Rel binds to the promoters of perforin and granzyme B, two key proteins required for NK cytotoxicity, and controls their expression. We generated a NK cell specific c-Rel conditional knockout to study NK cell intrinsic role of c- Rel and found that both global and conditional c-Rel deficiency leads to decreased perforin and granzyme B expression and thereby cytotoxic function. We also confirmed the role of c-Rel in perforin and granzyme B expression in human NK cells. c-Rel reconstitution rescued perforin and granzyme B expressions in c-Rel deficient NK cells and restored their cytotoxic function. Our results show a previously unknown role of c-Rel in transcriptional regulation of perforin and granzyme B expressions and control of NK cell cytotoxic function.


2021 ◽  
Vol 12 ◽  
Author(s):  
Weiwen Zhang ◽  
Anna B. Morris ◽  
Erica V. Peek ◽  
Geeta Karadkhele ◽  
Jennifer M. Robertson ◽  
...  

Cytomegalovirus (CMV) is one of the most commonly recognized opportunistic pathogens and remains the most influential known parameter in shaping an individual’s immune system. As such, T cells induced by CMV infection could have a long-term impact on subsequent immune responses. Accumulating evidence indicates that memory T cells developed during past bacterial and viral infection can cross-react with unrelated pathogens, including transplant antigens, and can alter responses to de novo infections, vaccines, cancers, or rejection. Therefore, careful examination of T cell responses elicited by CMV is warranted to understand their potentially beneficial or harmful roles in future major immune events. Our detailed exploration of the distribution, phenotype, TCR repertoire and transcriptome of CD4+ T cells within CMV seropositive healthy individuals using high-dimensional flow cytometry and single cell multi-omics sequencing reveals that CMV seropositivity has highly significant age-independent effects, leading to a reduction in CD4+ naïve T cells and an expansion of CD4+ effector memory T cells and CD45RA+ effector memory T cells. These induced CD4+ effector memory T cells undergo a specific differentiation trajectory resulting in a subpopulation of CD57+CD27-CD28-CD244+ CD4+ T cells with cytotoxic function and TCR oligoclonality for optimal controlled coexistence with cytomegalovirus. Through gene set enrichment analysis, we found that this subpopulation is similar to virus-specific CD8+ T cells and T cells that mediate acute rejection in patients using tacrolimus and belatacept, a selective costimulation blocker. Together, these data suggest that memory CD4+ T cells induced by cytomegalovirus are formed via a distinct differentiation program to acquire cytotoxic function and can be potentially detrimental to transplant patients adopting costimulation blockade immunosuppressive regimen.


2021 ◽  
Vol 12 ◽  
Author(s):  
Morgane Humbel ◽  
Florence Bellanger ◽  
Natalia Fluder ◽  
Alice Horisberger ◽  
Madeleine Suffiotti ◽  
...  

Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease characterized by multiple cellular and molecular dysfunctions of the innate and adaptive immunity. Cytotoxic function of NK cells is compromised in patients with SLE. Herein, we characterized the phenotypic alterations of SLE NK cells in a comprehensive manner to further delineate the mechanisms underlying the cytotoxic dysfunction of SLE NK cells and identify novel potential therapeutic targets. Therefore, we examined PBMC from SLE patients and matched healthy controls by single-cell mass cytometry to assess the phenotype of NK cells. In addition, we evaluated the cell function of NK cells (degranulation and cytokine production) and the killing of B cell subpopulations in a B cell-NK cell in vitro co-culture model. We found that SLE NK cells expressed higher levels of CD38 and were not able to adequately upregulate SLAMF1 and SLAMF7 following activation. In addition, ligation of SLAMF7 with elotuzumab or of CD38 with daratumumab on SLE NK cells enhanced degranulation of both healthy and SLE NK cells and primed them to kill circulating plasma cells in an in vitro co-culture system. Overall, our data indicated that dysregulated expression of CD38, SLAMF1 and SLAMF7 on SLE NK cells is associated with an altered interplay between SLE NK cells and plasma cells, thus suggesting their contribution to the accumulation of (auto)antibody producing cells. Accordingly, targeting SLAMF7 and CD38 may represent novel therapeutic approaches in SLE by enhancing NK cell function and promoting elimination of circulating plasma cell.


2021 ◽  
Author(s):  
Alexandra Laine ◽  
Ossama Labiad ◽  
Hector Hernandez-Vargas ◽  
Sebastien This ◽  
Amélien Sanlaville ◽  
...  

Abstract Among the strategies allowing cancer cells to escape the immune system, the presence of TGF-b in the tumor micro-environment is one of the most potent. However, TGF-b is secreted in an inactive form and mechanisms responsible for its activation within the tumor remain unknown. Here, we demonstrate that regulatory T cells (Tregs) compose the main cells expressing the b8 chain of avb8 integrin (Itgb8) in the tumors and that the Itgb8pos Treg population activates TGF-b produced by the cancer cells and stored in the tumor micro-environment. Itgb8 ablation in Tregs impaired TGF-b signaling in T lymphocytes present in the tumor but not in the tumor draining lymph nodes. The cytotoxic function of CD8pos T lymphocytes infiltrating the tumors was subsequently exacerbated leading to an efficient control of the tumor growth. Similar observations were made in patient tumors after anti-Itgb8 antibody treatment. Thus, this study reveals that Tregs work in concert with cancer cells to produce bioactive-TGF-b and create a powerful-immunosuppressive micro-environment.


Sign in / Sign up

Export Citation Format

Share Document