4,536 Matching Annotations
  1. Apr 2021
    1. Dephosphorylation of PGK1 by PTEN was found to inhibit its activity, downstream glycolytic functions, and glioblastoma cell proliferation xref , thereby presenting another mechanism in which PTEN functions as a tumour suppressor.

      PTEN dephosphorylates PGK1.

    2. It was reported that PTEN could dephosphorylate PGK1, a glycolytic enzyme and protein kinase with a tumorigenic role in glioblastoma 99.

      PTEN dephosphorylates PGK1.

    3. Dephosphorylation of PGK1 by PTEN was found to inhibit its activity, downstream glycolytic functions, and glioblastoma cell proliferation 99, thereby presenting another mechanism in which PTEN functions as a tumour suppressor.

      PTEN dephosphorylates PGK1.

    4. Newer studies add to this small body of data, including an intriguing study where a novel PTEN/ARID4B/PI3K pathway in which PTEN inhibits the expression of ARID4B was characterised.

      PTEN decreases the amount of ARID4B.

    5. PTEN inhibits ARID4B expression and thus prevents the transcriptional activation of ARID4B transcriptional targets PIK3CA and PIK3R2 (PI3K subunits) 79.

      PTEN decreases the amount of ARID4B.

    6. Furthermore, nuclear PTEN directly interacted with and inhibited RNA polymerase II (RNAPII)-mediated transcription, where it was involved in direct downregulation of critical transcriptional control genes including AFF4 and POL2RA 80.

      RNApo_II binds PTEN.

    7. Colocalisation of PTEN and PTENalpha promoted the function of PINK1, a mitochondrial-target kinase, and subsequently promoted energy production 105.

      PTEN activates PINK1.

    8. It is known that AKT signaling plays a critical role in the regulation of pre-mRNA splicing 77 and PTEN has been shown to modulate G6PD pre-mRNA splicing in an AKT independent manner 78.

      PTEN activates AKT.

    9. Numb inhibits Notch1, leading to the downregulation of RBP-Jkappa 94, which upregulates PTEN and anti-EMT effectors, leading to the downregulation of p-FAK and pro EMT effectors 94.

      NOTCH1 activates PTEN.

    1. Thus, while p53 deletion and missense mutations can enhance mTOR, emphasizing the functional interplay between AMPK and wild-type p53, some mutants can display effects on the canonical AMPK-mTOR signaling beyond the transcriptional repression.

      TP53 inhibits MTOR.

    2. By promoting glucose uptake, mutant p53 can limit autophagy dependent energy production.

      Mutated TP53 inhibits glucose.

    3. Thus, while mutant p53 enhanced glucose metabolism can correspondingly suppressed autophagy in proliferating cancer cells, it is reasonable that a reduced glycolysis by mutant p53 can induce autophagy in quiescent cells.

      Mutated TP53 inhibits glucose.

    4. This is in fact in line with the observations that CHIP, beyond targeting wild-type p53 by K48 polyubiquitinition, preferentially degrades aggregation prone mutant p53 proteins through K63 polyubiquitinition chains.

      STUB1 inhibits mutated TP53.

    5. Although p21 expression generally contributes to the induction of an irreversible proliferative arrest, transient p53 mediated induction of p21 is reversible, allowing cells to re-enter the cell cycle once stress or damage has been resolved.

      TP53 activates CDKN1A.

    6. A comparable example of this possibility is the activation of the cyclin-dependent kinase inhibitor p21 by p53.

      TP53 activates CDKN1A.

    7. Therefore, it is reasonable that some mutant p53 forms may enhance autophagy required to prevent energy crisis and maintain nucleotide pools during starvation in cancer cells caused by hypoxia and nutrition depletion in tumor microenvironment.

      Mutated TP53 activates autophagy.

    8. Alternatively, the mutant p53 driven autophagy suppressive function might be overridden by additional signaling, mutations or epigenetic changes.

      Mutated TP53 activates autophagy.

    9. Secondly, while mutant p53 have been linked to promote glycolysis through distinct mechanisms, emerging data supports the notion that not all mutants display enhanced glycolysis.

      Mutated TP53 activates glycolytic process.

    10. Therefore, sophisticated animal studies are needed for tumors that have undergone mutant p53 induced EMT program to provide an in vivo correlate in preclinical models.
    1. Down-regulated of RAC1 expression or loss of its function significantly suppressed cancer cell proliferation and metastasis.
    2. Furthermore, IR induced RAC1 expression and activity via the activation of PI3K/AKT signaling pathway, and then enhancing cell proliferation, survival, migration and metastasis and increasing levels of epithelial-to-mesenchymal transition (EMT) markers, which facilitated the cell survival and invasive phenotypes.

      Radiation, Ionizing increases the amount of RAC1.

    3. Additionally, IR could enhance the expression and activation of RAC1, positively associated with the up-regulation of PAK1, p-PAK1, LIMK1, p-LIMK1, Cofilin and p-Cofilin.

      Radiation, Ionizing increases the amount of RAC1.

    4. In this report, we found that IR could induce the up-regulation of RAC1 expression and activity via activating the PI3K and AKT signaling pathway.

      Radiation, Ionizing increases the amount of RAC1.

    5. IR significantly promoted the expression of GST-RAC1, RAC1, PAK1, p-PAK1, LIMK1, p-LIMK1, Cofilin, and p-Cofilin in the cells treated with IR.

      Radiation, Ionizing increases the amount of RAC1.

    6. These results indicate that IR increases the expression and activity of Rac1 via activating the PI3K and AKT signaling pathway.

      Radiation, Ionizing increases the amount of RAC1.

    7. A question is how IR induces Rac1 expression.

      Radiation, Ionizing increases the amount of RAC1.

    8. In addition, as shown in XREF_FIG, the results of GST-pull down assays showed Rac1 expression and activity was significantly increased after 6 Gy dose of IR in lung cancer cells, suggesting that IR could promote the Rac1 expression and activity.

      Radiation, Ionizing increases the amount of RAC1.

    9. IR Induces RAC1 Expression and EMT in Lung Cancer Cells.

      Radiation, Ionizing increases the amount of RAC1.

    10. As exhibited in xref , RAC1 overexpression led to the up-regulation of GST-RAC1, RAC1, PAK1, p-PAK1, LIMK1, p-LIMK1, Cofilin, and p-Cofilin in A549 and PC9 cells, while the opposite pattern of these genes was found in the A549 and PC9 cells after Rac1 knockdown.

      GST binds RAC1.

    11. E.g., RAC1 is activated by IR and the inhibition of RAC1 abrogates G2 checkpoint activation and cell survival following IR in breast cancer cells ( xref , xref ).
    12. Consistent with the in vitro results, RAC1 significantly enhanced tumor xenograft growth treated with IR (XREF_FIG).
    13. Furthermore , IR induced RAC1 expression and activity via the activation of PI3K / AKT signaling pathway , and then enhancing cell proliferation , survival , migration and metastasis and increasing levels of epithelial-to-mesenchymal transition ( EMT ) markers , which facilitated the cell survival and invasive phenotypes .
    14. The western blot results showed that IR could significantly increase the PI3K, p-AKT, AKT, and RAC1, whereas the LY294002 reversed this effect in both A549 and PC9 cells (XREF_FIG).
    15. Furthermore, IR induced RAC1 expression and activity via the activation of PI3K and AKT signaling pathway, and then enhancing cell proliferation, survival, migration and metastasis and increasing levels of epithelial-to-mesenchymal transition (EMT) markers, which facilitated the cell survival and invasive phenotypes.
    16. In this study, we also found that overexpression of Rac1 significantly promoted the migration and invasion, and radioresistance of lung cancer cells, whereas the knockdown of Rac1 markedly inhibited these capabilities of lung cancer cells in vivo and in vitro.
    17. In this article, we uncovered inhibition of RAC1 in lung cancer cells is sufficient to abrogate the IR induced and RAC1 mediated tumor migration and invasion, as evidenced by cell proliferation, colony formation, and Transwell assay.
    18. RAC1 Promotes Radioresistance, Invasion and Migration in Lung Cancer Cells.
    19. As exhibited in XREF_FIG, RAC1 overexpression led to the up-regulation of GST-RAC1, RAC1, PAK1, p-PAK1, LIMK1, p-LIMK1, Cofilin, and p-Cofilin in A549 and PC9 cells, while the opposite pattern of these genes was found in the A549 and PC9 cells after Rac1 knockdown.

      RAC1 activates RAC1.

    20. As exhibited in XREF_FIG, RAC1 overexpression led to the up-regulation of GST-RAC1, RAC1, PAK1, p-PAK1, LIMK1, p-LIMK1, Cofilin, and p-Cofilin in A549 and PC9 cells, while the opposite pattern of these genes was found in the A549 and PC9 cells after Rac1 knockdown.

      RAC1 activates PAK1.

    21. The colony formation assays showed that ectopic overexpression of RAC1 promoted growth of both A549 and PC9 cells compared to empty vector control (XREF_FIG).

      RAC1 activates PCSK9.

    22. The results demonstrated that RAC1 increased survival capacity of A549 and PC9 cells after IR at 0, 2, 4, 6, and 8 Gy (XREF_FIG).

      RAC1 activates PCSK9.

    23. These results suggest that RAC1 promotes proliferation of lung cancer cells.
    24. Furthermore, CCK-8 assays demonstrated that overexpression of RAC1 promoted cell proliferation, but silencing of RAC1 expression inhibited cell proliferation in both A549 and PC9 cells (XREF_FIG).
    25. Furthermore, IR induced RAC1 expression and activity via the activation of PI3K and AKT signaling pathway, and then enhancing cell proliferation, survival, migration and metastasis and increasing levels of epithelial-to-mesenchymal transition (EMT) markers, which facilitated the cell survival and invasive phenotypes.
    26. Collectively, we further found that RAC1 enhanced radioresistance by promoting EMT via targeting the PAK1-LIMK1-Cofilins signaling in lung cancer.
    27. Silencing Rac1 significantly inhibited the EMT phenotype in lung cancer cells, accompanied with a significant down-regulation of RAC1, PAK1, p-PAK1, LIMK1, p-LIMK1, Cofilin, and p-Cofilin in lung cancer cells.
    28. Our previous study found that RAC1 could significantly induce the EMT of colon cancer cells, which may be related to the positive regulation of Rac1/PAK1/LIMK1/Cofilins signaling pathway.
    1. Interestingly, CRP-induced MMP9 expression and invasion on RA-FLSs were p38-dependent as addition of a p38 inhibitor (SB202190) but not a NF-κB inhibitor (PDTC) was capable of inhibiting CRP-induced MMP9 expression and cell invasion.

      CRP increases the amount of MMP9.

    2. In contrast, blockade of CD16 produced no inhibitory effect on CRP-induced expression of CXCL8, CCL2, MMP9 and IL-6 by RA-FLSs ( xref ), suggesting that CRP may not signal through the CD16 to induce joint inflammation in vitro .

      CRP increases the amount of MMP9.

    3. However, blocking CD32 but not CD64 to inhibit CRP induced FLS proliferation, invasiveness, and proinflammatory cytokine CXCL8 production revealed a major role for CD32 signaling in synovial inflammation, although CRP via CD64, not CD32, to induce MMP9 expression was noticed.

      CRP increases the amount of MMP9.

    4. In contrast, blockade of CD16 produced no inhibitory effect on CRP-induced expression of CXCL8, CCL2, MMP9 and IL-6 by RA-FLSs ( xref ), suggesting that CRP may not signal through the CD16 to induce joint inflammation in vitro .

      CRP increases the amount of CXCL8.

    5. However, CRP- induced expression of CXCL8 was CD32-dependent as it was blunted by the antibody against CD32, whereas CRP-induced MMP9 was blocked by the antibody to CD64, demonstrating that differential signaling mechanisms for CRP in regulating CXCL8 and MMP9 expression in RA-FLSs.

      CRP increases the amount of CXCL8.

    6. In contrast, blockade of CD16 produced no inhibitory effect on CRP-induced expression of CXCL8, CCL2, MMP9 and IL-6 by RA-FLSs ( xref ), suggesting that CRP may not signal through the CD16 to induce joint inflammation in vitro .

      CRP increases the amount of IL6.

    7. As shown in xref , CRP-induced expression of CCL2 and IL-6 was blocked by either neutralizing antibody to CD32 or CD64 or both, suggesting that CRP signals through both CD32/CD64 to induce expression of CCL2 and IL-6.

      CRP increases the amount of IL6.

    8. As shown in XREF_FIG, CRP induced expression of CCL2 and IL-6 was blocked by either neutralizing antibody to CD32 or CD64 or both, suggesting that CRP signals through both CD32 and CD64 to induce expression of CCL2 and IL-6.

      CRP increases the amount of IL6.

    9. In contrast, blockade of CD16 produced no inhibitory effect on CRP-induced expression of CXCL8, CCL2, MMP9 and IL-6 by RA-FLSs ( xref ), suggesting that CRP may not signal through the CD16 to induce joint inflammation in vitro .

      CRP increases the amount of CCL2.

    10. As shown in xref , CRP-induced expression of CCL2 and IL-6 was blocked by either neutralizing antibody to CD32 or CD64 or both, suggesting that CRP signals through both CD32/CD64 to induce expression of CCL2 and IL-6.

      CRP increases the amount of CCL2.

    11. As shown in XREF_FIG, CRP induced expression of CCL2 and IL-6 was blocked by either neutralizing antibody to CD32 or CD64 or both, suggesting that CRP signals through both CD32 and CD64 to induce expression of CCL2 and IL-6.

      CRP increases the amount of CCL2.

    12. It has been reported that CRP-induced cytokine expression is also regulated by TWIST transcriptionally in myeloma cells ( xref ).

      CRP increases the amount of cytokine.

    13. We thus blocked CD32 or CD64 or both with neutralizing antibodies to differentially determine the signaling mechanisms of CRP-induced cytokine expression.

      CRP increases the amount of cytokine.

    14. Thus, it is highly possible that high concentration of CRP in synovial fluid in patients with RA may directly bind primarily CD32 to activate p38 MAP kinase and NF-kappaB signaling in RA-FLSs to differentially regulate synovial inflammation.

      CD32 binds CRP.

    15. In vitro studies confirmed this notion and found that CRP was able to upregulate both CD32 and CD64 and induced FLS proliferation, invasion, and pro inflammatory expression by increasing production of CCL2, CXCL8, IL-6, MMP2, MMP9 while suppressing an anti-inflammatory cytokine IL-10 expression.
    16. RA-FLS was a major cell type responsible for CRP production in RA patients, accounting for more than 65% of CRP producing cells as identified by co-expressing CRP and vimentin in the inflamed synovial tissues in patients with RA.
    17. As shown in XREF_FIG, multiplex cytokine assay kits assays showed that addition of CRP dose-dependently upregulated CCL2, CXCL8, IL-6, MMP2, MMP9 in RA-FLS but not in HFLS, although expression of IL-1beta and TNFalpha was not significantly changed (XREF_FIG).

      CRP activates MMP9.

    18. In vitro studies confirmed this notion and found that CRP was able to upregulate both CD32 and CD64 and induced FLS proliferation, invasion, and pro inflammatory expression by increasing production of CCL2, CXCL8, IL-6, MMP2, MMP9 while suppressing an anti-inflammatory cytokine IL-10 expression.

      CRP activates MMP9.

    19. In vitro studies confirmed this notion and found that CRP was able to upregulate both CD32 and CD64 and induced FLS proliferation, invasion, and pro inflammatory expression by increasing production of CCL2, CXCL8, IL-6, MMP2, MMP9 while suppressing an anti-inflammatory cytokine IL-10 expression.

      CRP activates MMP2.

    20. As shown in XREF_FIG, multiplex cytokine assay kits assays showed that addition of CRP dose-dependently upregulated CCL2, CXCL8, IL-6, MMP2, MMP9 in RA-FLS but not in HFLS, although expression of IL-1beta and TNFalpha was not significantly changed (XREF_FIG).

      CRP activates MMP2.

    21. In vitro studies confirmed this notion and found that CRP was able to upregulate both CD32 and CD64 and induced FLS proliferation, invasion, and pro inflammatory expression by increasing production of CCL2, CXCL8, IL-6, MMP2, MMP9 while suppressing an anti-inflammatory cytokine IL-10 expression.

      CRP activates CXCL8.

    22. As shown in XREF_FIG, multiplex cytokine assay kits assays showed that addition of CRP dose-dependently upregulated CCL2, CXCL8, IL-6, MMP2, MMP9 in RA-FLS but not in HFLS, although expression of IL-1beta and TNFalpha was not significantly changed (XREF_FIG).

      CRP activates CXCL8.

    23. In vitro studies confirmed this notion and found that CRP was able to upregulate both CD32 and CD64 and induced FLS proliferation, invasion, and pro inflammatory expression by increasing production of CCL2, CXCL8, IL-6, MMP2, MMP9 while suppressing an anti-inflammatory cytokine IL-10 expression.

      CRP activates IL6.

    24. As shown in XREF_FIG, multiplex cytokine assay kits assays showed that addition of CRP dose-dependently upregulated CCL2, CXCL8, IL-6, MMP2, MMP9 in RA-FLS but not in HFLS, although expression of IL-1beta and TNFalpha was not significantly changed (XREF_FIG).

      CRP activates IL6.

    25. In vitro studies confirmed this notion and found that CRP was able to upregulate both CD32 and CD64 and induced FLS proliferation, invasion, and pro inflammatory expression by increasing production of CCL2, CXCL8, IL-6, MMP2, MMP9 while suppressing an anti-inflammatory cytokine IL-10 expression.

      CRP activates CCL2.

    26. As shown in XREF_FIG, multiplex cytokine assay kits assays showed that addition of CRP dose-dependently upregulated CCL2, CXCL8, IL-6, MMP2, MMP9 in RA-FLS but not in HFLS, although expression of IL-1beta and TNFalpha was not significantly changed (XREF_FIG).

      CRP activates CCL2.

    27. In vitro studies confirmed this notion and found that CRP was able to upregulate both CD32 and CD64 and induced FLS proliferation, invasion, and pro inflammatory expression by increasing production of CCL2, CXCL8, IL-6, MMP2, MMP9 while suppressing an anti-inflammatory cytokine IL-10 expression.
    28. This was further confirmed by the ability of pre-treating RA-FLS with a NF-kappaB inhibitor, PDTC (100 mumol/L) to inhibit CRP induced proliferation (XREF_FIG) and upregulation of CXCL8, CCL2.
    29. CRP can induce synovial inflammation via mechanisms associated with activation of CD32/64-p 38 and NF-kappaB signaling.
    30. Here we tested the hypothesis that CRP may be produced locally by FLSs and functions to induce the synovial inflammation in patients with RA.
    31. CRP may promote synovial inflammation via mechanism associated with activation of CD32/64- p38 and NF-kappaB signaling.
    32. In the present study, we found that CRP signaled primarily through CD32, to a less extent of CD64, to differentially regulate joint inflammation.
    33. CRP Promotes RA-FLS Pro inflammatory Response Differentially via the CD32/64-p 38 and NF-kappaB-Dependent Mechanisms in vitro.
    34. This was supported by the findings that CRP induced activation of p38 MAP kinase and NF-kappaB signaling was blunted by neutralizing antibodies against CD32 but not CD64.

      CRP activates p38.

    35. To examine whether CRP induces NF-kappaB nuclear translation, immunofluorescence and subcellular fractionation were performed.

      CRP activates NFkappaB.

    1. Also, the phosphorylation of c-Jun N-terminal kinase (JNK) and extracellular-signal-regulated kinase (ERK) were stimulated by S100A8, which had an analogous effect to the lipopolysaccharide (LPS) treatment ( xref C–E).

      S100A8 leads to the phosphorylation of JNK.

    2. S100A8 Induced Pro Inflammatory Cytokine Production Via Phosphorylation of ERK and JNK in BV-2 Cells.

      S100A8 leads to the phosphorylation of JNK.

    3. Also, the phosphorylation of c-Jun N-terminal kinase (JNK) and extracellular-signal-regulated kinase (ERK) were stimulated by S100A8, which had an analogous effect to the lipopolysaccharide (LPS) treatment (XREF_FIG C-E).

      S100A8 leads to the phosphorylation of JNK.

    4. Also, the phosphorylation of c-Jun N-terminal kinase (JNK) and extracellular-signal-regulated kinase (ERK) were stimulated by S100A8, which had an analogous effect to the lipopolysaccharide (LPS) treatment ( xref C–E).

      S100A8 leads to the phosphorylation of ERK.

    5. S100A8 Induced Pro Inflammatory Cytokine Production Via Phosphorylation of ERK and JNK in BV-2 Cells.

      S100A8 leads to the phosphorylation of ERK.

    6. Also, the phosphorylation of c-Jun N-terminal kinase (JNK) and extracellular-signal-regulated kinase (ERK) were stimulated by S100A8, which had an analogous effect to the lipopolysaccharide (LPS) treatment (XREF_FIG C-E).

      S100A8 leads to the phosphorylation of ERK.

    7. The S100A8 knockdown using shRNA revealed that COX-2 and PGE 2 expression was regulated by S100A8, which suggested that the intracellular increase of microglial S100A8 levels upregulated COX-2 expression and PGE2 secretion, contributing to neuronal death under hypoxic conditions.

      S100A8 increases the amount of PTGS2.

    8. Our study showed that hypoxia increased the production of S100A8 in microglia .

      Hypoxia activates S100A8.

    9. FACS analysis showed that the increase of S100A8 levels in microglia by hypoxia promoted neuronal apoptosis , which was confirmed by immunofluorescence .

      Hypoxia activates S100A8.

    10. In agreement with previous reports, the results of this study confirmed that S100A8 significantly increased the production of IL-6, TNF-alpha, and IL-1beta.

      S100A8 activates IL6.

    11. In our study , the increase of IL-1beta expression by S100A8 indicated that S100A8 was involved in the priming signal for NLRP3 inflammasome assembly in microglia ( Figure 2B ) .

      S100A8 activates IL1B.

    12. In agreement with previous reports, the results of this study confirmed that S100A8 significantly increased the production of IL-6, TNF-alpha, and IL-1beta.

      S100A8 activates IL1B.

    13. These results strongly suggested that S100A8 induced the NLRP3 inflammasome priming via NF-kappaB activation.

      S100A8 activates NLRP3.

    14. The results suggested that S100A8, secreted by neuronal cells under hypoxic conditions, triggered the priming of NLRP3 in microglial cells, through the TLR4 and NF-kappaB signaling.

      S100A8 activates NLRP3.

    15. In addition, the translocation of NF-kB, which played a pivotal role in regulating the expression and activation of NLRP3, was also increased when cells were treated with S100A8.

      S100A8 activates NLRP3.

    16. These results suggested that S100A8, secreted by neuronal cells under hypoxic conditions, combined with TLR4 of microglia cells, activated the NLRP3 inflammasome priming.

      S100A8 activates NLRP3.

    17. In agreement with previous reports, the results of this study confirmed that S100A8 significantly increased the production of IL-6, TNF-alpha, and IL-1beta.

      S100A8 activates TNF.

    18. However, for the first time, we showed that up-regulation of microglial S100A8 levels increased neuronal apoptosis after hypoxia, in primary multicellular cultures consisting of neurons, astrocytes, and microglia.
    19. Therefore, this study determined whether S100A8 induced neuronal apoptosis during cerebral hypoxia and elucidated its mechanism of action using in vitro systems, including astrocytes and microglial and neuronal cells, under hypoxic conditions.
    20. S100A8 Knockdown on Microglia Attenuated Neuronal Apoptosis by Hypoxia.
    21. To investigate whether S100A8 expression in microglia induced apoptosis of neuronal cells under hypoxic condition, SH-SY5Y cells were co-cultured with BV-2 cells transfected with S100A8 shRNA for 48 h in a 0.4 mum pore transwell system and under hypoxic conditions (XREF_FIG A, B).
    22. These findings indicated that the expression of S100A8, induced in microglia cells under hypoxic conditions, activated COX-2 expression and PGE 2 secretion to induce the apoptosis of neurons.
    23. FACS analysis showed that the increase of S100A8 levels in microglia by hypoxia promoted neuronal apoptosis, which was confirmed by immunofluorescence.
    1. Knockdown of S100A8 levels by using shRNA revealed that microglial S100A8 expression activated COX-2 expression, leading to neuronal apoptosis under hypoxia.

      S100A8 increases the amount of PTGS2.

    2. S100A8, secreted from neurons under hypoxia, activated the secretion of tumor necrosis factor (TNF-alpha) and interleukin-6 (IL-6) through phosphorylation of extracellular-signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) in microglia.

      S100A8 activates IL6.

    3. S100A8, secreted from neurons under hypoxia, activated the secretion of tumor necrosis factor (TNF-alpha) and interleukin-6 (IL-6) through phosphorylation of extracellular-signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) in microglia.

      S100A8 activates TNF.

    4. The aim of this study was to determine whether S100A8 induced neuronal apoptosis during cerebral hypoxia and elucidate its mechanism of action.
    1. The coherent molecular mechanisms underlying the NO related inhibition of IDO remain unknown.

      nitric oxide inhibits IDO1.

    2. The antioxidant properties of IDO were proven, so the inhibiting of IDO by NO may restrict the antioxidant properties and induce increased free radicals.

      nitric oxide inhibits IDO1.

    3. In vivo studies indicate that NO can inhibit IDO catalytic activity by directly interacting [XREF_BIBR] or by stimulating IDO degradation through the proteasome pathway [XREF_BIBR].

      nitric oxide inhibits IDO1.

    1. Cells expressing IDO can co-express iNOS in response to IFN-gamma, which produces NO that inhibits, in turn, IDO.

      nitric oxide inhibits IDO1.

    1. Previous studies have suggested nitric oxide (NO) can inhibit IDO activity and expression [XREF_BIBR, XREF_BIBR] most likely through post-translational regulation leading to proteasomal degradation of IDO rather than transcriptional regulation [XREF_BIBR].

      nitric oxide inhibits IDO1.

    2. Previous studies have suggested nitric oxide (NO) can inhibit IDO activity and expression [XREF_BIBR, XREF_BIBR] most likely through post-translational regulation leading to proteasomal degradation of IDO rather than transcriptional regulation [XREF_BIBR].

      nitric oxide inhibits IDO1.

    1. For example, nitric oxide (NO) production may block IDO enzyme activity since NO inhibits the activity of heme containing enzymes such as IDO (Thomas et al., 2001).

      nitric oxide inhibits IDO1.

    2. For example, nitric oxide (NO) production may block IDO enzyme activity since NO inhibits the activity of heme containing enzymes such as IDO (Thomas et al., 2001).

      nitric oxide inhibits IDO1.

    1. In sharp contrast to macrophages, murine microglial cell clones immortalized with the activated c-myc oncogene have been reported to be resistant to NO inhibition of IDO.

      nitric oxide inhibits IDO1.

    2. In conclusion, results of the present series of experiments indicate that IDO in primary murine microglia costimulated with IFNgamma + LPS is not impaired by the production of NO, as is known to occur in murine macrophages.

      nitric oxide inhibits IDO1.

    3. Conversely, induction of NO by activation of iNOS down-regulates IDO activity in cell types as diverse as human uroepithelial transformed cells, murine bone marrow derived myeloid dendritic cells, human transformed and primary macrophages and mouse peritoneal cells.

      nitric oxide inhibits IDO1.

    4. As a result of this crosstalk, NO is well known to inhibit IDO activity in many types of cells.

      nitric oxide inhibits IDO1.

    5. In addition, direct binding of NO to heme iron in IDO, which is one of the mechanisms by which NO inhibits IDO at the post-translational level, is dependent on a number of cellular factors, including NO abundance, pH, redox environment and tryptophan availability.

      nitric oxide inhibits IDO1.

    6. NO inhibition of IDO is apparently specific to both certain species and cell types.

      nitric oxide inhibits IDO1.

    7. In macrophages, the inhibition of IDO by NO occurs at both the transcriptional and post-transcriptional levels.

      nitric oxide inhibits IDO1.

    1. Previous studies have reported that NO is able to inhibit the activity of IDO by reacting with the heme iron situated in its active site.

      nitric oxide inhibits IDO1.

    2. NO inhibits IDO at the transcriptional level and accelerates the decomposition of IDO protein, thereby affecting its stability.

      nitric oxide inhibits IDO1.

    1. Therefore, simultaneous induction of nitric oxide, respiratory burst and tryptophan degradation responses would antagonize PKC and thus NADPH oxidase activation and the IDO enzyme.

      nitric oxide inhibits IDO1.

    1. Several findings have demonstrated that NO is able to inhibit the IDO enzyme by direct interaction or accelerating proteasomal degradation [XREF_BIBR, XREF_BIBR].

      nitric oxide inhibits IDO1.

    1. Although not yet examined in the context of exercise, nitric oxide has been shown to inhibit IDO activity.

      nitric oxide inhibits IDO1.

    2. 33 Thus, a strong rationale suggests that exercise induced changes in nitric oxide may mediate an inhibition of IDO activity, possibly leading to a chronic downregulation and stabilization of the KYN pathway as reported by Zimmer et al. 23 The mechanisms underlying both acute and chronic exercise induced elevations in the metabolic flux towards KA could be driven by KAT expression in different tissues or cell types.

      nitric oxide inhibits IDO1.

    1. XREF_BIBR Nitric oxide, interleukin-4, peroxynitrite and transforming growth factor beta inhibit IDO-1 but it is unclear whether these also inhibit IDO-2.

      nitric oxide inhibits IDO1.

    2. XREF_BIBR 3-HAA also inhibits nitric oxide synthetase (although not in microglial cells) and nuclear factor kappaB expression; XREF_BIBR, XREF_BIBR of which the former could result in positive feedback and upregulation of IDO activity, which is inhibited by nitric oxide, as well as neuronal dysfunction through impairment of nitric oxide 's neurotransmitter function.

      nitric oxide inhibits IDO1.

    1. Furthermore, CCL11 induced the mRNA expression of CCL11 and CCR3.

      CCL11 increases the amount of CCR3.

    2. Furthermore, CCL11 induced the mRNA expression of CCL11 and CCR3.

      CCL11 increases the amount of CCL11.

    1. The results of the present study demonstrated that FBXW7 efficiently inhibited SKOV3 cell invasion and migration, as well as tube formation of HUVECs.
    2. It has also been reported that FBXW7 suppresses oral squamous cell carcinoma proliferation and invasion regulated by miR-27a through the PI3K and AKT signaling pathway.
    3. The results of the present study demonstrated that overexpression of FBXW7 inhibited the invasion, migration, EMT and angiogenesis of OC cells.
    4. In conclusion, the results of the present study demonstrated that FBXW7 inhibited the invasion, migration and angiogenesis of OC cells.
    5. Previous studies have demonstrated that beta-catenin signaling helps VEGF regulate angiogenesis , and that FBXW7 promotes the degradation of beta-catenin ( 42,43 ) .

      FBXW7 inhibits CTNNB1.

    6. Previous studies have demonstrated that beta-catenin signaling helps VEGF regulate angiogenesis, and that FBXW7 promotes the degradation of beta-catenin.

      FBXW7 inhibits CTNNB1.

    7. It has also been reported that FBXW7 suppresses oral squamous cell carcinoma proliferation and invasion regulated by miR-27a through the PI3K and AKT signaling pathway.
    8. The results of the present study demonstrated that overexpression of FBXW7 inhibited the invasion, migration, EMT and angiogenesis of OC cells.
    9. The results of the present study demonstrated that overexpression of FBXW7 inhibited the invasion, migration, EMT and angiogenesis of OC cells.

      FBXW7 inhibits angiogenesis.

    10. In conclusion, the results of the present study demonstrated that FBXW7 inhibited the invasion, migration and angiogenesis of OC cells.

      FBXW7 inhibits angiogenesis.

    11. FBXW7 inhibits VEGF expression through inactivation of beta-catenin signaling To further elucidate the potential molecular mechanism by which FBXW7 mediates its antitumor effects in OC , western blot analysis was performed to detect the expression levels of key proteins in beta-catenin signaling .

      FBXW7 inhibits VEGF.

    12. Collectively , these results suggested that FBXW7 may inhibit VEGF expression through inactivation of beta-catenin signaling in SKOV3 cells .

      FBXW7 inhibits VEGF.

    13. Mechanistically , FBXW7 suppressed VEGF expression by inactivating beta-catenin signaling .

      FBXW7 inhibits VEGF.

    14. Overall , the current results suggested that FBXW7 may inhibit VEGF expression through inactivation of beta-catenin signaling in SKOV3 cells .

      FBXW7 inhibits VEGF.

    15. Notably, overexpression of FBXW7 significantly decreased VEGF mRNA and protein expression compared with the vector control group (XREF_FIG and XREF_FIG).

      FBXW7 inhibits VEGF.

    16. Collectively, these results indicated that overexpression of FBXW7 inhibited the invasion, migration and EMT process of OC cells by suppressing VEGF expression.

      FBXW7 increases the amount of VEGF.

    17. Overall, these results suggested that overexpression of FBXW7 suppressed the angiogenesis of OC cells by suppressing VEGF expression.

      FBXW7 increases the amount of VEGF.

    18. The present study aimed to investigate VEGF expression following overexpression of FBXW7 in OC cells.

      FBXW7 increases the amount of VEGF.

    19. FBXW7 inhibits invasion, migration and angiogenesis in ovarian cancer cells by suppressing VEGF expression through inactivation of beta-catenin signaling.

      FBXW7 increases the amount of VEGF.

    20. Subsequently, FBXW7 was overexpressed to determine VEGF expression in SKOV3 cells.

      FBXW7 increases the amount of VEGF.

    21. Overall, the results of the present study suggested that FBXW7 inhibited invasion, migration and angiogenesis of OC cells by suppressing VEGF expression through inactivation of beta-catenin signaling.

      FBXW7 increases the amount of VEGF.

    22. Furthermore, overexpression of FBXW7 markedly suppressed beta-catenin and c-Myc expression, whereas the decreased expression levels of VEGF, VEGFR1 and VEGFR2 following overexpression of FBXW7 were increased after treatment of SKOV3 cells with LiCl.

      FBXW7 decreases the amount of MYC.

    23. As presented in XREF_FIG, overexpression of FBXW7 significantly decreased the expression levels of beta-catenin and c-Myc compared with the empty vector group.

      FBXW7 decreases the amount of MYC.

    24. The results of the present study demonstrated that overexpression of FBXW7 inhibited the expression levels of VEGF, beta-catenin and c-Myc.

      FBXW7 decreases the amount of MYC.

    25. As presented in XREF_FIG, overexpression of FBXW7 significantly decreased the expression levels of beta-catenin and c-Myc compared with the empty vector group.

      FBXW7 decreases the amount of CTNNB1.

    26. The results of the present study demonstrated that overexpression of FBXW7 inhibited the expression levels of VEGF, beta-catenin and c-Myc.

      FBXW7 decreases the amount of CTNNB1.

    27. Furthermore, overexpression of FBXW7 markedly suppressed beta-catenin and c-Myc expression, whereas the decreased expression levels of VEGF, VEGFR1 and VEGFR2 following overexpression of FBXW7 were increased after treatment of SKOV3 cells with LiCl.

      FBXW7 decreases the amount of CTNNB1.

    28. FBXW7 inhibits VEGF expression through inactivation of beta-catenin signaling.

      FBXW7 decreases the amount of VEGF.

    29. Collectively, these results suggested that FBXW7 may inhibit VEGF expression through inactivation of beta-catenin signaling in SKOV3 cells.

      FBXW7 decreases the amount of VEGF.

    30. Mechanistically, FBXW7 suppressed VEGF expression by inactivating beta-catenin signaling.

      FBXW7 decreases the amount of VEGF.

    31. The results of the present study demonstrated that overexpression of FBXW7 suppressed VEGF expression, while overexpression of VEGF partially counteracted the inhibitory effects of FBXW7 overexpression on the invasion, migration, EMT and angiogenesis of OC cells.

      FBXW7 decreases the amount of VEGF.

    32. Overall, the current results suggested that FBXW7 may inhibit VEGF expression through inactivation of beta-catenin signaling in SKOV3 cells.

      FBXW7 decreases the amount of VEGF.

    33. The results of the present study demonstrated that overexpression of FBXW7 inhibited the expression levels of VEGF, beta-catenin and c-Myc.

      FBXW7 decreases the amount of VEGF.

    34. Overexpression of FBXW7 significantly downregulates VEGF expression in OC cells.

      FBXW7 decreases the amount of VEGF.

    35. Overall, these results suggested that overexpression of FBXW7 inhibited VEGF expression in OC cells.

      FBXW7 decreases the amount of VEGF.

    36. Overexpression of FBXW7 significantly decreased VEGF expression in SKOV3 cells.

      FBXW7 decreases the amount of VEGF.

    37. The results demonstrated that overexpression of FBXW7 downregulated the expression levels of CD31, VEGFR1 and VEGFR, whereas co-transfection with FBXW7 and VEGF plasmids significantly increased their expression levels compared with the Ov-FBXW7+ pc-NC group (XREF_FIG and XREF_FIG), which is consistent with the results of the tube formation assay.

      FBXW7 decreases the amount of VEGFR.

    38. For instance, FBXW7 can target salt inducible kinase 2 for degradation, leading to the disruption of target of rapamycin 2-AKT signaling to inhibit pancreatic cancer cell proliferation and cell cycle progression.

      FBXW7 activates SIK2.

    1. Limonin induces apoptosis of HL-60 cells by inhibiting NQO1 activity.

      limonin inhibits NQO1.

    2. At the same time , limonin down-regulated the expression of NQO1 , indicating that limonin may indirectly act on the apoptosis pathway by regulating the expression activity of antioxidant enzymes in vivo , thus exerting its inhibitory effect on tumor cells , which provides an idea for the molecular mechanism that natural products can indirectly exert their anticancer effect by regulating the activity of antioxidant enzymes .

      limonin inhibits NQO1.

    3. Limonin induces apoptosis of HL-60 cells by inhibiting NQO1 activity.

      limonin inhibits NQO1.

    4. At the same time , limonin down-regulated the expression of NQO1 , indicating that limonin may indirectly act on the apoptosis pathway by regulating the expression activity of antioxidant enzymes in vivo , thus exerting its inhibitory effect on tumor cells , which provides an idea for the molecular mechanism that natural products can indirectly exert their anticancer effect by regulating the activity of antioxidant enzymes .

      limonin inhibits NQO1.

    5. At the same time, limonin down-regulated the expression of NQO1, indicating that limonin may indirectly act on the apoptosis pathway by regulating the expression activity of antioxidant enzymes in vivo, thus exerting its inhibitory effect on tumor cells, which provides an idea for the molecular mechanism that natural products can indirectly exert their anticancer effect by regulating the activity of antioxidant enzymes.

      limonin decreases the amount of NQO1.

    6. At the same time, limonin down-regulated the expression of NQO1, indicating that limonin may indirectly act on the apoptosis pathway by regulating the expression activity of antioxidant enzymes in vivo, thus exerting its inhibitory effect on tumor cells, which provides an idea for the molecular mechanism that natural products can indirectly exert their anticancer effect by regulating the activity of antioxidant enzymes.

      limonin decreases the amount of NQO1.

    7. In order to explore the anticancer activity based on interaction between limonin and NQO1, Human promyelocytic leukemia cells (HL-60) were studied in vitro.

      limonin binds NQO1.

    8. In order to explore the anticancer activity based on interaction between limonin and NQO1, Human promyelocytic leukemia cells (HL-60) were studied in vitro.

      limonin binds NQO1.

    1. However, CAPE did not affect NLRP3 or IL-1beta transcription, but instead enhanced NLRP3 binding to ubiquitin molecules, promoting NLRP3 ubiquitination, and contributing to the anti-tumor effect in the AOM and DSS mouse model.

      IL1B leads to the ubiquitination of NLRP3.

    2. However, CAPE did not affect NLRP3 or IL-1beta transcription, but instead enhanced NLRP3 binding to ubiquitin molecules, promoting NLRP3 ubiquitination, and contributing to the anti-tumor effect in the AOM and DSS mouse model.

      IL1B leads to the ubiquitination of NLRP3.

    3. However, CAPE did not affect NLRP3 or IL-1beta transcription, but instead enhanced NLRP3 binding to ubiquitin molecules, promoting NLRP3 ubiquitination, and contributing to the anti-tumor effect in the AOM and DSS mouse model.

      NLRP3 leads to the ubiquitination of NLRP3.

    4. However, CAPE did not affect NLRP3 or IL-1beta transcription, but instead enhanced NLRP3 binding to ubiquitin molecules, promoting NLRP3 ubiquitination, and contributing to the anti-tumor effect in the AOM and DSS mouse model.

      NLRP3 leads to the ubiquitination of NLRP3.

    5. In this study, we provide evidence that CAPE facilitates NLRP3 ubiquitination by inhibiting ROS in THP-1 cells and inhibits enteritis and tumor burden by inhibiting NLRP3 in an AOM/DSS mouse model.

      phenethyl caffeate leads to the ubiquitination of NLRP3.

    6. CAPE Promotes NLRP3 Ubiquitination by Inhibiting ROS.

      phenethyl caffeate leads to the ubiquitination of NLRP3.

    7. These findings indicate that CAPE also enhances NLRP3 ubiquitination in vivo .

      phenethyl caffeate leads to the ubiquitination of NLRP3.

    8. CAPE Increases NLRP3 Ubiquitination in AOM/DSS Mouse Model.

      phenethyl caffeate leads to the ubiquitination of NLRP3.

    9. Thus, CAPE suppresses the interaction between NLRP3 and deubiquitinating enzymes, and enhances its interaction with a ubiquitin conjugating enzyme in vivo and in vitro, promoting NLRP3 ubiquitination.

      phenethyl caffeate leads to the ubiquitination of NLRP3.

    10. Moreover, CAPE enhanced the binding of NLRP3 to ubiquitin molecules, promoted NLRP3 ubiquitination (XREF_FIG), and significantly blocked the formation of NLRP3 inflammasome, which were again reversed by rotenone (XREF_FIG).

      phenethyl caffeate leads to the ubiquitination of NLRP3.

    11. CAPE Promotes NLRP3 Ubiquitination by Inhibiting ROS.

      phenethyl caffeate leads to the ubiquitination of NLRP3.

    12. Moreover, CAPE enhanced the binding of NLRP3 to ubiquitin molecules, promoted NLRP3 ubiquitination (XREF_FIG), and significantly blocked the formation of NLRP3 inflammasome, which were again reversed by rotenone (XREF_FIG).

      NLRP3 inhibits NLRP3.

    13. Moreover, CAPE significantly inhibited the formation of ASC dimers and reduced the abundance of NLRP3 inflammasome complexes in a dose dependent manner (XREF_FIG).
    14. We first investigated whether CAPE inhibits the activation of NLRP3 inflammasome induced by ATP and LPS in macrophages in vitro.
    15. CAPE Decreases NLRP3 Inflammasome Activation in BMDMs and THP-1 Cells.
    16. We found that CAPE decreased NLRP3 inflammasome activation in BMDMs and THP-1 cells and protected mice from colorectal cancer induced by AOM and DSS.
    17. In conclusion, CAC can be prevented by CAPE-induced NLRP3 inflammasome inhibition, highlighting CAPE as a potential candidate for reducing the risk of CAC in patients with inflammatory bowel disease.
    18. In conclusion, CAC can be prevented by CAPE induced NLRP3 inflammasome inhibition, highlighting CAPE as a potential candidate for reducing the risk of CAC in patients with inflammatory bowel disease.
    19. Overall, the results indicate that activated NLRP3 in AOM and DSS mouse model is suppressed by CAPE.
    20. To determine whether CAPE inhibits NLRP3 inflammasome in vivo, we assessed NLRP3 expression in the AOM and DSS mouse model by immunohistochemistry and western blotting.