scholarly journals Lymphadenopathy driven by TCR-Vγ8Vδ1 T-cell expansion in FAS-related autoimmune lymphoproliferative syndrome

2017 ◽  
Vol 1 (15) ◽  
pp. 1101-1106 ◽  
Author(s):  
Stefano Vavassori ◽  
Jacob D. Galson ◽  
Johannes Trück ◽  
Anke van den Berg ◽  
Rienk Y. J. Tamminga ◽  
...  

Key Points FAS-dependent apoptosis in Vδ1 T cells makes the latter possible culprits for the lymphadenopathy observed in patients with FAS mutations. Rapamycin and methylprednisolone resistance should prompt clinicians to look for Vδ1 T cell proliferation in ALPS-FAS patients.

Blood ◽  
2004 ◽  
Vol 104 (3) ◽  
pp. 775-780 ◽  
Author(s):  
Irini Sereti ◽  
Kara B. Anthony ◽  
Hector Martinez-Wilson ◽  
Richard Lempicki ◽  
Joseph Adelsberger ◽  
...  

Abstract Administration of interleukin 2 (IL-2) leads to selective and sustained CD4+ T-cell expansions in patients infected with HIV. It has been hypothesized that persistent CD4+ T-cell proliferation is the primary mechanism maintaining these expansions. T-cell proliferation was studied by ex vivo bromodeoxyuridine (BrdU) incorporation and intracellular Ki67 staining in HIV-infected patients treated with antiretroviral therapy (ART) with or without IL-2. In contrast to the tested hypothesis, HIV-infected patients treated with IL-2 had lower CD4+ T-cell proliferation compared to patients treated with ART alone. Independently of viral load changes, administration of IL-2 led to a decrease in basal CD4+ T-cell proliferation. Total numbers of CD4+ T cells with naive and recall, but not effector, memory phenotype were increased. The degree of CD4+ T-cell expansion correlated with the decreases in proliferation and a strong association was seen between these decreases and the expansion of the CD4+/CD25+ subset. Intermittent IL-2 in HIV-infected patients leads to expansions of CD4+/CD25+ T cells with naive and recall memory phenotypes that strongly correlate with decreases in proliferation. These data suggest that decreased T-cell proliferation is central in the CD4+ T-cell expansions induced by IL-2.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3255-3255
Author(s):  
Nicholas Leigh ◽  
Guanglin Bian ◽  
Wei Du ◽  
George L. Chen ◽  
Hong Liu ◽  
...  

Abstract Graft versus tumor (GVT) effect is the desired and integral outcome for successful allogeneic bone marrow transplantation (allo-BMT) for cancer patients. This effect is dependent on T cell mediated recognition and elimination of residual host tumor cells present after allo-BMT. T cell killing is mediated primarily via three pathways: perforin/granzymes, Fas/FasL, and cytotoxic cytokines. Recent work from our lab has revealed a detrimental role for granzyme B (GzmB) in GVT effect due to its role in activation induced cell death (AICD) of CD8+ T cells. As a result, GzmB-/- CD8+ T cells exhibited higher expansion after allo-BMT and subsequently provided better tumor control. Our current study sought to determine the role of perforin (Prf1) in GVT effect mediated by both CD4+ and CD8+ T cells. Using the MHC-mismatched C57BL/6 (H-2b) to BALB/c (H-2d) allo-BMT model, we first confirmed previous findings that when transplanting CD8+ T cells along with T cell depleted (TCD) BM cells, donor CD8+ T cells require Prf1 to mediate GVT effect against allogeneic A20 lymphoma (Fig 1A, Prf1-/- (n=4) vs WT (n=4), *P<0.05). In addition, our data suggest that Prf1 is also required for CD4+ T cells to effectively mediate GVT effect against A20, as transplant with Prf1-/- CD4+CD25- T cells does not control tumor growth as well as WT controls (Fig 1B). Our previous work showed that GzmB deficiency allows for less AICD and subsequently more CD8+ T cell expansion. New data now show a similar effect for Prf1 in CD8+ T cell accumulation, as Prf1-/- CD8+ T cells outcompete WT CD8+ T cells (CD45.1+) when these two genotypes are mixed in equal numbers and transplanted into tumor bearing BALB/c mice (n=5/time point, *P=0.02 day 9)(Fig 1C). This competitive advantage was due to less AICD in the Prf1-/- CD8+ T cells. However, Prf1 appears to be required for efficient GVT activity, because the higher number of Prf1-/- CD8+ T cells are still less capable than WT counterparts in controlling tumor growth. We next tested the effect of Prf1 in AICD in CD4+CD25- T cells, and again co-transplanted WT CD45.1+ and Prf1-/- CD4+CD25- T cells into tumor bearing mice for a competition assay. Unexpectedly, WT CD4+CD25- T cells accumulate to significantly higher numbers when in direct competition with Prf1-/- CD4+CD25- T cells (n=4/time point, **,P<0.01)(Fig 1D). When we measured apoptotic cells with Annexin V staining, we found that WT CD4+CD25- T cells still had significantly more AICD (Prf1-/- 38.3 ± 4.2% vs. WT 48.1 ± 5.1%, P<0.01 on day 7 post-BMT; Prf1-/- 12.7 ± 1.0% vs. WT 18.1 ± 3.4%, P<0.03 on day 9 post-BMT). This result suggests that while Prf1 has an important role in AICD, it may also play a role in another feature of CD4+ T cell biology. We then explored the hypothesis that may Prf1 promote CD4+ T cell proliferation by evaluating Hoescht staining on day 9 post-BMT. Preliminary results suggest that Prf1 may enhance T cell proliferation, as Prf1-/- CD4+ T cells have less actively dividing cells at this time point. Therefore, Prf1 appears to have a surprising role after allo-BMT in sustaining T cell expansion for CD4+ T cells, but not for CD8+ T cells. Another factor influencing GVT effect may be T cell phenotype. Our previous work with CD8+ T cells suggests that more effector memory (CD62LLOWCD44HIGH) T cells accumulate in the absence of GzmB, and that GzmB-/- CD8+ T cells exhibited higher GVT activity than WT controls. We now found that while Prf1-/- CD4+ T cells also skewed towards the effector memory phenotype (CD62LLOWCD44HIGH), loss of Prf1 still reduced the ability of CD4+ T cells to control tumor growth in this model of allo-BMT. In summary, our results suggest that Prf1 plays an important role in GVT responses mediated not only by CD8+ T cells but also by CD4+ T cells, which were shown in previous literature to mainly utilize Fas ligand and cytokine systems to mediate GVT activity. In addition, Prf1 can cause AICD to both CD4+ and CD8+ T cells after allo-BMT. While Prf1-induced AICD reduces CD8+ T cell expansion, Prf1 appears to play a previously unrecognized role enhancing CD4+ T cell proliferation via an unidentified mechanism. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (8) ◽  
pp. 804-814 ◽  
Author(s):  
John Rossi ◽  
Patrick Paczkowski ◽  
Yueh-Wei Shen ◽  
Kevin Morse ◽  
Brianna Flynn ◽  
...  

Key Points The PSI of manufactured CAR T cells was associated with clinical response and toxicities. Monitoring CAR T-cell polyfunctionality as a key product attribute may complement other characteristics including T-cell proliferation.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 242-242
Author(s):  
Daigo Hashimoto ◽  
Andrew Chow ◽  
Melanie Greter ◽  
Marylene Leboeuf ◽  
Florent Ginhoux ◽  
...  

Abstract Abstract 242 Host antigen presenting cells are critical to activate allo-reactive T cells and to initiate acute graft versus host disease (GVHD) after allogeneic bone marrow transplantation (BMT). However, while the role of host dendritic cells (DCs) in GVHD has been established, the contribution of host macrophages to GVHD has not been clearly addressed. In this study we revisited the role of host macrophages in GVHD. To this end we used an anti-CSF-1R monoclonal antibody (aCSF1R) to reduce macrophages but not DC numbers in lymphoid organs. We treated recipient C57BL/6 mice with αCSF1R on days -5 to -3 followed on day 0 by TBI and i.v. injection of BM cells and splenocytes isolated from MHC-mismatched BALB/c donor. We found that host macrophages that persist in the spleen and LN of recipient mice were severely reduced by αCSF1R, whereas host DCs remained unaffected by the treatment. We also found that αCSF1R-treatement significantly enhanced GVHD morbidity and mortality (Figure left panel) after allogeneic BMT, enhanced donor T cell expansion in recipient spleen, LN and liver, and increased IFN-gamma and TNF-alfa sera levels compared to mice treated with control IgG. Similar results were obtained when low dose Lip-Clod was administered 10 days prior to transplant in order to deplete macrophages in lymphoid tissues but not host DC whose half-life in lymphoid tissues does not exceed three days allowing them to recover to normal at the time of transplant. Our results revealed that in contrast to a previous report in which higher Lip-Clod doses administered 7 and 2 days prior to transplant led to the depletion of both DCs and macrophages and improved GVHD, low dose Lip-Clod administered 10 days prior to transplant, depleted host macrophages but not DCs and aggravated GVHD. We also explored the mechanisms by which host macrophages could potentially control donor T cell expansion induced by host DC after allo-HCT. We found that host macrophages isolated from allogeneic recipients inhibited the proliferation of allo-reactive T cells co-cultured with host C57BL/6 DC partly through a cell-cell contact pathway. Host macrophages were also able to engulf and reduce alloreactive T cells after few hrs of culture. Consistently, 18 hrs after allo-HCT, the number of donor T cells accumulating in the recipient spleen and mesenteric LNs were higher in Ab-treated mice compared to the control groups and CFSE-labeled donor T cells were engulfed by splenic macrophages during the first day of transplant and prior to the initiation of donor T cell proliferation. These results suggest that host macrophages limit the expansion of donor T cells through their ability to engulf donor T cell and to inhibit donor T cell proliferation induced by host DC stimulation. Further supporting the importance of host macrophages in regulating the development of GVHD, we found that pre-transplant administration of the cytokine CSF-1 increased the number of spleen macrophages, limited the expansion of donor allo-reactive T cells and improved GVHD outcome (Figure right panel). Altogether, we showed that in contrast to host DCs, host macrophages modulate GVHD through their ability to limit donor T cell activation and expansion. In addition, our data also identify pre-transplant CSF-1 injection as a novel clinical strategy for the prevention of GVHD in patient candidate for allo-HCT. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2017 ◽  
Vol 130 (3) ◽  
pp. 348-359 ◽  
Author(s):  
Trisha A. Dant ◽  
Kaifeng L. Lin ◽  
Danny W. Bruce ◽  
Stephanie A. Montgomery ◽  
Oleg V. Kolupaev ◽  
...  

Key Points Donor T cells lacking AhR demonstrate decreased aGVHD because of reduced donor T-cell proliferation early after transplant. Absence of AhR on donor cells increased pTreg cells in the colon; in vitro blockade increased the number of human iTreg from CD4+ T cells.


Blood ◽  
2018 ◽  
Vol 132 (12) ◽  
pp. 1279-1292 ◽  
Author(s):  
Qing Wang ◽  
Ran Yan ◽  
Nancy Pinnell ◽  
Anna C. McCarter ◽  
Yeonjoo Oh ◽  
...  

Key Points Notch1 cofactor Zmiz1 induces a subset of Notch target genes and drives pre–T-cell proliferation during normal and stress thymopoiesis. Disrupting the Zmiz1-Notch1 protein-protein interaction impairs Myc induction, pre–T-cell expansion, and leukemic proliferation.


Blood ◽  
2013 ◽  
Vol 121 (8) ◽  
pp. 1265-1275 ◽  
Author(s):  
Alicia N. McMurchy ◽  
Jana Gillies ◽  
Maria Concetta Gizzi ◽  
Michela Riba ◽  
Jose Manuel Garcia-Manteiga ◽  
...  

Key Points FOXP3 functions as a negative regulator of T-cell proliferation and cytokine production in human conventional T cells. Expression of FOXP3 in human Th17 cells functions to suppress IFN-γ production.


Blood ◽  
2015 ◽  
Vol 125 (16) ◽  
pp. 2519-2529 ◽  
Author(s):  
Le Xuan Truong Nguyen ◽  
Yunqin Lee ◽  
Lenore Urbani ◽  
Paul J. Utz ◽  
Anne W. Hamburger ◽  
...  

Key Points MPA suppresses ribosomal RNA (rRNA) synthesis and cell proliferation in T cells through TIF-IA, a GTP binding protein. The combination of MPA and sotrastaurin potently suppresses T-cell proliferation and inhibits IL-2 secretion through TIF-IA and ErbB3-binding protein 1 (Ebp1).


1993 ◽  
Vol 22 (2-3) ◽  
pp. 80-85
Author(s):  
A. Malley ◽  
N. Pangares ◽  
S.K. Mayo ◽  
M. Zeleny‐Pooley ◽  
J.V. Torres ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document