Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 May 21.
Published in final edited form as: Immunotherapy. 2011 Nov;3(11):1353–1366. doi: 10.2217/imt.11.126

Notch and Inflammatory T Cell Response: New Developments and Challenges

Kazuhiro Mochizuki 1, Shan He 1, Yi Zhang 1,2
PMCID: PMC4439399  NIHMSID: NIHMS534667  PMID: 22053886

Abstract

Inflammatory T cell response is important for protecting the host against infections and tumors. However, dysregulated generation of effector T cells that produce high levels of inflammatory cytokines and cytotoxic molecules may cause inflammatory disorders, such as chronic infections, autoimmune diseases and graft-versus-host disease after allogeneic bone marrow transplantation. Thus, tight regulation of effector T cells is essential. Accumulating evidence indicates that Notch plays critical roles in regulating the differentiation of antigen-activated T cells into distinct lineages of effector T cells. Furthermore, aberrant regulation of Notch signaling has been shown to implicate in several inflammatory disorders and tumorigenesis. On the other hand, significant progresses have been made in the development of new methods to modulate Notch for the disease treatment. In this review, we will discuss recent findings that help to gain insight into the impact of Notch in regulation of effector T cells and highlight the beneficial effects of modulating Notch on inflammatory diseases.

Keywords: Notch effector T cell differnetiation, inflammatory disorder, modulation of Notch signaling

INTRODUCTION

Inflammatory T cell response is a part of complex immune response against antigens, such as pathogens, autoantigens, alloantigens and tumor antigens14. Upon antigenic stimulation, naïve T cells are activated to become different lineages of effector T cells that produce high levels of inflammatory cytokines (e.g., TNF-α, IFN-γ, IL-4 and IL-17, etc.) and cytotoxic molecules (e.g., granzyme B, perforin and Fas ligand, etc.). It is well known that this differentiation of naïve T cells to different effector subsets is regulated, first by T cell receptor (TCR) and costimulatory signals, and then by cytokines14. For example, IL-12 and IL-4 induce T helper 1 (Th1) and Th2 differentiation, respectively2,5. Accumulating evidence indicate that Notch signaling plays important roles in regulating effector T cell differentiation via a mechanism independent of IL-12 and IL-469. In the setting of allogeneic bone marrow transplantation (BMT), inactivation of Notch signaling leads to reduced production of a broad spectrum of inflammatory cytokines in alloantigen-activated T cells10. Thus, Notch signaling may represent a critical pathway regulating inflammatory T cell responses.

In mammalians, there are five Notch ligands (i.e., Delta-like 1 (Dll1), Dll3, Dll4, Jagged1 (J1) and J2) and four Notch receptors (Notch 1, 2, 3 and 4)9,11,12. Notch receptor composed of an extracellular ligand-binding domain, a single-pass transmembrane domain and an intracellular domain. In the canonical signaling pathway, binding of a Notch ligand to its receptor results in the cleavage of the receptor by γ-secretase complex and the subsequent release of its Notch intracellular domain (NICD)9,1114. NICD associates with the DNA-binding factor RBP/J and co-activator Mastermind-like 1 (MAML1) in the nucleus, where it recruits additional co-activators to activate the transcription of Notch target genes. This review will focus on discussing the impact of Notch ligands and their interactions with Notch receptors in effector T cell differentiation (Fig. 1) and the beneficial effects of modulating Notch on inflammatory diseases.

Fig. 1. Notch ligand regulation of effector differentiation of antigen-activated T cells.

Fig. 1

(A), Delta-like ligand regulation of effector T cell differentiation. Activation of dendritic cells (DCs) with inflammatory stimuli upregulates the expression of Dll1 and Dll4. Dll1 can induce Th1 differentiation of antigen-activated CD4+ T cells and the generation of CD8+ cytotoxic T lymphocytes (CTL). Notch 3 and Notch 2 are found to be important for the development Th1 cells and CTLs, respectively. Whether Dll1 may affect Th17 cell differentiation remains unknown. Dll4 can promote the development of both Th1 and Th17 cells, whereas its impact on CD8+ CTL differentiation is poorly understood. Both Dll1 and Dll4 are found to repress Th2 differentiation via an unknown mechanism. (B), Jagged ligand regulation of effector T cell differentiation. J1 instructs the differentiation of antigen-specific Th2 cells. However, the impact of J1 in the generation of Th1 cells, Th17 cells and CD8+ CTL has yet to be determined. Unlike J1, J2 has been shown to be incapable of stimulating Th2 cell differentiation. In addition, whether J2 is engaged in regulation of other lineages of effector T cells has not been reported.

A. DELTA-LIKE NOTCH LIGANDS AND EFFECTOR T CELL DIFFERENTIATION

Dll1 and Dll4 induce Th1 cell differentiation

Th1 cells produce IFN-γ to promote the clearance of intracellular pathogens and inflammatory antigens2,5,6. Several studies have demonstrated that Dll1 may promote Th1 cell differentiation6,1518. In vitro stimulation of activated T cells with functional Dll1 induced Th1 cells15,16. In vivo administration of active Dll1 increased IFN-γ secretion in CD4+ T cells in mice infected with Leishmania major (L. major)15 and in mice with experimental autoimmune encephalomyelitis (EAE)18, where as blocking Dll1 by its neutralizing antibody (Ab) decreased the frequency of IFN-γ-producing CD4+T cells in these EAE mice18.

Dll4 was also reported to promote Th1 cell differentiation1922. Gain-of-function assays showed that activation of CD4+ T cells with Dll4-expressing I-Ek fibroblast L cells enhanced the generation of Th1 cells in cultures19. In contrast, soluble Dll4 fused to mutant Fc fragment of immunoglobulin G (sDll4-mFc), which can effectively compete with endogenous Dll4 on binding to Notch receptors, decreased IFN-γ production in activated CD4+ T cells without impairing their proliferation and activation19. Most importantly, Dll4 is found to be critical to the development of IL-12-independent Th1 cell differentiation19.

Dll1 and Dll4 suppress Th2 cell differentiation

Th2 cells produce IL-4, IL-5 and IL-13 to promote the control of parasite infection2,6,16,23,24 and are thought to be important for mediating allergic inflammation such as airway hyper-responsiveness and inflammation18,22,2528. Several studies suggest that both Dll1 and Dll4 may modulate Th2 cell differentiation18,22,2528. DCs over-expressing Dll1 suppressed IL-4 production in OT-II CD4+ T cells in vitro under a Th2-skewed culture condition22. In a mouse model of airway hyper-responsiveness and inflammation, administration of active Dll1-Fc led to increased levels of IFN-γ in bronchoalveolar lavage fluid but decreased Th2 responses in the lung26.

Dll4 is also found to negatively influence Th2 cell differentiation20,24. Anti-Dll4 Abtreatment caused an increase in airway hyper-responsiveness in respiratory syncytial virus (RSV)-infected animals as compared with control Ab treatment24. CD4+ T cells from the lung of these anti-Dll4 Ab-treated mice demonstrated a significant increase in Th2-type cytokines and a decrease in IFN-γ production than those derived from control mice24. In vitro culture assays showed that addition of recombinant Dll4 reduced the production of both IL-4 and IL-13 in CD4+ T cells recovered from RSV-infected mice24. Interestingly, this suppressive effect of Dll4 on Th2 cytokine production was observed not only in wild type but in MyD88 deficient lymphocytes, which suggested this suppressive effect was independent of MyD8824, a pathway used by all toll-like receptors (TLRs)29,30. A recent study also revealed the negative role of Dll4 in Th2 cell development27. In this study, in vivo anti-Dll4 Ab treatment during induction phase significantly augmented Th2 cell-mediated experimental allergic conjunctivitis in WT mice but not in IFN-γ-deficient mice27. This finding implies that the suppressive effect of Dll4 on Th2 differentiation might be associated with IFN-γ. However, a separate, in vitro study suggested that Dll4-mediated inhibition of Th2 cell development could occur independently of IFN-γ or T-bet22. Thus, an alternative explanation could be that Dll4-mediated Th2 inhibition might result in a block in IL-4-initiated commitment to the Th2 lineage22, which has yet to be formally examined.

Dll4 induces Th17 cell differentiation

Th17 cells are recently characterized Th cells that produce IL-17 and recruit neutrophils to the site of inflammation2,3133. Several experimental studies reveal an important role of Dll4 in regulating IL-17 production21,28,34. In vivo administration of neutralizing Dll4 Ab reduced the production of IL-17 and IL-17-related cytokines IL-6 and IL-21 in the lung of mice immunized with mycobacteria34. A separate study showed that in vitro treatment with anti-Dll4 Ab markedly reduced the mRNA levels of IL-17 and RORγt in mice with Theiler’s murine encephalomyelitis virus (TMEV)-induced demyelinating disease (TMEV-IDD)21. In vitro assays further demonstrate that blocking Dll4 inhibited IL-17 production by TCR transgenic DO10.11 CD4+ T cells, whereas stimulating these CD4+ T cells with active Dll4 promoted their generation of Th17 cells28. However, how Dll4 integrates other signals to promote T cell production of IL-17 and whether Dll4 is required for the generation of Th17 cells or maintenance of differentiated Th17 cells have not been reported.

Dll1 plays differential roles in the generation of CD8+ cytotoxic T lymphocytes (CTL)

The acquisition of cytotoxic effector function by antigen-specific CD8+ T cells is crucial for the control of intracellular infection and tumor invasion3. Several studies have demonstrated that Dll1 may play important roles in CD8+ CTL differentiation. DCs over-expressing Dll1 induced higher levels of granzyme B in alloantigen-activated CD8+ T cells with than control DCs35. CTL induced by Dll1-expressing DCs had enhanced capability to kill target leukemic cells35. Silencing Dll1 in DCs significantly reduced their ability to induce the generation of granzyme B-producing T cells35. Further studies showed that Dll1-expressing DCs augment CD8+CTL-mediated anti-tumor immunity36. These data suggest that Dll1 promotes CTL differentiation.

Controversially, one early study reports that constitutive expression of Dll1 on alloantigen-bearing non-professional I-Ek fibroblast L cells rendered them nonimmunogenic and able to induce specific unresponsiveness to a challenge with the same alloantigen, even in the form of a cardiac allograft37. These effects could be reversed by depletion of CD8+ T cells at the time of transplantation37. Furthermore, ligation of Dll1 on CD8+ T cells resulted in a dramatic decrease in IFN-γ with a concomitant enhancement of IL-10 production. These data suggest that Dll1 can alter the differentiation potential of CD8+ T cells37. The mechanisms by which Dll1 expressing I-Ek fibroblast L cells induce CD8+ T cell-dependent specific unresponsiveness is unclear37. Since DCs have different features in many aspects as compared to non-professional APCs such as fibroblasts, it will be intriguing to investigate whether the impact of Notch ligands such as Dll1 on T cell responses may depend on the presence of other signals delivered by APCs. (On the other hand, the effect of Dll4 on the differentiation of CD8+ cytotoxic T lymphocytes is largely unknown.)

B. JAGGED NOTCH LIGANDS AND EFFECTOR T CELL DIFFERENTIATION

The impact of Jagged Notch ligands in Th1 and Th17 has not been well characterized. A recent study reports that curdlan, one of the β-glucans derived from Candida albicans, was able to upregulate the expression of J1 in human monocyte-derived DCs that stimulated Th17 differentiation39. Addition of polyclonal anti-J1 Abinhibited DC-mediated Th17 differentiation in the culture. Further studies are needed to assess the impact of J1 in regulating Th1 and Th17 differentiation in vivo.

Jagged Notch ligands induces Th2 cell differentiation

Jagged ligands have been demonstrated to be important for the induction of Th2 responses16,18,40. Initial studies demonstrate that BM-derived DCs expressing high levels of J1 and I-Ek-fibroblast cells that were transduced with J1 stimulated Th2 cell differentiation in vitro16. Later studies suggested that in vivo administration of active J1-Fc enhanced the generation of CD4+ T cells producing IL-4 and IL-10 in EAE mice, whereas treatment of mice with anti-J1 Ab resulted in a decrease of IL-4- and IL-10-producing CD4+ T cells18. It has also been shown that in vivo treatment of J1-Fc enhanced Th2 cell response-mediated airway hyper-responsiveness and inflammation in mice40. These data support the influence of J1 on Th2 cell differentiation.

However, available data indicates that although J2 is found to be induced in response to several stimuli, including Schistosoma egg antigen (SEA), dust-mite extracts, prostaglandin E2(PGE2) and cholera toxin16,41,42, inactivation of J2 in APCs showed no effect on either Th1 or Th2 cell differentiation in vivo42. In addition, suppression of J2 expression using retrovirally delivered small interfering RNA failed to affect Th2 cell responses42.

C. NOTCH SIGNALING IN EFFECTOR T CELL DIFFERENTIATION

Notch signaling in Th1 cells

Notch ligands regulate Th1 differentiation through signaling mediated by Notch receptors. Treatment of T cells with γ-secretase inhibitors (GSI), which can block signaling of all four Notch receptors, decreased IFN-γ and IL-2 production in TCR-activated T cells(Adler et al, Palaga et al). In addition, blockade of Notch signaling by dominant-negative MAML (DNMAML), which is a pan Notch inhibitor, inhibited alloantigen-activated donor CD4+ T cells to produce IFN-γ in the setting of allogeneic BMT10. These studies provide strong evidence that Notch signaling is critical to Th1 cell differentiation.

Several studies tried to dissect the role of individual Notch receptors in Th1 cell differentiation. Some in vitro studies suggested that inhibition of Notch 1 by its antisense or neutralizing Ab reduced production of IFN-γ in activated CD4+ T cells17, Palaga,43. However, Notch 1-deficient CD4+ T cells develop Th1 immune responses in mice infected by L. major44. Furthermore, Notch 1 and Notch 2 double-deficient CD4+ T cells that were sensitized by parasite antigens showed no consistent effect on reduced production of IFN-γ45. In contrast, some other studies suggested that silencing Notch 3 resulted in a decrease in production of IFN-γ by TCR-activated T cells, whereas ectopic expression of NICD3 increased IFN-γ secretion by CD4+ T cells15. Most recently, it has been shown that in vivo treatment with anti-Notch 3 Ab but not anti-Notch 1 Ab significantly inhibited IFN-γ production of lymphocytes from mice with EAE46. It appears that Notch 3 may play a dominant role in regulating the production of Th1 type cytokine during EAE. These apparent discrepancies might be the consequence of different experimental systems.

Molecular cues directing Th1 differentiation include expression of the Th1-specific transcription factor T-bet, encoded by Tbx-21. It has been found that activation of Notch by Dll1 induces T-bet expression in activated CD4+ T cells early after stimulation22,15. This finding supports an earlier study showing that inhibition of Notch signaling by GSI extinguished expression of T-bet in activated CD4+ T cells, whereas ectopic expression of activated Notch 1 restored Tbx21 transcription17. Activated Notch binds to the RBP/J(CSL)complex, displacing the SMART-SKIP corepressors, and recruits the coactivator MAML1 to convert RBP/J(CSL) to a transcriptional activator. Chromatin immunoprecipitation assay (ChIP) revealed that both active Notch 1 and RBP/J bound to the promoter region of Tbx21, suggesting the direct effect of Notch on regulating Tbx21 transcription17. This may explain that Dll1 or Dll4 activation of Notch signaling induce T-bet expression independent of IL-1222,15. However, controversial results were reported about the role of IFN-γ in Delta like ligand mediated Th1 cell development. Early syudy showed the neutralizing IFN-γ reduced T-bet expression in Dll1 or Dll4 inducing CD4+ T cells and the subsequent generation of Th1 cells22, while recent study demonstrated the independence of anti-IFN-γ regulating T-bet expression.

Other studies also show that Notch may promote the generation of Th1 cells by interacting with the NF-kB pathway. Complexes of NICD1 and NF-kB1 (p50) or NICD1 and RelA (p65) could be immunoprecipitated on the Ifng promoter, in regions lacking RBP/J(CSL) binding sites4749. This raises the possibility that Notch 1 can contribute to Th1 cell differentiation in a RBP/J(CSL)-independent manner4749. Interestingly, activated Notch may also involve in molecular pathway(s) associated with TCR signaling46. Inhibition of Notch in T cells is correlated with the down-regulated expression of protein kinase C (PKC) θ, which is an important regulator of the mature T cell activation and polarization46.

Notch signaling in Th2 cells

Notch signaling is an important regulator of type 2 immunity. Initial studies suggest that inactivation of RBP/J in mature T cells resulted in the defect in production of IgG1 and IgE, an indication of Th2-related immune responses50. Inactivation of RBP/J in mature T cells also reduced their capability to generate Th2 cells producing IL-4 upon in vitro stimulation with LPS-pretreated MyD88-deficient DC and in vivo in mice immunized with SEA16. Later studies demonstrate that mature T cells expressing the pan Notch inhibitor DNMAML failed to develop protective Th2 responses against the gastro intestinal helminth Trichurismuris51, suggesting the critical role of Notch in regulating Th2 cell differentiation. However, mice lacking individual Notch genes have no defect in T helper differentiation45. In contrast, Notch 1-and Notch 2-double deficient CD4+ T cells failed to make IL-445. These experiments reveal that either Notch 1 or Notch 2 are required for Th2 cell differentiation.

The transcription factor GATA3 is critically required for promoting IL-4 production and Th2 cell differentiation2,5,6,45,51,52. Several studies indicate that GATA3 is a direct target of Notch signaling. Ectopic expression of active NICD greatly increases the expression of GATA3, whereas inactivation of Notch signaling results in markedly diminished GATA3 expression45,52. Furthermore, mature T cells lacking RBP/J have significantly reduced expression of GATA3 as compared to WT T cells and loss of RBP-J may cause the failure of efficient induction of GATA3 expression, leading to impaired Th2 differentiation50. ChIP assays confirm that both active Notch and RAB/J directly bind to the promoter region of Gata3 to regulating the gene transcription45,52. Interestingly, several RBP/J(CSL) binding sites are found in enhancer region downstream of the Il4 coding region, and one site in the Il4 promoter16,52. Therefore, Il4 gene is also a direct target of Notch effector16. This may explain that naïve T cells lacking STAT6, which is required for IL-4 mediated Th2 cell differentiation, retained their capability to differentiate into Th2 cells. Furthermore, ectopic expression of either NICD1 or NICD2 in STAT6-deficient T cells promotes IL-4 production in effector T cells16,52. Thus, Notch signaling acts on both Il4 and GATA3 genes to activate IL-4 expression52.

Notch signaling in Th17 cells

Recent studies suggest that Notch signaling may play an important role in Th17 cells. Inhibition of Notch signaling by the pan Notch inhibitor DNMAML drastically reduced the number of alloreactive CD4+ T cells producing IL-17 in the setting of allogeneic BMT10. Treatment of EAE mice induced by proteolipid protein (PLP) immunization with anti-Notch 3 Ab, but not anti-Notch 1 Ab, impaired Th17 responses, leading to amelioration of EAE in animals46. Thus, it appears that Notch 3 may play an important role in Th17 cells.

Several studies show that Dll4 activation of Notch signaling results in increased production of IL-17 in activated CD4 T cells21,28,34. Rorc, which is a transcription factor required for Th17 differentiation2,31,32, was significantly increased in CD4+ T cells cultured under Th17 skewing condition, and further enhanced when cells were incubated with Dll4 in a time dependent fashion28. A second required factor for Th17 differentiation is STAT3 protein activation (pSTAT3)2,31,32. While Th17 skewing condition increased pSTAT3, addition of Dll4 did not alter pSTAT3 in these cells. Thus, increased Th17 cell differentiation by Dll4 appears to be associated with increased expression of Rorc, but not altered STAT3 activation 28.

Notch signaling in CD8+effector T cells

Notch signaling directly regulates the transcription of effector molecules in CTLs. Naïve CD8+ T cells express higher levels of Notch 2 on their surface than Notch 1 as assessed by flow cytometry, whereas activated CD8+ T cells expressed similarly high levels of Notch 1 and Notch 235. In contrast, both naïve and activated CD8+ T cells expressed extremely low levels of Notch 3 and Notch 435. Conditional inactivation of Notch 2 but not Notch 1 in mature CD8+ T cells led to impaired generation of functional CTLs35,36. Mechanistic analysis revealed that active NICD2 interacted with a phosphorylated form of the transcription factor CREB1, and subsequently bound to the transcription coactivator p300 to form a complex on the promoter of the gene encoding granzyme B35. Thus, Notch signaling may control CTL differentiation through phosphorylated CREB1 and p300 coactivator35.

Transcription factors T-bet and Eomes control CD8+ CTL differentiation1,3. Previous reports have suggested that Notch could control the expression of these genes17,35,36,49. Notch 1 signaling was also found to regulate the expression of perforin and granzyme B through direct binding to the promoters of these crucial effector molecules54. However, Notch 2-mediated CTL differentiation appears to be independent of Eomes35. Interestingly, a recent study demonstrates that inhibition of pan Notch signaling by DNMAML reduced the expression of granzyme B and perforin in alloantigen-activated CD4+ and CD8+ T cells without altering the expression of T-bet and Eomes10. Furthermore, Tbx21 mRNA was preserved in alloreactive DNMAML T cells, while Eomes transcripts were significantly elevated10. These observations suggest that Notch signaling-mediated CD8+ CTL differentiation may involve multiple molecular pathways.

D. REGULATION OF NOTCH LIGAND EXPRESSION

Notch ligand in APCs

APCs are crucial in directing the differentiation of distinct subsets of effector T cells6,55. We now appreciate that Notch ligands presented by APCs are also important for promoting the generation of different lineages of effector T cells6. Many studies have demonstrated that different inflammatory stimuli may stimulate APCs to express different type of Notch ligands at different levels6,15,16,1820,24,28,35,56. As summarized in Table 1, TLR ligands or TLR agonists stimulate APC expression of both Delta ligands and Jagged ligands, whereas other inflammatory stimuli such as helminthes, allergens, toxins and PGE2 may cause the selective increase in the expression of Jagged ligands6,15,16,1820,24,28,35,56. These observations could explain, in part, how different pro-inflammatory stimuli induce the development of different type of immune responses.

Table I.

Regulation of Notch ligand expression.

A. TLR stimulation
Cells Stimulation mRNA Reference
Dll1 Dll4 J1 J2
Splenic DC: CD8(−)/CD11c(+) LPS No change Increase No change No change 19
Mediastinal LN DC: CD11c(+) OVA + LPS(hi) Increase Increase 62
OVA + LPS(lo) No change Increase 62
BM DC LPS Increase Increase No change No change 28
LPS Increase Increase No change 16
LPS Increase Decrease 41
Monocyte derived DC LPS + R848 Increase Decrease 63
B. Non-TLR stimulation
Cells Stimulation mRNA Reference
Dll1 Dll4 J1 J2
Astrocyte TGF-β Increase 18
IFN-γ, TNF-α Decrease 18
BM DC CT No change Increase 16
CT Increase Increase 41
BM DC SEA No change No change No change No change 42
SEA Increase Increase 41
BM DC PGE2 No change Increase 16
PGE2 Increase Increase 41
BM DC RSV Increase Increase 28
RSV Increase 24
BM DC TMEV Increase 21
BM DC P. acnes No change Increase No change Decrease 42
P. acnes Increase Decrease 41

APC expression of Notch ligands under steady condition

Flow cytometry analysis of Notch ligand revealed that under steady condition different APC subsetsin lymphoid organs expressed distinct patterns of Notch ligands (Table 1). Myeloid CD11b+CD11c+ cells expressed low to moderate level of Dll1, J1 and J2 but not Dll4, whereas lymphoid CD11b CD11c+ cells expressed low to moderate levels of Dll1 and J2 but not Dll4 and J157. A similar pattern of Notch ligand expression in CD8+CD11c+ DCs and CD8 CD11c+ DCs has been reported58. Macrophages expressed low to moderate levels of Dll1 and Jagged 2 but not Dll4 and J159. Interestingly, plasmacytoid DCs (pDCs) expressed high levels of Dll1 and Dll4 but lacked J1 and J257. These pDCs had greater ability than either myeloid or lymphoid DCs to induce the generation of IL-10-producing Th1 cells. This effect of pDCs could be reduced by GSI or by inactivating the Notch effector RBP/J(CSL)57. Whether these Notch ligands impact mature T cells under steady condition and what kind of Notch ligands is expressed in APCs resided in non-lymphoid tissues have yet to be carefully examined.

TLR ligand induction of Notch ligands in APCs

TLRs are potent stimuli for the induction of Notch ligands in DCs16,19,24,28,6063. These included TLR-specific signals, poly(I:C) (TLR3), LPS (TLR4), and CpG (TLR9), as well as RSV, which activates by both RIG-I and TLR-mediated pathways16,25,28,6163,19. LPS could rapidly induce Dll4 expression in DCs in mice6163. Similar results were observed by using human monocyte-derived DCs stimulated by LPS63. Combined stimulation of human DCs with LPS with TLR7 agonist R848 further increased the expression of Dll4 and down-regulated J163. These data suggest that activation of TLRs may lead to opposing effects on the expression of Notch ligands in DCs. Notably, while LPS induction of Dll4 in BM-derived DCs was found to be dependent on MyD8816, cytokines including IL-1β and IL-18, which are two IL-1R family MyD88-dependent signals, did not induce the expression of any Notch ligands28. This suggests that activation of MyD88 is not sufficient to the induction of Dll4 in DCs. Further studies are necessary to determine the molecular mechanisms by which LPS induces Dll4 expression in APCs.

An elegant study recently demonstrates that high but not low concentration of LPS induced a marked upreguration of both Dll4 and J1 mRNA in DCs in mice.62 In contrast Dll4 mRNA expression was only weakly upregulated by low concentration of LPS, while it was sufficient to lead to the upregulation of J162. This coincided with the finding that high concentration of LPS induces Th1 and low concentration of LPS resulted in Th2 responses in an allergic lung inflammatory disease mouse model62. Furthermore, LPS has different effects on regulating Notch ligand expression in distinct APC subsets. Skokos et al. report that LPS stimulation rapidly induced Dll4 expression in CD8 DCs but not in CD8+ DCs19. In contrast, Dll1, Dll3, J1 and J2 were not up-regulated in these DCs upon LPS stimulation19. It appears that different mechanisms may be involved in regulating the expression of distinct Notch ligands in APCs.

Non-TLR ligand regulation of Notch ligands in APCs

Several studies have demonstrated that different antigens may possess the ability to regulate the expression of distinct Notch ligands in APCs. SEA stimulation moderately enhanced the expression of J1 and J2, whereas P. acnes infection resulted in notable reduction in expression of J2 in DCs41. Other studies showed the stimulation of DCs with P. acnes induces the expression of both Dll1 and Dll4, with Dll4 the most extent42. In an EAE mouse model, immunization of mice with antigen MOGe mulsified in CFA induced the expression of Dll1 and J1 mRNA in DCs in the spleen and lymph node, but did not affect the expression of Notch ligands in macrophages18. Interestingly, viruses such as RSV and TMEV also increase the expression of Dll4 in DCs 21,24,25. Thus, a complex mechanism is involved in regulating the expression of Notch ligands in APCs during immune response. Some studies suggested that proinflammatory cytokines were able to regulate Notch ligand expression during inflammatory response. In vitro assays showed that IFN-γ and TNF-α down-regulated the expression of J1 on astrocytes. In contrast, a significant increase in J1 mRNA levels was detected in cells exposed to TGF-β118. Cholera toxin and PGE-2 are found to upregulate the expression of J216. However, none of these inflammatory stimuli shows the impact on APC expression of Dll416,18

Non APC expressing Notch ligands

In slot blot analysis, Dll1 was detected in all immature lineages as well as mature hematopoietic cells. In the same analysis, expression of J1 transcripts was restricted mainly to terminally maturing cells, most strikingly to mast cells and megakaryocytes, and at lower levels in B cells and macrophages (Singh, JExp Hem 2000). In a separate study, bone marrow stroma cell (BMS) were shown to express a substantially higher level of J1 than D1, while spleen stroma (SS) had just the opposite expression pattern. Interestingly, those two stroma cells have different roles in DC development. BMS stimulated accumulation of DC precursors but prevent their transition to terminally differentiated DCs, while SS promoted generation of fully differentiated DCs (Cheng, Blood 2006). Another study showed that CD4+ and CD8+ T cells also express mRNA of J1 more abundant in DC (Hoyone. IntImm 2000). In Airway hyperresponsiveness (AHR) mouse model, increased expression of Dll1 in effector CD8+ T cells were shown which skewed the T cell response from a Th1 to Th2 phenotype (26). Although non APC expressing Notch ligands are intriguingly investigated, it is still controversial that the precise roles of those Notch ligands in effecter T cell deferentiation.

E. MODULATION OF NOTCH SIGNALING TO CONTROL INFLAMMATORY DISEASES

Approaches to modulate Notch signaling

Significant progresses have been made in understanding the impact of each individual Notch ligands and receptors in various inflammatory conditions6. These studies clearly demonstrate that Notch signaling may play critical and multiple roles in inflammatory T cell-mediated diseases6. Both genetic and pharmacologic approaches have been used for modulating Notch to control inflammatory diseases and improve tumor immunity. These include the pharmacologic Notch inhibitor GSI, neutralizing Abs specific for Notch receptors and ligands, ectopic expression of Notch ligands and Notch receptors and transgenic expression of pan Notch inhibitor-DNMAML.

GSI has been in the clinical development for the treatment of advanced solid tumor, acute T lymphoblastic leukemia and Alzheimer’s disease64,72. Preclinical experimental studies suggest that mice treated with a GSI resulted in significantly delayed onset of EAE, reduced clinical symptoms and decreased inflammatory lesions in the spinal cord17. This inhibitory effect of GSI was associated with significantly decreased proliferation and reduction of Th1-associated with cytokines17. Treatment of systemic lupus erythematosis (SLE) mice with GSI resulted in a reduction of autoreactive T cells and a decrease in production of inflammatory cytokines (e.g., IFN-γ, TNF-α, IL-6, and IL-2) and improved nephritis65. Despite the potential benefit in cancer and immune mediated disease, GSI treatment causes significant accumulation of goblet cells in the small intestine66. This results in severe diarrhea and has hindered the clinical application67,68. However, progress has recently been made in reducing the adverse effects of GSI on GI tract, while preserving its beneficial effects 67.

Abs that specifically antagonize Notch 1 and Notch 2 have been developed to elucidate the discrete functions of Notch 1 and Notch 2 and reduce intestine toxicity69. These Abs specifically reacted to each receptor paralogue and cross-reacted with the human and mouse sequences, enabling the discrimination of Notch 1 versus Notch 2 function in human patients and rodent models69. Selectively blocking of Notch 1 inhibited cancer cell growth and induced deregulation of angiogenesis. Importantly, both antibodies inhibited signaling induced through the ligands J1, J2, Dll1 and Dll4, and thus inhibition occurs irrespective of the ligand69. Furthermore, whereas inhibition of Notch 1 plus Notch 2 caused severe intestinal toxicity, inhibition of either receptor alone reduced or avoided this effect, demonstrating a clear advantage over pan-Notch inhibitors69.

Alternatively, targeting individual Notch ligands may have significant implications in modulating Notch signaling. Many Abs specific to Notch ligands are available now21,27,35,53,59,70. Different Notch ligands may differentially regulate the outcome of inflammatory responses via interacting with distinct Notch receptor. It is possible that blocking one Notch ligand may allow the other Notch ligands to trigger the corresponding Notch receptor, leading to the preservation of some Notch functions in the target cells and possible reduction of toxicity resulted from inhibition of pan Notch signaling10.

Notch and multiple sclerosis (MS)

Tight regulation of Th1 and Th2 responses is critical, as aberrant Th1 conditions perpetuate destructive inflammation characteristic of autoimmune diseases such as MS7176. MS is a chronic demyelinating disease of the human central nervous system (CNS). Although its etiology remains unclear, MS is generally considered to be an autoimmune Th1/Th17-mediated disease7176. EAE represents a useful animal model for studying human MS, in which antigen-specific CD4+ Th1 and Th17 cells mediate inflammatory damage in CNS, with consequent demyeliation and progressive paralysis7176. Data from recent studies indicate that selective inhibition of Notch 3 by its neutralizing Ab ameliorated EAE46,77. Furthermore, treatment of EAE mice with anti-Dll1 Abor active J1-Fc from the induction phase led to reduced CD4+ T cells secreting IFN-γ, increased frequency of CD4+ T cells producing IL-4 and IL-10, and the subsequent amelioration of the disease18.

A separate study demonstrates that administration of anti-Dll4 mAb ameliorated demyelinating disease induced by TMEV21. Treatment with mAb to Dll4, especially during the effector phase, resulted in significantly decreased expression of transcripts of Th1- and Th17-derived cytokines, reduced number of infiltrating mononuclear inflammatory cells in the spinal cords and suppression of the disease development21. Thus, aberrant activation of Notch signaling by Dll4 may be also important to the development of Th1 cell-mediated demyelinating disease, in particularly during effector phase. Most recently, in EAE mouse model, anti-Dll4 treatment was shown to reduce T cell surface expression of the CCR2 and CCR6, and decreased clinical severity with diminished T cell accumulation in CNS. This result suggests a role of Dll4 in EAE pathogenesis through modulation of T cell chemokine receptor expression and migration to the CNS (Reynolds JI 2011). All these observations suggest that Notch ligands or Notch receptors may be new and attractive targets in treating MS.

Notch and airway hyper-responsiveness and inflammation

A dysregulated Th2 milieu can promote airway hyper-responsiveness and inflammation20,23,24,26,40. Allergic asthma is characterized by persistent airway hyperresponsiveness and inflammation due to cellular and molecular responses induced by allergen in sensitized hosts20,23,24,26,40. Antigen-specific memory T cells, especially CD4+ T cells, are believed to play an integral role in orchestrating the disease process through the secretion of a variety of Th2 cytokines, including IL-4, IL-5 and IL-1320,23,24,26,40. Treatment of allergen-sensitized mice with active Dll1-Fc resulted in decreased airway hyper-responsiveness and inflammation in response to secondary allergen rechallenge26. This effect was associated with a significant increase in number of CD4+ T cells producing IFN-γ and the reduction of eosinophils in the lung tissues26.

Emerging evidence indicate that Dll4 also plays important roles in the pathogenesis of airway hyper-responsiveness. In vivo administration of anti-Dll4Ab substantially increased airway hyper-responsiveness in mice infected by RSV26. Examination of lymphocytic lung infiltrate revealed a significant increase in total CD4+ T cells and activated CD8+ T cells in RSV-infected mice following anti-Dll4 Ab treatment. These lung CD4+ T cells produced high levels of type 2 inflammatory cytokines25. All these observations suggest that both Dll1 and Dll4 could be therapeutic targets for modulating airway hyper-responsiveness and inflammation.

Notch and graft-versus-host disease (GVHD)

GVHD is caused by donor T cells that recognize and react to alloantigens of the recipient. Upon APC activation, donor T cells become alloreactive effector T cells producing high levels of inflammatory cytokines (e.g., TNF-α, IFN-γ, IL-4 and IL-17) and cytotoxic molecules, leading to host tissue injury and the subsequent life-threatening complication7885. However, blockade of individual effector molecules has limited efficacy in controlling GVHD79,85. We recently demonstrate that Notch signaling is a potent regulator of T cell activation, differentiation and function during acute GVHD10. Inhibition of canonical Notch signaling by expressing pan Notch inhibitor DNMAML in donor T cells markedly reduced GVHD severity and mortality in mouse models of allogeneic HSCT. Whereas Notch-deprived T cells proliferated and expanded in response to alloantigens in vivo, they were incapable of producing several inflammatory cytokines, including TNF-α, IFN-γ, IL-4 and IL-1710. Furthermore, inactivation of Notch resulted in a decrease in expression of cytolytic effector molecules in both CD4+ and CD8+ T cells activated by alloantigens10. Most importantly, Notch-deprived alloreactive CD4+ T cells retained significant cytotoxic potential and anti-leukemia activity, leading to improved overall survival of the recipients. These results identify Notch as a novel essential regulator of pathogenic CD4+ T cell responses during acute GVHD and suggest that Notch signaling in T cells should be investigated as a therapeutic target following allogeneic HSCT10.

This work presents the first example of a disease model in which Notch inhibition prevents otherwise lethality in recipients after allogeneic BMT10. Since systemic Notch inhibition can cause adverse reactions, in particular gastrointestinal toxicity, strategies are needed to overcome this important limitation. To this end, further experiments are needed to identify the individual Notch ligand(s) and Notch receptor(s) that are critical for the induction of effector differentiation of alloantigen-activated T cells.

Notch and cancer immunotherapy

CD8+ T cells play a central role in cancer immune surveillance, and the efficient induction of CTLs against tumor antigens is required for successful immunotherapy for cancer patients86,87. Several studies suggest that Notch signaling is critical to effector differentiation of antigen-activated CD8+ T cells17,35. For instance, Dll1 activation of Notch 2 signaling directly regulates granzyme B expression and promotes CTL differentiation.35. Consistent with their higher expression of granzyme B, CD8+ T cells stimmulated with Dll1 upregurated DC lysed allogeneic EL-4 (mouse thymoma) target cells more efficientry than those stimulated with control DC. Most recently, Sugimoto et al. report that Notch 2 signaling is required for potent anti-tumor immunity in vivo36. Mice with Notch 2-deficient-but not Notch 1-deficient-CD8+ T cells die earlier than control mice after inoculation with OVA-expressing EG7 thymoma cells36. These data suggest that new strategies capable of enhancing Notch 2 signaling in CD8+ T cells may increase the anti-tumor activity in vivo. Indeed, administration of anti-Notch 2 agonist Ab or Dll1-overexpressing DCs augment tumor immunity36.

F. FUTURE PERSPECTIVES

Significant progresses have been made during the past few years in understanding the important roles of Notch in inflammatory T cell responses. However, many critical issues have yet to be determined. For example, why do distinct Notch ligands have differential roles in regulating development of different lineages of effector T cells? It has been shown that while Dll1 and Dll4 promote the development of Th1 response, they may repress the production of Th2-type immune responses under some circumstances20,24. In sharp contrast, J1 can instruct Th2 cell differentiation but has little effect on the generation of Th1 cells6,16,18,40,45. One possible explanation is that different Notch ligands may activate distinct Notch receptors for the subsequent development of Th1 and Th2 cells. Indeed, whereas Notch 3 is critical to Th1 differentiation15,17,46, both Notch 1 and Notch 2 play a redundant role in regulating the development of Th2 cells45. However, there is no evidence indicating that J1 is incapable of binding to Notch 36,15,16,45,70. Furthermore, both Dll1 and Dll4 were also shown to be able to bind to both Notch 1 and Notch 269,70. It would be interesting to identify whether additional mechanisms such as co-stimmulatory/co-inhibitory signals, or Notch ligands on other cell popurations such as stroma cells may determine the consequence of Notch regulation of effector differentiation from antigen-activated T cells.

Another important question is what the role of Notch ligands is in regulating APC development and function. Accumulating evidence indicate that Notch receptors are highly expressed in APCs and Notch signaling may be critical to the development and functional differentiation of APCs58,8890. Inactivation of the Notch effector RBP/J(CSL)led to impaired differentiation and survival of DCs88. Blocking of Notch ligands drastically reduced the number of DCs58. Thus, a better understanding of the role of Notch in APC generation and function will be important for the development of novel strategies to modulate inflammatory responses.

Executive summary.

Introduction

Delta-like Notch ligands and effector T cell differentiation

  • Both Dll1 and Dll4 promote Th1 cell differentiation.

  • Dll4 is related the development of IL-12 independent Th1 cell differentiation.

  • Both Dll1 and Dll4 supress Th2 cell differentiation.

  • Dll4 is related the promotion of Th17 cell differentiation.

  • Dll1 may promote CD8+ cytotoxic T lymphocyte differentiation.

Jagged Notch ligands and effecter T cell differentiation

  • J1 induces Th2 cell differentiation.

  • The role of J2 in effector T cell differentiation is unclear.

Notch signalling in effecter T cell differentiation

  • Notch signaling is clitical to Th1 cell differentiation.

  • Notch signalling acts GATA3 directry, and promote Th2 cell differentiation.

  • Notch signalling acts RORγt directory, and may play a role in Th17 cell differentiation.

  • Notch signalling directory regurates the transcription factor CREB1 and EOMES in CD8+ effector T cells.

Reguration of Notch ligand expression

  • Under steady condition, different APC subset expressed distinct pattern of Notch ligands (?).

  • TLRs stimulate APC expression of both Delta and Jagged ligands (Table ?).

  • Non-TLR ligands have the ability to regulate the expression of distinct Notch ligands in APC (Table ?).

  • The precise role of non-APC expressing Notch ligands is yet to be determined.

Modulation of Notch signalling to control inflammatory diseases

  • Both genetic and pharmacologic approaches have been used for modulating Notch to control inflammatory diseases and improve tumor immunity.

  • These include the pharmacologic Notch inhibitor GSI, neutralizing Abs specific for Notch receptors and ligands, ectopic expression of Notch ligands and Notch receptors.

  • GIS,

  • Abs to Notch R,

  • Abs to Notch Ligands….

  • Engineering APCs

  • Engineering stromal cells/fibroblast cells

  • Notch 3, Dll1, Dll4, and J1 may be new targets in treating MS.

  • Both Dll1 and Dll4 could be therapeutic targets for modulating airway hyper-responsiveness.

  • Notch signalling is an essential regulator of allogeneic CD4+ T cell response during acute GVHD, and should be investigated as a therapeutic target following HSCT.

  • Enhancing Notch 2 signalling in CD8+ T cells may increase the anti-tumor activity in vivo.

Acknowledgments

This work was supported by a Damon Runyon-Rachleff Innovation Award (YZ) and the American Cancer Society (YZ).

References

  • 1.Reiner SL. Epigenetic control in the immune response. Hum Mol Genet. 2005;14(Spec No 1):R41–46. doi: 10.1093/hmg/ddi115. [DOI] [PubMed] [Google Scholar]
  • 2.Zhu J, Yamane H, Paul WE. Differentiation of effector CD4 T cell populations (*) Annu Rev Immunol. 2010;28:445–489. doi: 10.1146/annurev-immunol-030409-101212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Fearon DT. The expansion and maintenance of antigen-selected CD8(+) T cell clones. Adv Immunol. 2007;96:103–139. doi: 10.1016/S0065-2776(07)96003-4. [DOI] [PubMed] [Google Scholar]
  • 4.Lanzavecchia A, Sallusto F. Progressive differentiation and selection of the fittest in the immune response. Nat Rev Immunol. 2002;2:982–987. doi: 10.1038/nri959. [DOI] [PubMed] [Google Scholar]
  • 5.O’Shea JJ, Paul WE. Mechanisms underlying lineage commitment and plasticity of helper CD4+ T cells. Science. 2010;327:1098–1102. doi: 10.1126/science.1178334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Amsen D, Antov A, Flavell RA. The different faces of Notch in T-helper-cell differentiation. Nature reviews. 2009;9:116–124. doi: 10.1038/nri2488. [DOI] [PubMed] [Google Scholar]
  • 7.Osborne BA, Minter LM. Notch signalling during peripheral T-cell activation and differentiation. Nature reviews. 2007;7:64–75. doi: 10.1038/nri1998. [DOI] [PubMed] [Google Scholar]
  • 8.Sandy AR, Maillard I. Notch signaling in the hematopoietic system. Expert opinion on biological therapy. 2009;9:1383–1398. doi: 10.1517/14712590903260777. [DOI] [PubMed] [Google Scholar]
  • 9.Radtke F, Fasnacht N, Macdonald HR. Notch Signaling in the Immune System. Immunity. 2010;32:14–27. doi: 10.1016/j.immuni.2010.01.004. [DOI] [PubMed] [Google Scholar]
  • 10.Zhang Y, et al. Notch signaling is a critical regulator of allogeneic CD4+ T cell responses mediating graft-versus-host disease. Blood. 2011 doi: 10.1182/blood-2010-03-271940. The first report that Notch can be the target for modulation of allogeneic T cell response. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Maillard I, Fang T, Pear WS. Regulation of lymphoid development, differentiation, and function by the Notch pathway. Annual review of immunology. 2005;23:945–974. doi: 10.1146/annurev.immunol.23.021704.115747. [DOI] [PubMed] [Google Scholar]
  • 12.Grabher C, von Boehmer H, Look AT. Notch 1 activation in the molecular pathogenesis of T-cell acute lymphoblastic leukaemia. Nat Rev Cancer. 2006;6:347–359. doi: 10.1038/nrc1880. [DOI] [PubMed] [Google Scholar]
  • 13.Gordon WR, Arnett KL, Blacklow SC. The molecular logic of Notch signaling--a structural and biochemical perspective. J Cell Sci. 2008;121:3109–3119. doi: 10.1242/jcs.035683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kopan R, Ilagan MX. The canonical Notch signaling pathway: unfolding the activation mechanism. Cell. 2009;137:216–233. doi: 10.1016/j.cell.2009.03.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Maekawa Y, et al. Delta1-Notch 3 interactions bias the functional differentiation of activated CD4+ T cells. Immunity. 2003;19:549–559. doi: 10.1016/s1074-7613(03)00270-x. Along with (15,16), The first report that Notch signaling is important for Th1/Th2 differentiation. [DOI] [PubMed] [Google Scholar]
  • 16.Amsen D, et al. Instruction of distinct CD4 T helper cell fates by different notch ligands on antigen-presenting cells. Cell. 2004;117:515–526. doi: 10.1016/s0092-8674(04)00451-9. [DOI] [PubMed] [Google Scholar]
  • 17.Minter LM, et al. Inhibitors of gamma-secretase block in vivo and in vitro T helper type 1 polarization by preventing Notch upregulation of Tbx21. Nature immunology. 2005;6:680–688. GSI regulating of Th1. [PubMed] [Google Scholar]
  • 18.Elyaman W, et al. JAGGED1 and delta1 differentially regulate the outcome of experimental autoimmune encephalomyelitis. J Immunol. 2007;179:5990–5998. doi: 10.4049/jimmunol.179.9.5990. [DOI] [PubMed] [Google Scholar]
  • 19.Skokos D, Nussenzweig MC. CD8− DCs induce IL-12-independent Th1 differentiation through Delta 4 Notch-like ligand in response to bacterial LPS. J Exp Med. 2007;204:1525–1531. doi: 10.1084/jem.20062305. Dll4 is important for Il-12 independent Th1 differentiation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jang S, Schaller M, Berlin AA, Lukacs NW. Notch ligand delta-like 4 regulates development and pathogenesis of allergic airway responses by modulating IL-2 production and Th2 immunity. J Immunol. 2010;185:5835–5844. doi: 10.4049/jimmunol.1000175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Takeichi N, et al. Ameliorating effects of anti-Dll4 mAb on Theiler’s murine encephalomyelitis virus-induced demyelinating disease. Int Immunol. 2010;22:729–738. doi: 10.1093/intimm/dxq059. [DOI] [PubMed] [Google Scholar]
  • 22.Sun J, Krawczyk CJ, Pearce EJ. Suppression of Th2 cell development by Notch ligands Delta1 and Delta4. J Immunol. 2008;180:1655–1661. doi: 10.4049/jimmunol.180.3.1655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Cohn L, Homer RJ, Marinov A, Rankin J, Bottomly K. Induction of airway mucus production By T helper 2 (Th2) cells: a critical role for interleukin 4 in cell recruitment but not mucus production. J Exp Med. 1997;186:1737–1747. doi: 10.1084/jem.186.10.1737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Schaller MA, et al. Notch ligand Delta-like 4 regulates disease pathogenesis during respiratory viral infections by modulating Th2 cytokines. J Exp Med. 2007;204:2925–2934. doi: 10.1084/jem.20070661. anti-Dll4 antibody regurating Th2 immunity. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Schaller MA, et al. Delta-like 4 differentially regulates murine CD4 T cell expansion via BMI1. PLoS One. 2010;5:e12172. doi: 10.1371/journal.pone.0012172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Okamoto M, et al. Essential role of Notch signaling in effector memory CD8+ T cell-mediated airway hyperresponsiveness and inflammation. J Exp Med. 2008;205:1087–1097. doi: 10.1084/jem.20072200. Dll1 skews Th2 to Th1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Fukushima A, et al. Notch ligand Delta-like4 inhibits the development of murine experimental allergic conjunctivitis. Immunol Lett. 2008;121:140–147. doi: 10.1016/j.imlet.2008.10.006. [DOI] [PubMed] [Google Scholar]
  • 28.Mukherjee S, Schaller MA, Neupane R, Kunkel SL, Lukacs NW. Regulation of T cell activation by Notch ligand, DLL4, promotes IL-17 production and Rorc activation. J Immunol. 2009;182:7381–7388. doi: 10.4049/jimmunol.0804322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Iwasaki A, Medzhitov R. Toll-like receptor control of the adaptive immune responses. Nature immunology. 2004;5:987–995. doi: 10.1038/ni1112. [DOI] [PubMed] [Google Scholar]
  • 30.Iwasaki A, Medzhitov R. Regulation of adaptive immunity by the innate immune system. Science. 2010;327:291–295. doi: 10.1126/science.1183021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Weaver CT, Hatton RD, Mangan PR, Harrington LE. IL-17 family cytokines and the expanding diversity of effector T cell lineages. Annual review of immunology. 2007;25:821–852. doi: 10.1146/annurev.immunol.25.022106.141557. [DOI] [PubMed] [Google Scholar]
  • 32.Stockinger B, Veldhoen M. Differentiation and function of Th17 T cells. Curr Opin Immunol. 2007;19:281–286. doi: 10.1016/j.coi.2007.04.005. [DOI] [PubMed] [Google Scholar]
  • 33.Harrington LE, et al. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nature immunology. 2005;6:1123–1132. doi: 10.1038/ni1254. [DOI] [PubMed] [Google Scholar]
  • 34.Ito T, et al. TLR9 regulates the mycobacteria-elicited pulmonary granulomatous immune response in mice through DC-derived Notch ligand delta-like 4. J Clin Invest. 2009;119:33–46. doi: 10.1172/JCI35647. Along with (28,34), Dll4 regurates Th17 cell differentiation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Maekawa Y, et al. Notch 2 integrates signaling by the transcription factors RBP-J and CREB1 to promote T cell cytotoxicity. Nature immunology. 2008;9:1140–1147. doi: 10.1038/ni.1649. Notch signal promotes CTL differentiation. [DOI] [PubMed] [Google Scholar]
  • 36.Sugimoto K, et al. Notch 2 signaling is required for potent antitumor immunity in vivo. J Immunol. 2010;184:4673–4678. doi: 10.4049/jimmunol.0903661. [DOI] [PubMed] [Google Scholar]
  • 37.Wong KK, et al. Notch ligation by Delta1 inhibits peripheral immune responses to transplantation antigens by a CD8+ cell-dependent mechanism. J Clin Invest. 2003;112:1741–1750. doi: 10.1172/JCI18020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Eagar TN, et al. Notch 1 signaling regulates peripheral T cell activation. Immunity. 2004;20:407–415. doi: 10.1016/s1074-7613(04)00081-0. [DOI] [PubMed] [Google Scholar]
  • 39.Higashi T, et al. Curdlan induces DC-mediated Th17 polarization via Jagged1 activation in human dendritic cells. Allergol Int. 2010;59:161–166. doi: 10.2332/allergolint.09-OA-0103. [DOI] [PubMed] [Google Scholar]
  • 40.Okamoto M, et al. Jagged1 on dendritic cells and Notch on CD4+ T cells initiate lung allergic responsiveness by inducing IL-4 production. J Immunol. 2009;183:2995–3003. doi: 10.4049/jimmunol.0900692. [DOI] [PubMed] [Google Scholar]
  • 41.Krawczyk CM, Sun J, Pearce EJ. Th2 differentiation is unaffected by Jagged2 expression on dendritic cells. J Immunol. 2008;180:7931–7937. doi: 10.4049/jimmunol.180.12.7931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Worsley AG, et al. Dendritic cell expression of the Notch ligand jagged2 is not essential for Th2 response induction in vivo. Eur J Immunol. 2008;38:1043–1049. doi: 10.1002/eji.200737335. [DOI] [PubMed] [Google Scholar]
  • 43.Luty WH, Rodeberg D, Parness J, Vyas YM. Antiparallel segregation of notch components in the immunological synapse directs reciprocal signaling in allogeneic Th:DC conjugates. J Immunol. 2007;179:819–829. doi: 10.4049/jimmunol.179.2.819. [DOI] [PubMed] [Google Scholar]
  • 44.Tacchini-Cottier F, Allenbach C, Otten LA, Radtke F. Notch 1 expression on T cells is not required for CD4+ T helper differentiation. Eur J Immunol. 2004;34:1588–1596. doi: 10.1002/eji.200324337. [DOI] [PubMed] [Google Scholar]
  • 45.Amsen D, et al. Direct regulation of Gata3 expression determines the T helper differentiation potential of Notch. Immunity. 2007;27:89–99. doi: 10.1016/j.immuni.2007.05.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Jurynczyk M, Jurewicz A, Raine CS, Selmaj K. Notch 3 inhibition in myelin-reactive T cells down-regulates protein kinase C theta and attenuates experimental autoimmune encephalomyelitis. J Immunol. 2008;180:2634–2640. doi: 10.4049/jimmunol.180.4.2634. [DOI] [PubMed] [Google Scholar]
  • 47.Hao L, et al. Notch-1 activates estrogen receptor-alpha-dependent transcription via IKKalpha in breast cancer cells. Oncogene. 2010;29:201–213. doi: 10.1038/onc.2009.323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Song LL, et al. Notch-1 associates with IKKalpha and regulates IKK activity in cervical cancer cells. Oncogene. 2008;27:5833–5844. doi: 10.1038/onc.2008.190. [DOI] [PubMed] [Google Scholar]
  • 49.Shin HM, et al. Notch 1 augments NF-kappaB activity by facilitating its nuclear retention. EMBO J. 2006;25:129–138. doi: 10.1038/sj.emboj.7600902. Along with (47,48,49), Notch promotes Th1 by NFkB pathway. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Tanigaki K, et al. Regulation of alphabeta/gammadelta T cell lineage commitment and peripheral T cell responses by Notch/RBP-J signaling. Immunity. 2004;20:611–622. doi: 10.1016/s1074-7613(04)00109-8. RBP/J(CSL) is important for Notch reguration of T cell response. [DOI] [PubMed] [Google Scholar]
  • 51.Tu L, et al. Notch signaling is an important regulator of type 2 immunity. J Exp Med. 2005;202:1037–1042. doi: 10.1084/jem.20050923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Fang TC, et al. Notch directly regulates Gata3 expression during T helper 2 cell differentiation. Immunity. 2007;27:100–110. doi: 10.1016/j.immuni.2007.04.018. Along with (45,52), Notch regulates Th2 through GATA3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kijima M, et al. Jagged1 suppresses collagen-induced arthritis by indirectly providing a negative signal in CD8+ T cells. J Immunol. 2009;182:3566–3572. doi: 10.4049/jimmunol.0803765. [DOI] [PubMed] [Google Scholar]
  • 54.Cho OH, et al. Notch regulates cytolytic effector function in CD8+ T cells. J Immunol. 2009;182:3380–3389. doi: 10.4049/jimmunol.0802598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–252. doi: 10.1038/32588. [DOI] [PubMed] [Google Scholar]
  • 56.Benedito R, et al. The notch ligands Dll4 and Jagged1 have opposing effects on angiogenesis. Cell. 2009;137:1124–1135. doi: 10.1016/j.cell.2009.03.025. [DOI] [PubMed] [Google Scholar]
  • 57.Kassner N, et al. Cutting edge: Plasmacytoid dendritic cells induce IL-10 production in T cells via the Delta-like-4/Notch axis. J Immunol. 2010;184:550–554. doi: 10.4049/jimmunol.0903152. [DOI] [PubMed] [Google Scholar]
  • 58.Sekine C, et al. Differential regulation of splenic CD8− dendritic cells and marginal zone B cells by Notch ligands. Int Immunol. 2009;21:295–301. doi: 10.1093/intimm/dxn148. [DOI] [PubMed] [Google Scholar]
  • 59.Moriyama Y, et al. Delta-like 1 is essential for the maintenance of marginal zone B cells in normal mice but not in autoimmune mice. Int Immunol. 2008;20:763–773. doi: 10.1093/intimm/dxn034. Along with (57,58,59), Notch ligand expression under steady condition. [DOI] [PubMed] [Google Scholar]
  • 60.Kuroda K, et al. Regulation of marginal zone B cell development by MINT, a suppressor of Notch/RBP-J signaling pathway. Immunity. 2003;18:301–312. doi: 10.1016/s1074-7613(03)00029-3. [DOI] [PubMed] [Google Scholar]
  • 61.Yamaguchi E, et al. Expression of Notch ligands, Jagged1, 2 and Delta1 in antigen presenting cells in mice. Immunol Lett. 2002;81:59–64. doi: 10.1016/s0165-2478(01)00326-1. [DOI] [PubMed] [Google Scholar]
  • 62.Tan AM, et al. TLR4 signaling in stromal cells is critical for the initiation of allergic Th2 responses to inhaled antigen. J Immunol. 2010;184:3535–3544. doi: 10.4049/jimmunol.0900340. [DOI] [PubMed] [Google Scholar]
  • 63.Napolitani G, Rinaldi A, Bertoni F, Sallusto F, Lanzavecchia A. Selected Toll-like receptor agonist combinations synergistically trigger a T helper type 1-polarizing program in dendritic cells. Nature immunology. 2005;6:769–776. doi: 10.1038/ni1223. Along with (62,63), TLR induction of Notch ligands in APC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Henley DB, May PC, Dean RA, Siemers ER. Development of semagacestat (LY450139), a functional gamma-secretase inhibitor, for the treatment of Alzheimer’s disease. Expert Opin Pharmacother. 2009;10:1657–1664. doi: 10.1517/14656560903044982. [DOI] [PubMed] [Google Scholar]
  • 65.Teachey DT, et al. Targeting Notch signaling in autoimmune and lymphoproliferative disease. Blood. 2008;111:705–714. doi: 10.1182/blood-2007-05-087353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Shih Ie M, Wang TL. Notch signaling, gamma-secretase inhibitors, and cancer therapy. Cancer Res. 2007;67:1879–1882. doi: 10.1158/0008-5472.CAN-06-3958. [DOI] [PubMed] [Google Scholar]
  • 67.Wei P, et al. Evaluation of selective gamma-secretase inhibitor PF-03084014 for its antitumor efficacy and gastrointestinal safety to guide optimal clinical trial design. Mol Cancer Ther. 2010;9:1618–1628. doi: 10.1158/1535-7163.MCT-10-0034. [DOI] [PubMed] [Google Scholar]
  • 68.Dahan S, et al. Notch-1 signaling regulates intestinal epithelial barrier function, through interaction with CD4+ T cells, in mice and humans. Gastroenterology. 2011;140:550–559. doi: 10.1053/j.gastro.2010.10.057. Along with (65,66,67,68), GSI treatment in Notch related inflamatry disease. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wu Y, et al. Therapeutic antibody targeting of individual Notch receptors. Nature. 2010;464:1052–1057. doi: 10.1038/nature08878. The first report about the establishment of new method to regulate Notch. [DOI] [PubMed] [Google Scholar]
  • 70.Abe N, Hozumi K, Hirano K, Yagita H, Habu S. Notch ligands transduce different magnitudes of signaling critical for determination of T-cell fate. Eur J Immunol. 2010;40:2608–2617. doi: 10.1002/eji.200940006. [DOI] [PubMed] [Google Scholar]
  • 71.Zamvil SS, Steinman L. The T lymphocyte in experimental allergic encephalomyelitis. Annual review of immunology. 1990;8:579–621. doi: 10.1146/annurev.iy.08.040190.003051. [DOI] [PubMed] [Google Scholar]
  • 72.Begolka WS, Vanderlugt CL, Rahbe SM, Miller SD. Differential expression of inflammatory cytokines parallels progression of central nervous system pathology in two clinically distinct models of multiple sclerosis. J Immunol. 1998;161:4437–4446. [PubMed] [Google Scholar]
  • 73.Sospedra M, Martin R. Immunology of multiple sclerosis. Annual review of immunology. 2005;23:683–747. doi: 10.1146/annurev.immunol.23.021704.115707. [DOI] [PubMed] [Google Scholar]
  • 74.Kebir H, et al. Human TH17 lymphocytes promote blood-brain barrier disruption and central nervous system inflammation. Nat Med. 2007;13:1173–1175. doi: 10.1038/nm1651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Bailey SL, Schreiner B, McMahon EJ, Miller SD. CNS myeloid DCs presenting endogenous myelin peptides ‘preferentially’ polarize CD4+ T(H)-17 cells in relapsing EAE. Nature immunology. 2007;8:172–180. doi: 10.1038/ni1430. [DOI] [PubMed] [Google Scholar]
  • 76.Segal BM. Th17 cells in autoimmune demyelinating disease. Semin Immunopathol. 2010;32:71–77. doi: 10.1007/s00281-009-0186-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Jurynczyk M, Selmaj K. Notch: a new player in MS mechanisms. J Neuroimmunol. 2010;218:3–11. doi: 10.1016/j.jneuroim.2009.08.010. [DOI] [PubMed] [Google Scholar]
  • 78.Martin PJ, et al. Evaluation of mycophenolate mofetil for initial treatment of chronic graft-versus-host disease. Blood. 2009;113:5074–5082. doi: 10.1182/blood-2009-02-202937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Ferrara JL, Levine JE, Reddy P, Holler E. Graft-versus-host disease. Lancet. 2009;373:1550–1561. doi: 10.1016/S0140-6736(09)60237-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Das R, Chen X, Komorowski R, Hessner MJ, Drobyski WR. Interleukin-23 secretion by donor antigen-presenting cells is critical for organ-specific pathology in graft-versus-host disease. Blood. 2009;113:2352–2362. doi: 10.1182/blood-2008-08-175448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Cutler C, Antin JH. Novel drugs for the prevention and treatment of acute GVHD. Curr Pharm Des. 2008;14:1962–1973. doi: 10.2174/138161208785061436. [DOI] [PubMed] [Google Scholar]
  • 82.Chao NJ. Are there effective new strategies for the treatment of acute and chronic GvHD? Best Pract Res Clin Haematol. 2008;21:93–98. doi: 10.1016/j.beha.2007.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Shlomchik WD. Graft-versus-host disease. Nat Rev Immunol. 2007;7:340–352. doi: 10.1038/nri2000. [DOI] [PubMed] [Google Scholar]
  • 84.Deeg HJ. How I treat refractory acute GVHD. Blood. 2007;109:4119–4126. doi: 10.1182/blood-2006-12-041889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Blazar BR, Murphy WJ. Bone marrow transplantation and approaches to avoid graft-versus-host disease (GVHD) Philos Trans R Soc Lond B Biol Sci. 2005;360:1747–1767. doi: 10.1098/rstb.2005.1701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.June CH. Adoptive T cell therapy for cancer in the clinic. J Clin Invest. 2007;117:1466–1476. doi: 10.1172/JCI32446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.June CH. Principles of adoptive T cell cancer therapy. J Clin Invest. 2007;117:1204–1212. doi: 10.1172/JCI31446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Caton ML, Smith-Raska MR, Reizis B. Notch-RBP-J signaling controls the homeostasis of CD8− dendritic cells in the spleen. J Exp Med. 2007;204:1653–1664. doi: 10.1084/jem.20062648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Zhou J, Cheng P, Youn JI, Cotter MJ, Gabrilovich DI. Notch and wingless signaling cooperate in regulation of dendritic cell differentiation. Immunity. 2009;30:845–859. doi: 10.1016/j.immuni.2009.03.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ishifune C, et al. Notch signaling regulates the development of a novel type of Thy1-expressing dendritic cell in the thymus. Eur J Immunol. 2011;41:1309–1320. doi: 10.1002/eji.201041159. Along with (88,89,90), Notch signaling in APC differentiation. [DOI] [PubMed] [Google Scholar]

RESOURCES