T Cell Proliferation and Homeostasis: An Emerging Role for the Cell Cycle Inhibitor Geminin

2011 ◽  
Vol 31 (3) ◽  
pp. 209-231 ◽  
Author(s):  
Dimitris Karamitros ◽  
Panorea Kotantaki ◽  
Zoi Lygerou ◽  
Dimitris Kioussis ◽  
Stavros Taraviras
Blood ◽  
2014 ◽  
Vol 123 (22) ◽  
pp. 3452-3461 ◽  
Author(s):  
Yang Wang ◽  
Xiaoguang Gu ◽  
Gaolei Zhang ◽  
Lin Wang ◽  
Tingting Wang ◽  
...  

Key Points SATB1 is specifically overexpressed in the CD30+ lymphoma cells in cutaneous CD30+ lymphoproliferative disease. SATB1 promotes proliferation of CD30+ lymphoma cells by direct transcriptional repression of cell cycle inhibitor p21.


10.1038/72272 ◽  
2000 ◽  
Vol 6 (2) ◽  
pp. 171-176 ◽  
Author(s):  
Dimitrios Balomenos ◽  
Juan Martín-Caballero ◽  
Maria I. García ◽  
Ignacio Prieto ◽  
Juana M. Flores ◽  
...  

2000 ◽  
Vol 164 (12) ◽  
pp. 6188-6192 ◽  
Author(s):  
Xuefeng Ling ◽  
Salar Kamangar ◽  
Michelle L. Boytim ◽  
Zvi Kelman ◽  
Philip Huie ◽  
...  

2021 ◽  
Vol 12 ◽  
Author(s):  
Stephanie A. Amici ◽  
Wissam Osman ◽  
Mireia Guerau-de-Arellano

Multiple Sclerosis (MS) is a debilitating central nervous system disorder associated with inflammatory T cells. Activation and expansion of inflammatory T cells is thought to be behind MS relapses and influence disease severity. Protein arginine N-methyltransferase 5 (PRMT5) is a T cell activation-induced enzyme that symmetrically dimethylates proteins and promotes T cell proliferation. However, the mechanism behind PRMT5-mediated control of T cell proliferation and whether PRMT5 contributes to diseases severity is unclear. Here, we evaluated the role of PRMT5 on cyclin/cdk pairs and cell cycle progression, as well as PRMT5’s link to disease severity in an animal model of relapsing-remitting MS. Treatment of T helper 1 (mTh1) cells with the selective PRMT5 inhibitor, HLCL65, arrested activation-induced T cell proliferation at the G1 stage of the cell cycle, suggesting PRMT5 promotes cell cycle progression in CD4+ T cells. The Cyclin E1/Cdk2 pair promoting G1/S progression was also decreased after PRMT5 inhibition, as was the phosphorylation of retinoblastoma. In the SJL mouse relapsing-remitting model of MS, the highest PRMT5 expression in central nervous system-infiltrating cells corresponded to peak and relapse timepoints. PRMT5 expression also positively correlated with increasing CD4 Th cell composition, disease severity and Cyclin E1 expression. These data indicate that PRMT5 promotes G1/S cell cycle progression and suggest that this effect influences disease severity and/or progression in the animal model of MS. Modulating PRMT5 levels may be useful for controlling T cell expansion in T cell-mediated diseases including MS.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1393-1393
Author(s):  
Kazuya Sato ◽  
Katsutoshi Ozaki ◽  
Iekuni Oh ◽  
Keiko Hatanaka ◽  
Tadashi Nagai ◽  
...  

Abstract Mesenchymal stem cells (MSCs) are attractive source for regenerative therapy as they have been shown to be capable of differentiating into adipocytes, chondrocytes, osteoblasts, myocytes, cardiomyocytes, and neural precursors. MSCs have also been shown to suppress T cell proliferation in vitro and were reported to be effective as a treatment for acute graft-versus-host disease (GVHD) but the underlying molecular mechanisms for T cell suppression are uncertain. So far, TGF-β, HGF, and PGE2 were shown to be candidates as molecules causing the suppression. To address the molecular mechanisms, we used primary mouse MSCs derived from bone marrow cells and CFSE (carboxyfluorescein diacetate succinimidyl ester) or thymidine uptake for T cell proliferation assay. Co-culture of MSCs inhibited T cell proliferation induced by PMA plus Ionomycin, suggesting that TCR and signaling molecules interacting with TCR such as Lck and ZAP70 are not involved and that downstream signals of PMA plus Ionomycin are essential for the suppression by MSCs. The proliferation of either purified CD4 or CD8 cells induced by PMA plus Ionomycin was also inhibited by co-culture with MSCs, indicating MSCs suppression is active on both CD4 and CD8 cells. Stat5 phosphorylation in activated T cells was suppressed by co-culture with MSCs. Induction of cell-cycle promoting proteins such as CDK6, Cyclin D2, and Cyclin E by mitogenic stimulation were inhibited and suppression of a cell-cycle inhibitor, Kip1, was abolished. A previous report showed that T cells from stat5 deficient mice failed to induce cell-cycle promoting proteins and were not be able to proliferate on the stimulation through TCR. It was also reported that Nitric Oxide (NO) suppressed stat5 phosphorylation. Taken together with these reports, we hypothesized that NO is another candidate for the cause of suppression. In fact, NO synthase inhibitor (N-nitro-L-arginine methyl ester) recovered T cell proliferation from the suppression by MSCs in a dose-dependent manner. The amount of NO production and the strength of T cell suppression were parallel and dependent on the number of MSCs. MSCs blocked production of IFNγ but induction of T cell activation markers such as CD25 and CD69 and production of IL-2 were unaffected as reported. Our data suggest that MSCs block stat5 phosphorylation by production of NO, resulting in that T cells can neither proliferate nor produce high level of IFNγ. Here we demonstrate a new critical NO-stat5 dependent mechanism for T cell suppression by MSCs.


2010 ◽  
Vol 140 (8) ◽  
pp. 1509-1515 ◽  
Author(s):  
Munkyong Pae ◽  
Zhihong Ren ◽  
Mohsen Meydani ◽  
Fu Shang ◽  
Simin Nikbin Meydani ◽  
...  

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.


2004 ◽  
Vol 173 (8) ◽  
pp. 5077-5085 ◽  
Author(s):  
Markus Falk ◽  
Sandra Ussat ◽  
Norbert Reiling ◽  
Daniela Wesch ◽  
Dieter Kabelitz ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document