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
Filter
  • Boyd, Richard L  (2)
  • 1
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 1043-1043
    Abstract: Abstract 1043 Thymopoiesis is a highly complex process involving cross-talk interactions between developing thymocytes and the supporting non-hematopoietic stromal microenvironment, which includes highly specialized thymic epithelial cells (TECs). Paradoxical to its importance for continually generating a diverse repertoire for effective adaptive immunity, the thymus undergoes profound atrophy with age. Age-related thymic involution is characterized by severe structural dysregulation of the supporting epithelial microenvironment (and in humans linked to a buildup of fatty tissue), reduced thymopoiesis, and subsequently reduced export of na•ve lymphocytes into the periphery. Together this degeneration in thymic function significantly narrows the T cell receptor repertoire and can causally linked to increased infection, autoimmunity and malignancy. Moreover, progressive thymic involution can also be a considerable hindrance to the regeneration of adaptive immunity following cytoreductive treatments such as chemotherapy or the conditioning required for successful hematopoietic stem cell transplant. Despite considerable work, little is understood about the underlying causes of age-related thymic involution. We have recently demonstrated a novel role for interleukin-22 (IL-22), a recently identified cytokine predominantly associated with maintenance of barrier function at mucosal surfaces, in endogenous thymic regeneration from acute immune injury. Our studies suggested that 1) the depletion of DP thymocytes triggers, 2) upregulation of IL-23 by dendritic cells (DCs), which induces 3) the production of IL-22 by intrathymic innate lymphoid cells (ILCs). IL-22 promotes the proliferation and survival of TECs, therefore this cascade of events leads to regeneration of the supporting microenvironment and, ultimately, to rejuvenation of thymopoiesis. Given these recent findings demonstrating a role for IL-22 in endogenous thymic regeneration following acute immune injury, one hypothesis would be that a breakdown in the IL-22 pathway contributes towards chronic age-related thymus involution. However, in contrast to this initial hypothesis, our studies revealed that rather than being depleted with age, there was actually a significant increase in the level of intrathymic IL-22 in aged (18+ months old) compared to young (2 months old) mice (Figure 1a). These findings highlighted that, in addition to being triggered by the depletion of CD4+CD8+ double positive thymocytes during acute immune injury, the IL-22 regenerative pathway can also be activated by the chronic atrophy that is a hallmark of age-related thymic involution. Similar to our findings in models of thymic injury in young mice, we found that these increased levels of IL-22 with age were predicated on the increased production of IL-22 by thymic innate lymphoid cells (Figure 1b). Moreover, in keeping with our findings in young mice with acute thymic injury, intrathymic levels of IL-22 in aged mice correlated with those of IL-23 - production of which by dendritic cells was significantly increased with age (Figure 1c). As predicted by this increase in the production of IL-22 with age, TECs from aged mice displayed all the hallmarks of increased IL-22 signaling including increased expression of the IL-22 receptor (Figure 1d) as well as increased phosphorylation of STAT-3 (Y705) (Figure 1e). However, although in vitro incubation of aged TECs with IL-22 led to increased proliferation, consistent with our findings in young mice, in vivo analysis revealed significantly reduced proliferation among TECs in aged mice (Figure 1f), as has been previously reported. Given the role for inflammasome components in mediating thymic involution, it is possible that although endogenous regenerative pathways are triggered with age (in the case of IL-22 likely due to the depletion of DP thymocytes), these regular processes fail in the face of an overwhelming inflammatory milieu in the thymus with age. Although further studies need to elucidate the specific inhibitory interactions constraining thymic regeneration, it is clear that strategies harnessing these endogenous pathways for enhancing immunity in the aging thymus first need to overcome these negative stimuli for effective regeneration. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    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: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 143-143
    Abstract: Abstract 143 Despite being exquisitely sensitive to insult, the thymus is remarkably resilient in young healthy animals. Endogenous regeneration of the thymus is a crucial function that allows for renewal of immune competence following infection or immunodepletion caused by cytoreductive chemotherapy or radiation. However, the mechanisms governing this regeneration remain poorly understood. Thymopoiesis is a highly complex process involving cross-talk between developing thymocytes and their supporting non-hematopoietic stromal microenvironment, which includes highly specialized thymic epithelial cells (TECs) that are crucial for T cell development. IL-22 is a recently identified cytokine predominantly associated with maintenance of barrier function at mucosal surfaces. Here we demonstrate for the first time a critical role for IL-22 in endogenous thymic repair. Comparing IL-22 KO and WT mice we observed that while IL-22 deficiency was redundant for steady-state thymopoiesis, it led to a pronounced and prolonged loss of thymus cellularity following sublethal total body irradiation (SL-TBI), which included depletion of both thymocytes (p=0.0001) and TECs (p=0.003). Strikingly, absolute levels of IL-22 were markedly increased following thymic insult (p 〈 0.0001) despite the significant depletion of thymus cellularity. This resulted in a profound increase in the production of IL-22 on a per cell basis (p 〈 0.0001). These enhanced levels of IL-22 peaked at days 5 to 7 after SL-TBI, immediately following the nadir of thymic cellularity. This was demonstrated by a strong negative correlation between thymic cellularity and absolute levels of IL-22 (Fig 1a). In mucosal tissues the regulation of IL-22 production has been closely associated with IL-23 produced by dendritic cells (DCs) and ex vivo incubation of cells with IL-23 stimulates the production of IL-22. Following thymic insult there was a significant increase in the amount of IL-23 produced by DCs (Fig 1b) resulting in similar kinetics of intrathymic levels of IL-22 and IL-23. We identified a population of radio-resistant CD3−CD4+IL7Ra+RORg(t)+ thymic innate lymphoid cells (tILCs) that upregulate both their production of IL-22 (Fig 1c) and expression of the IL-23R (p=0.0006) upon exposure to TBI. This suggests that they are responsive to IL-23 produced by DCs in vivo following TBI and, in fact, in vitro stimulation of tILCs by IL-23 led to upregulation of Il-22 production by these cells (Fig 1d). We found expression of the IL-22Ra on cortical and medullary TECs (cTECs and mTECs, respectively), and uniform expression across both mature MHCIIhi mTEC (mTEChi) and immature MHCIIlo mTECs (mTEClo). However, in vitro stimulation of TECs with recombinant IL-22 led to enhanced TEC proliferation primarily in cTEC and mTEClo subsets (p=0.002 and 0.004 respectively). It is currently unclear if IL-22 acts as a maturation signal for mTECs, however, the uniform expression of IL-22Ra between immature mTEClo and mature Aire-expressing mTEChi, together with the preferential promotion of proliferation amongst mTEClo and cTEC seem to argue against IL-22 as a maturational signal but rather as promoter of proliferation, which ultimately leads to terminal differentiation of TECs. Of major clinical importance, administration of exogenous IL-22 led to enhanced thymic recovery (Fig. 1e) following TBI, primarily by promoting the proliferation of TECs. Consistent with this, the administration of IL-22 also led to significantly enhanced thymopoiesis following syngeneic BMT. Taken together these findings suggest that following thymic insult, and specifically the depletion of developing thymocytes, upregulation of IL-23 by DCs induces the production of IL-22 by tILCs and regeneration of the supporting microenvironment. This cascade of events ultimately leads to rejuvenation of the thymocyte pool (Fig. 1f). These studies not only reveal a novel pathway underlying endogenous thymic regeneration, but also identify a novel regenerative strategy for improving immune competence in patients whose thymus has been damaged from infection, age or cytoreductive conditioning required for successful hematopoietic stem cell transplantation. Finally, these findings may also provide an avenue of study to further understand the repair and regeneration of other epithelial tissues such as skin, lung and breast. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    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...