GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 3050-3050
    Abstract: Graft-versus-host disease (GVHD) mediated by alloreactive donor T cells is the most dreaded complication after allogeneic bone marrow transplantation (BMT). Conditioning therapy in the context of BMT creates a proinflammatory milieu, which is thought to be central to the development of GVHD. Interfering with the conditioning-induced inflammatory response could be an approach to prevent GVHD without compromising the graft-versus-malignancy reaction. Histone deacetylase (HDAC) inhibitors belong to a new family of anti-cancer drugs with potent anti-inflammatory properties and have recently been shown to reduce the development of GVHD. The aim of this study was understand the mechanisms underlying the downregulation of GVHD after treatment with the HDAC inhibitor suberonylanilide hydroxamic acid (SAHA). Using the fully MHC-mismatched strain combination B6 to BALB/c, treatment with SAHA resulted in a significantly reduced GVHD mortality. Thus, at days +10 or +37 post-BMT survival for vehicle-treated or SAHA-treated mice was 33 % versus 86 % and 8 % versus 57 % respectively (Chi2 test, p = 0,027 and p = 0,02, respectively). This was associated with a significant reduction in IFN-g and IL-5 serum levels of SAHA-treated animals. As we could not detect any effect of SAHA treatment on T cell activation or T cell expansion in vitro and in vivo, we hypothesized that the inhibitory effect of SAHA treatment on the development of GVHD might be primarily due to an interference in the early events of the inflammatory cascade occurring after conditioning and initial alloactivation. Therefore, we performed gene expression profiling studies in classical GVHD target organs of animals treated with SAHA or vehicle to further understand the mechanisms underlying this effect. SAHA treated animals revealed a significant upregulation of the mRNA expression of the Protein inhibitor of activated stat 1 (PIAS1) gene in the liver compared to vehicle-treated animals. To further strengthen the hypothesis that SAHA might exert its action by interfering with inflammatory reaction and subsequent signaling through the JAK/STAT pathway, we analyzed the effects of SAHA on STAT-1, 3, and 5 activation and expression of SOCS-1 and SOCS-3 in vitro and in vivo. Thus, BALB/c responder splenocytes were incubated with or without irradiated B6 stimulators in the presence or absence of LPS in order to allow for the separate analysis of LPS and alloactivation-induced JAK/STAT activation. Treatment for 24 hours with SAHA completely inhibited phosphorylation of STAT-1 and STAT-3 in response to LPS and alloactivation using western blot analysis. Furthermore, analysis of liver tissue from GVHD animals showed a sustained expression of SOCS-1 protein in SAHA treated animals whereas SOCS-1 was downregulated in the absence of SAHA. In conclusion our data suggest that the inhibitory effect of SAHA on the development of GVHD is associated with an inhibition of the JAK/STAT signaling pathway. Further studies are warranted to understand the precise mechanisms how SAHA interferes with JAK/STAT signaling and how this leads to inhibition of GVHD. However, it is conceivable that interfering with inflammatory signaling pathways using pharmacological inhibitors of the JAK/STAT pathway might provide a highly attractive treatment strategy for the prevention of GVHD.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Nature Immunology, Springer Science and Business Media LLC, Vol. 7, No. 2 ( 2006-2), p. 207-215
    Type of Medium: Online Resource
    ISSN: 1529-2908 , 1529-2916
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2006
    detail.hit.zdb_id: 2026412-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 4988-4988
    Abstract: Delayed administration of DLI to mixed hematopoietic chimeras following allogeneic bone marrow transplantation (alloBMT) leads to conversion from mixed to full donor chimerism without GVHD. However, in the clinical setting development of GVHD is still the most common complication. Donor T cells are the main mediators of GVHD in alloBMT. However, the precise role of host dendritic cells (DC) and their different subsets needs still to be elucidated. CD8a+ DCs seem to have a regulatory function and to reduce experimental GVHD. Recently, it has been shown that the ICSBP (Interferon Consensus Sequence Binding Protein) gene is essential for the development and activation of CD8a+ DCs. ICSBP-gene deficient mice (ICSBP KO) are characterized by several immune defects and systemic expansion of myeloid cells mimicking human CML and may be therefore an attractive in vivo model to evaluate the role of DCs in alloBMT. The aim of the study was to evaluate the 1. feasibility of inducing mixed hematopoietic chimerism following nonmyeloablative conditioning in ICSBP KO mice using fully MHC-mismatched BMT (Balb/c [H2Dd] to B6 x 129 [H2b]), and 2. susceptibility of ICSBP KO mice to the development of GVHD following administration of DLI. Nonmyeloablative conditioning consisted of in-vivo T cell depletion (anti-CD4-[GK1.5] , anti-CD8-[2,43] mAb, d-5), fludarabine (30 mg/kg, d-4 to d-2), cyclophosphamid (200 mg/kg, d-1) and 3 Gy TBI (d0) followed by i.v. injection of 1,5 x 10E7 Balb/c bone marrow cells. Stable mixed chimerism was observed in ICSBP wildtype (wt) mice through week 35 post-BMT without signs of clinical GVHD. In contrast, we observed increasing donor CD4 and CD8 T cell chimerism over time in ICSBP KO mice suggesting a lack of donor T cell tolerance. Furthermore, chimerism was higher in granulocyte, B cells and monocyte compared to ICSBP wt mice. In a second experiment we were able to observe spontaneous conversion to full donor hematopoietic chimerism in 2 of 9 mice after nonmyeloablative BMT. In two separate experiments ICSBP wt or KO mice with mixed chimerism received (DLI) on day 35 post BMT. After injection of DLI all wt mice (n = 10) showed a rapid conversion to full donor chimerism and remained healthy until the end of the observation period without signs of GVHD. Despite myeloid hyperproliferation all ICSBP KO mice showed conversion to full donor chimerism. Furthermore, in ICSBP KO mice conversion to full donor chimerism was associated with development of severe GVHD (n = 9). In vitro studies using mixed lymphocyte reactions showed that splenic stimulator cells from ICSBP KO mice had a higher stimulatory capacity compared to wt stimulators (SI 1,5 versus 2,3). Using the same mixed lymphocyte culture (MLC) conditions, supernatants collected from BALB/c responders with ICSBP KO stimulators had higher levels of TNF-alpha and IFN-g compared to MLC using ICSBP wt stimulators. In conclusion, following alloBMT a defective development of host-specific tolerance and high susceptibility for GVHD in ICSBP KO mice is suggested. Further studies are warranted to delineate the precise underlying mechanism by addressing the potential contribution of defective regulatory host T cells and the lack of host CD8a+ DCs in ICSBP KO mice.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...