scholarly journals NK Cells Inhibit T Cell Proliferation via p21-Mediated Cell Cycle Arrest

2005 ◽  
Vol 174 (8) ◽  
pp. 4590-4597 ◽  
Author(s):  
Prachi P. Trivedi ◽  
Paul C. Roberts ◽  
Norbert A. Wolf ◽  
Robert H. Swanborg
Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4916-4916
Author(s):  
Tian-Hui Yang ◽  
Kathryn E Quintanilla ◽  
Amy M Cortez ◽  
Jeffrey J. Molldrem

Abstract Proteinase 3 (P3), a serine protease found in primary granules in granulocytes, is the target of T cell- and B cell-mediated autoimmunity in Wegener’s granulomatosis (WG) and of anti-leukemia immunity mediated by PR1 (VLQELNVTV)-specific cytotoxic T lymphocytes (PR1-CTL). Although aberrant P3 and neutrophil elasase (NE) expression in leukemia increases susceptibility to PR1-CTL-mediated killing, overexpression of P3 also induces apoptosis of the high affinity PR1-CTL leading to deletional tolerance and leukemia outgrowth. Because expression of P3 and NE in sera from leukemia patients is increased by 5-fold compared to healthy controls, we sought to determine whether such overexpression of P3 or NE impairs PR1-CTL immunity to leukemia by a direct affect on T lymphocytes. To study this, T cells from healthy donors were activated by anti-CD3 and anti-CD28 and exposed to increasing concentration of P3 or NE over one to five days, and the percentage of apoptotic cells and cell proliferation were determined by flow cytometry using PI, anti-Ki-67, and CFSE. P3, but not NE, induced dose-dependent apoptosis of up to 30% of T cells, and in the non-apoptotic cells, a 50% inhibition of CD4 and CD8 T cell proliferation at 1 μg/ml and 100% at 10 μg/ml compared to untreated cells. This effect was not enzyme-mediated since prior exposure of P3 to 56°C or co-incubation with the serine protease inhibitors Elafin and alpha-1 antitrypsin showed no affect on apoptosis or cell proliferation. P3 induced a cell cycle arrest at the G0/G1 interface, determined with PI and Ki-67 staining of healthy donor T cells that were exposed to P3 for up to 3 days. In contrast, at protein concentrations up to 25 μg/ml, NE showed no such inhibitory effect on apoptosis or cell proliferation. In addition to its role as a leukemia-associated antigen, P3 is also targeted by the cANCA antibody in patients with WG and the serum titer correlates with disease activity. Therefore, we hypothesized that the effect of P3 on T cell proliferation might also be affected by humoral immunity during circumstances of systemic autoimmunity. Co-incubation of P3 with a molar excess of cANCA reversed P3- mediated inhibition of both CD4 and CD8 T cells, consistent with a role of this antibody and the P3 target antigen in controlling T cell autoreactivity. Taken together, this data shows a new role for P3 in regulating T cell proliferation, which occurs only at high P3 concentration, similar to P3 in sera from leukemia patients, which is not enzymedependent. This supports a direct role for P3 in regulating both anti-leukemia immunity and autoimmunity. This data will need to be considered for effective immunotherapy targeting P3 in leukemia patients and these inhibitory effects also suggest a role for P3 in regulating autoimmunity at sites of inflammation, such as in patients with WG.


2014 ◽  
Vol 96 (3) ◽  
pp. 453-462 ◽  
Author(s):  
Johannes Wedel ◽  
Maximillia C. Hottenrott ◽  
Eleni Stamellou ◽  
Annette Breedijk ◽  
Charalambos Tsagogiorgas ◽  
...  

2013 ◽  
Vol 55 (4) ◽  
pp. 932-933
Author(s):  
Eyal Ben-Ami ◽  
Norberto Krivoy ◽  
Edna Efrati

2016 ◽  
Vol 2016 ◽  
pp. 1-11 ◽  
Author(s):  
Xiuying Li ◽  
Zhuo Xu ◽  
Jinping Bai ◽  
Shuyuan Yang ◽  
Shuli Zhao ◽  
...  

It has been reported that human mesenchymal stem cells are able to inhibit T lymphocyte activation; however, the discrepancy among different sources of MSCs is not well documented. In this study, we have compared the MSCs from bone marrow (BM), adipose tissue (AT), placenta (PL), and umbilical cord (UC) to determine which one displayed the most efficient immunosuppressive effects on phytohemagglutinin-induced T cell proliferation. Among them we found that hUC-MSC has the strongest effects on inhibiting T cell proliferation and is chosen to do the further study. We observed that T lymphocyte spontaneously released abundant IFN-γ. And IFN-γsecreted by T lymphocyte could induce the expression of indoleamine 2, 3-dioxygenase (IDO) in hUC-MSCs. IDO was previously reported to induce T lymphocyte apoptosis and cell cycle arrest in S phase. When cocultured with hUC-MSCs, T lymphocyte expression of caspase 3 was significantly increased, while Bcl2 and CDK4 mRNA expression decreased dramatically. Addition of 1-methyl tryptophan (1-MT), an IDO inhibitor, restored T lymphocyte proliferation, reduced apoptosis, and induced resumption of the cell cycle. In addition, the changes in caspase 3, CDK4, and Bcl2 expression were reversed by 1-MT. These findings demonstrate that hUC-MSCs induce T lymphocyte apoptosis and cell cycle arrest by expressing abundant IDO and provide an explanation for some of the immunomodulatory effects of MSCs.


Molecules ◽  
2020 ◽  
Vol 25 (21) ◽  
pp. 5031
Author(s):  
Hyun-Su Lee ◽  
Gil-Saeng Jeong

The appropriate regulation of T cell activity under inflammatory conditions is crucial for maintaining immune homeostasis. Salinosporamide A discovered as a self-resistance product from the marine bacterium Salinospora tropica, has been used as a potent proteasome inhibitor (PI). Although PIs have been developed as novel therapeutics for autoimmune diseases, due to their immunosuppressive effect, whether salinosporamide A inhibits T cell activation remains unknown. The current study finds that salinosporamide A is not cytotoxic, but controls T cell proliferation. Results from our cell cycle arrest analysis revealed that salinosporamide A leads to cell cycle arrest and regulates the expression of cyclin-dependent kinases. Under activated conditions, salinosporamide A abrogated T cell activation by T cell receptor-mediated stimulation, in which the production of cytokines was inhibited by pretreatment with salinosporamide A. Furthermore, we demonstrated that the regulation of T cell activation by salinosporamide A is mediated by suppressing the MAPK pathway. Therefore, our results suggest that salinosporamide A effectively suppresses T cell activation through regulating T cell proliferation and the cell cycle and provides great insight into the development of novel therapeutics for autoimmune diseases or graft-versus-host disease.


2021 ◽  
Vol 19 (1) ◽  
pp. 119-127
Author(s):  
Ibrahim O. Barnawi ◽  
Fahd A. Nasr ◽  
Omar M. Noman ◽  
Ali S. Alqahtani ◽  
Mohammed Al-zharani ◽  
...  

Abstract Different phytochemicals from various plant species exhibit promising medicinal properties against cancer. Juniperus phoenicea is a plant species that has been found to present medicinal properties. Herein, crude extract and fractions of J. phoenicea were examined to determine its anticancer properties against several cancer cells. The active fraction was chosen to assess its activity on cell cycle progression and apoptosis induction by annexin and propidium iodide (PI) biomarkers. Further, phytochemical screening for possible contents of active fraction using gas chromatography–mass spectrometry (GC-MS) analysis was conducted. It was demonstrated that cell proliferation was suppressed, and the MCF-7 cell line was the most sensitive to J. phoenicea chloroform fraction (JPCF), with the IC50 values of 24.5 μg/mL. The anti-proliferation activity of JPCF in MCF-7 cells was linked to the aggregation of cells in the G1 phase, increases in early and late apoptosis as well as necrotic cell death. Contents analysis of JPCF using GC-MS analysis identified 3-methyl-5-(2′,6′,6′-trimethylcyclohex-1′-enyl)-1-penten-3-ol (16.5%), methyl 8-oxooctanoate (15.61%), cubenol (13.48%), and 7-oxabicyclo [2.2.1] heptane (12.14%) as major constituents. Our present study provides clear evidence that J. phoenicea can inhibit cell proliferation, trigger cell cycle arrest, and induce apoptosis in tested cancer cells.


2021 ◽  
Vol 7 (1) ◽  
Author(s):  
Pan Wang ◽  
Sheng Gong ◽  
Jinyu Pan ◽  
Junwei Wang ◽  
Dewei Zou ◽  
...  

AbstractThere exists a consensus that combining hyperbaric oxygen (HBO) and chemotherapy promotes chemotherapy sensitivity in GBM cells. However, few studies have explored the mechanism involved. HIF1α and HIF2α are the two main molecules that contribute to GBM malignant progression by inhibiting apoptosis or maintaining stemness under hypoxic conditions. Moreover, Sox2, a marker of stemness, also contributes to GBM malignant progression through stemness maintenance or cell cycle arrest. Briefly, HIF1α, HIF2α and Sox2 are highly expressed under hypoxia and contribute to GBM growth and chemoresistance. However, after exposure to HBO for GBM, whether the expression of the above factors is decreased, resulting in chemosensitization, remains unknown. Therefore, we performed a series of studies and determined that the expression of HIF1α, HIF2α and Sox2 was decreased after HBO and that HBO promoted GBM cell proliferation through cell cycle progression, albeit with a decrease in stemness, thus contributing to chemosensitization via the inhibition of HIF1α/HIF2α-Sox2.


Sign in / Sign up

Export Citation Format

Share Document