Preemptive HMG-CoA Reductase Inhibition Significantly Attenuates a Murine Chronic Graft-Versus-Host Disease

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 809-809
Author(s):  
Ji-Young Lim ◽  
Hyung-Gyu Yoon ◽  
Chang-Ki Min

Abstract Chronic graft-versus-host disease (cGVHD) is a serious and increasingly common complication of allogeneic stem cell transplantation. Treatment of cGVHD remains a large challenge. Even though cGVHD differs among patients, the clinical complications of cGVHD often include fibrosis and scleroderma-like changes. A murine sclerodermatous graft-versus-host disease (Scl GVHD) model sharing critical characteristics with human cGVHD is characterized by skin thickening and lung fibrosis. Statins, 3-hydroxy-3-methyl-CoA (HMG-CoA) reductase inhibitors which are a class of cholesterol lowering drugs decreasing mortality from cardiovascular disease and stroke, have strong immunomodulatory effects on antigen-presenting cells and T cells interfering with synthesis of L-mevalonate and its downstream isoprenoid metabolites. It has been demonstrated that some statins inhibit the production of proinflammatory cytokines and improved chronic inflammatory disorders. We used B10.D2 → BALB/c model of cGVHD, which differ at minor histocompatibility loci, to address the therapeutic effect of statins on the development of cGVHD. Lethally irradiated (700 cGy) BALB/c mice were transplanted with either B10.D2 (allogeneic) or BALB/c (syngeneic) bone marrow (1.5 × 106) and spleen cells (3 × 106) to generate Scl GVHD. We have noted skin thickening as early as day 14, however 28 days was usually required to observe significant skin thickening in allogeneic recipients with Scl GVHD. The skin thickenings were still lingering on but weaken as late as 56 days post-alloSCT, whereas the loss of normal lacy alveolar pattern of lungs remained constant up to that time. Syngeneic recipients did not show any significant changes in each organ. In vivo treatment of pravastatin (30 mg/kg/day, intraperitoneally) for 10 days early after transplant led to greater suppression of the incidence and severity of clinical skin cGVHD compared with allogeneic controls injected with diluent. The occurrence of clinical cutaneous GVHD was significantly slower in onset in the recipients of pravastatin treatment than it in the control animals (36 days vs. 25 days, respectively, P<0.05) Blinded pathologic scoring of skin disease on day 28 confirmed the clinical result. Skin from pravastatin recipients had an average pathology score of 2.5 ± 0.3 compared with 6.9 ± 0.7 for the allogeneic controls (P<0.01). To qualify the cells infiltrating skin or lungs in early Scl GVHD, we examined the number of CD11b+ or CD3+ cells isolated from each organ of the animals with Scl GVHD with or without pravastatin treatment on day 14, 28 and 56 post-alloSCT. Compared with the allogeneic controls with diluent treatment, the injection of pravastatin significantly reduced the number of cells infiltrating each organ on day 14; for bronchoalveolar lavage fluid 3.2 ± 0.3 ×104 vs. 2.2 ± 0.3 ×104 (P<0.05, CD11b+) and 1.3 ± 0.1 ×104 vs. 0.33 ± 0.1 ×104 (P<0.05, CD3+), for skin 56 ± 4.3 ×104 vs. 10 ± 2.3 ×104 (P<0.01, CD11b+) and 12 ± 3.1 ×104 vs. 3 ± 1.8 ×104 (P<0.01, CD3+). On days 28 and 56, the infiltrating cells were also significantly reduced in the animals treated with pravastatin (data not shown). To test whether statins might produce an effect on attracting these monocytes to skin or lungs by influencing the chemokine expression, we performed semiquantitative RT-PCR analyses and ELISA on day 14 to examine the expression of MCP-1 and RANTES in skin and lungs from Scl GVHD animals treated by either pravastatin or the diluent. MCP-1 and RANTES mRNA levels as well as protein concentrations from each organ were significantly reduced in recipients treated with pravastatins compared to the allogeneic controls. In conclusion, preemptive HMG CoA reductase inhibition prevented murine Scl GVHD by effectively blocking the influx of monocytes into target organs and by down-regulating the expression of MCP-1 and RANTES, thereby reducing new collagen synthesis. The Scl GVHD model is valuable for testing the effect of statins on early fibrosing diseases, and chemokines may be the potential targets in cGVHD protection effect of statins.

Blood ◽  
2007 ◽  
Vol 110 (13) ◽  
pp. 4588-4598 ◽  
Author(s):  
Robert Zeiser ◽  
Sawsan Youssef ◽  
Jeanette Baker ◽  
Neeraja Kambham ◽  
Lawrence Steinman ◽  
...  

We investigated whether atorvastatin (AT) was capable of protecting animals from acute graft-versus-host disease (aGVHD) across major histocompatibility complex (MHC) mismatch barriers. AT treatment of the donor induced a Th-2 cytokine profile in the adoptively transferred T cells and reduced their in vivo expansion, which translated into significantly reduced aGVHD lethality. Host treatment down-regulated costimulatory molecules and MHC class II expression on recipient antigen-presenting cells (APCs) and enhanced the protective statin effect, without impacting graft-versus-leukemia (GVL) activity. The AT effect was partially reversed in STAT6−/− donors and abrogated by L-mevalonate, indicating the relevance of STAT6 signaling and the L-mevalonate pathway for AT-mediated aGVHD protection. AT reduced prenylation levels of GTPases, abolished T-bet expression, and increased c-MAF and GATA-3 protein in vivo. Thus, AT has significant protective impact on aGVHD lethality by Th-2 polarization and inhibition of an uncontrolled Th-1 response while maintaining GVL activity, which is of great clinical relevance given the modest toxicity profile of AT.


Blood ◽  
2010 ◽  
Vol 115 (9) ◽  
pp. 1669-1677 ◽  
Author(s):  
Sheng F. Cai ◽  
Xuefang Cao ◽  
Anjum Hassan ◽  
Todd A. Fehniger ◽  
Timothy J. Ley

Abstract Regulatory T (Treg) cells can suppress a wide variety of immune responses, including antitumor and alloimmune responses. The mechanisms by which Treg cells mediate their suppressive effects depend on the context of their activation. We previously reported that granzyme B is important for Treg cell–mediated suppression of antitumor immune responses. We therefore hypothesized that granzyme B may likewise be important for suppression of graft-versus-host disease (GVHD). We found that allogeneic mismatch induces the expression of granzyme B in mixed lymphocyte reactions and in a model of graft-versus-host disease (GVHD). However, wild-type and granzyme B–deficient Treg cells were equally able to suppress effector T (Teff) cell proliferation driven by multiple stimuli, including allogeneicantigen-presenting cells. Surprisingly, adoptive transfer of granzyme B–deficient Treg cells prevented GVHD lethality, suppressed serum cytokine production in vivo, and prevented target organ damage. These data contrast strikingly with our previous study, which demonstrated that granzyme B plays a nonredundant role in Treg cell–mediated suppression of antitumor responses. Taken together, these findings suggest that targeting specific Treg cell–suppressive mechanisms, such as granzyme B, may be therapeutically beneficial for segregating GVHD and graft-versus-tumor immune responses.


Blood ◽  
1994 ◽  
Vol 84 (8) ◽  
pp. 2815-2820 ◽  
Author(s):  
PY Dietrich ◽  
A Caignard ◽  
A Lim ◽  
V Chung ◽  
JL Pico ◽  
...  

In a series of patients transplanted with HLA-matched allogeneic bone marrow grafts (alloBMT), we previously showed that a few T-cell receptor (TCR) V alpha and V beta gene segment transcripts were overexpressed in skin compared with blood at the time of acute graft- versus-host disease (aGVHD). Here, in one selected patient with overexpressed V beta 16 and V alpha 11 transcripts in skin, we analyzed the junctional variability of these transcripts in donor blood, patient blood, and skin collected at aGVHD onset. A unique junctional region sequence accounted for 81% of in frame V beta 16 transcripts (13 of 16) in skin and 59% (13 of 22) in patient blood. Similarly, two recurrent junctional region sequences were found in skin V alpha 11 transcripts, one accounting for 66% (21 of 32) and the other for 16% (5 of 32). These recurrences were also found in patient blood (36% and 15% of V alpha 11 transcripts, respectively). To extend our analysis, a polymerase chain reaction (PCR)-based method was used to precisely determine TCR beta transcript length in run-off reactions using uncloned bulk cDNA samples. All V beta-C beta PCR products analyzed in donor blood, as well as the majority of those analyzed in patient blood, included transcripts with highly diverse junctional region sizes. As expected from the sequence data, most V beta 16-C beta PCR products in skin and patient blood were of the same size (ie, same junctional region). In addition, V beta 3, V beta 5, and V beta 17 transcripts in skin were shown to display highly restricted size variability. The clonality of the V beta 16-C beta and V beta 17-C beta transcripts was further supported by the results of run-off reactions using 13 J beta specific primers. We have identified several recurrent TCR transcripts in skin, some of them also present in patient blood. These data support the view that several T-cell subpopulations are clonally expanded in vivo at the time of aGVHD onset in this case of related HLA-matched alloBMT.


Blood ◽  
1999 ◽  
Vol 93 (9) ◽  
pp. 3140-3147 ◽  
Author(s):  
Joshua A. Grass ◽  
Tamim Wafa ◽  
Aaron Reames ◽  
David Wages ◽  
Laurence Corash ◽  
...  

Abstract Photochemical treatment (PCT) with the psoralen S-59 and long wavelength ultraviolet light (UVA) inactivates high titers of contaminating viruses, bacteria, and leukocytes in human platelet concentrates. The present study evaluated the efficacy of PCT to prevent transfusion-associated graft-versus-host disease (TA-GVHD) in vivo using a well-characterized parent to F1 murine transfusion model. Recipient mice in four treatment groups were transfused with 108 splenic leukocytes. (1) Control group mice received syngeneic splenic leukocyte transfusions; (2) GVHD group mice received untreated allogeneic splenic leukocytes; (3) gamma radiation group mice received gamma irradiated (2,500 cGy) allogeneic splenic leukocytes; and (4) PCT group mice received allogeneic splenic leukocytes treated with 150 μmol/L S-59 and 2.1 J/cm2UVA. Multiple biological and clinical parameters were used to monitor the development of TA-GVHD in recipient mice over a 10-week posttransfusion observation period: peripheral blood cell levels, spleen size, engraftment by donor T cells, thymic cellularity, clinical signs of TA-GVHD (weight loss, activity, posture, fur texture, skin integrity), and histologic lesions of liver, spleen, bone marrow, and skin. Mice in the control group remained healthy and free of detectable disease. Mice in the GVHD group developed clinical and histological lesions of TA-GVHD, including pancytopenia, marked splenomegaly, wasting, engraftment with donor derived T cells, and thymic hypoplasia. In contrast, mice transfused with splenic leukocytes treated with (2,500 cGy) gamma radiation or 150 μmol/L S-59 and 2.1 J/cm2 UVA remained healthy and did not develop detectable TA-GVHD. Using an in vitro T-cell proliferation assay, greater than 105.1 murine T cells were inactivated by PCT. Therefore, in addition to inactivating high levels of pathogenic viruses and bacteria in PC, these data indicate that PCT is an effective alternative to gamma irradiation for prevention of TA-GVHD.


Sign in / Sign up

Export Citation Format

Share Document