4,536 Matching Annotations
  1. Apr 2021
    1. Merlin expression in Meso-17 and Meso-25 cells decreased FAK Tyr397 phosphorylation and consequently disrupted FAK-Src and PI3K interaction, providing a mechanism for the observed enhancement of invasion and spreading caused by merlin inactivation.

      Modified NF2 leads to the dephosphorylation of PTK2 on Y397.

    2. Accordingly, merlin was shown to reduce the levels of ErbB2 and ErbB3 receptor levels at the plasma membrane.

      NF2 decreases the amount of ERBB3.

    3. Accordingly, merlin was shown to reduce the levels of ErbB2 and ErbB3 receptor levels at the plasma membrane.

      NF2 decreases the amount of ERBB3.

    4. Accordingly, merlin was shown to reduce the levels of ErbB2 and ErbB3 receptor levels at the plasma membrane.

      NF2 decreases the amount of ERBB2.

    5. In sub-confluent primary Schwann cells, we found that merlin binds to paxillin and mediates merlin localization at the plasma membrane and association with beta1-integrin and ErbB2, modifying the organization of the actin cytoskeleton in a cell density dependent manner.

      NF2 binds PXN.

    6. In sub-confluent primary Schwann cells, we found that merlin binds to paxillin and mediates merlin localization at the plasma membrane and association with β1-integrin and ErbB2, modifying the organization of the actin cytoskeleton in a cell density-dependent manner ( xref ).

      NF2 binds PXN.

    7. Merlin-Amot interaction was required for merlin regulation of mitogenic MAPK signaling.

      AMOT binds NF2.

    8. Moreover, in cultured Schwann cells, merlin interaction with Amot was demonstrated by co-immunoprecipitation of the endogenous proteins.

      AMOT binds NF2.

    9. Moreover, in cultured Schwann cells, merlin interaction with Amot was demonstrated by co-immunoprecipitation of the endogenous proteins ( xref ).

      AMOT binds NF2.

    10. Moreover, co-immunoprecipitation experiments revealed that merlin interacts with YAP1, although the interaction is not direct.

      YAP1 binds NF2.

    11. Moreover, co-immunoprecipitation experiments revealed that merlin interacts with YAP1, although the interaction is not direct ( xref ).

      YAP1 binds NF2.

    12. Studies in human meningioma tumors and in paired cell lines—KY21MG1 or MENII-1 meningioma cell lines and AC1 arachnoidal cells—demonstrated that merlin loss was associated with increased YAP expression and nuclear localization.

      YAP1 binds NF2.

    13. Amot-p130 isoform bound to the WW domains of YAP and blocked LATS1 access to YAP.

      YAP1 binds WW.

    14. The activation of Rac1 through CD44 was identified via the interaction of CD44 with Tiam-1, a Rac1 guanine nucleotide exchange factor (GEF) that catalyzes the replacement of the tightly-bound GDP with GTP.

      TIAM1 binds CD44.

    15. Merlin inactivation of Src signaling was also shown in CNS glial cells, where merlin competitively inhibits Src binding to ErbB2 thereby preventing ErbB2-mediated Src phosphorylation and downstream mitogenic signaling.

      SRC binds ERBB2.

    16. Merlin inactivation of Src signaling was also shown in CNS glial cells, where merlin competitively inhibits Src binding to ErbB2 thereby preventing ErbB2 mediated Src phosphorylation and downstream mitogenic signaling.

      SRC binds ERBB2.

    17. Merlin interacts with tubulin and acetylated-tubulin and stabilizes the microtubules by attenuating tubulin turnover -- lowering the rates of microtubule polymerization and depolymerization.

      Tubulin binds NF2.

    18. Merlin interacts with tubulin and acetylated-tubulin and stabilizes the microtubules by attenuating tubulin turnover—lowering the rates of microtubule polymerization and depolymerization.

      Tubulin binds NF2.

    19. Merlin inhibits PI3K activity by binding phosphatidylinositol 3-kinase enhancer-L (PIKE-L), the GTPase that binds and activates PI3K.

      GTPase binds PI3K.

    20. HDAC inhibitors disrupt the PP1-HDAC interaction facilitating Akt dephosphorylation and decrease human meningioma and schwannoma cell proliferation and schwannoma growth in an allograft model and meningioma growth in an intracranial xenograft model ( xref , xref , xref ).

      AKT binds HDAC and PPP1.

    21. In various cell types, the binding of hyaluronan to CD44 stimulates Tiam1 dependent Rac1 signaling and cytoskeleton mediated tumor cell migration.
    22. Hyaluronan-CD44 interaction in astrocytes and an immortalized mouse mammary epithelial cell line, EpH4, leads to Rac1 signaling activation and actin cytoskeleton rearrangement ( xref , xref ).
    23. In various cell types, the binding of hyaluronan to CD44 stimulates Tiam1 dependent Rac1 signaling and cytoskeleton mediated tumor cell migration.

      RAC1 activates cell migration.

    24. Pharmacological or genetic inhibition of Rac1 in Nf2 -/- MEFs reduced the Wnt signaling activation to basal levels as assessed by reporter assay of transactivation of the nuclear beta-catenin-dependent T-cell factor 4 transcription factor.

      RAC1 activates Wnt.

    25. In sub-confluent primary Schwann cells, we found that merlin binds to paxillin and mediates merlin localization at the plasma membrane and association with beta1-integrin and ErbB2, modifying the organization of the actin cytoskeleton in a cell density dependent manner.

      PXN bound to NF2 activates NF2.

    26. In sub-confluent primary Schwann cells, we found that merlin binds to paxillin and mediates merlin localization at the plasma membrane and association with beta1-integrin and ErbB2, modifying the organization of the actin cytoskeleton in a cell density dependent manner.

      PXN bound to NF2 activates localization.

    27. FAK silencing decreased schwannoma cell proliferation and was associated with increased levels of total and nuclear p53.
    28. In a similar fashion, NF2 mutations increased the resistance to dihydrofolate reductase inhibitors methotrexalate and pyremethamine as well as the JNK inhibitor JNK-9L.
    29. Furthermore, it was shown that overactive PAK and LIMK pathway activity contributed to cell proliferation through cofilin phosphorylation and auroraA activation.
    30. Interestingly, it was shown that schwannoma cells release insulin like growth factor binding protein 1 which in beta1-integrin dependent manner activates Src and FAK signaling.

      IGFBP1 activates PTK2.

    31. Interestingly, it was shown that schwannoma cells release insulin like growth factor binding protein 1 which in beta1-integrin dependent manner activates Src and FAK signaling.

      IGFBP1 activates SRC.

    32. Moreover, neuregulin survival signaling through the ErbB2 and ErbB3 receptor activates PI3K in rat Schwann cells through the activation of Akt and inhibition of Bad, a pro apoptotic Blc-2 family protein.

      ERBB3 activates PI3K.

    33. ErbB2 activation in mouse Nf2 deficient spinal cord neural progenitor cells was shown to be caused by Rac mediated retention of the receptor at the plasma membrane.

      ERBB2 activates NF2.

    34. Silencing DCAF1 in Meso-33, merlin deficient mesothelioma cells reduced their proliferation by arresting the cell cycle in G1 phase.
    35. Significantly, silencing of DCAF1 in schwannoma cells isolated from NF2 patients also reduced their proliferation.
    36. Silencing DCAF1 in Meso-33, merlin deficient mesothelioma cells reduced their proliferation by arresting the cell cycle in G1 phase.

      DCAF1 activates cell cycle.

    37. Furthermore, Amot silencing attenuated Rac1 and Ras and MAPK signaling pathway.

      AMOT activates RAC1.

    38. Silencing of Amot in Nf2 -/- Schwann cells (SC4) selectively reduced cell proliferation because it did not change the proliferation rate of SC4 with merlin re-expression.
    39. Furthermore, Amot silencing attenuated Rac1 and Ras and MAPK signaling pathway.

      AMOT activates RAS.

    40. Furthermore, Amot silencing attenuated Rac1 and Ras and MAPK signaling pathway.

      AMOT activates MAPK.

    41. In various cell types, the binding of hyaluronan to CD44 stimulates Tiam1 dependent Rac1 signaling and cytoskeleton mediated tumor cell migration.

      CD44 bound to hyaluronic acid activates RAC1.

    42. In various cell types, the binding of hyaluronan to CD44 stimulates Tiam1 dependent Rac1 signaling and cytoskeleton mediated tumor cell migration.

      CD44 bound to hyaluronic acid activates TIAM1.

    43. First, protein kinase C potentiated phosphatase inhibitor (CPI-17), which is frequently overexpressed in mesothelioma tumors, inhibits merlin phosphatase MYPT1-PP1delta, providing one potential pathway by which merlin 's tumor suppressor function might be inactivated through maintenance of phosphorylation at Ser518.

      PPP1R14A activates NF2.

    44. First, protein kinase C potentiated phosphatase inhibitor (CPI-17), which is frequently overexpressed in mesothelioma tumors, inhibits merlin phosphatase MYPT1-PP1delta, providing one potential pathway by which merlin 's tumor suppressor function might be inactivated through maintenance of phosphorylation at Ser518.

      PPP1R14A activates Phosphatase.

    45. In various cell types, the binding of hyaluronan to CD44 stimulates Tiam1 dependent Rac1 signaling and cytoskeleton mediated tumor cell migration.

      TIAM1 activates cell migration.

    46. Finally, a recent meningioma study of 73 patients found a high incidence of TERT promoter activating mutations in meningiomas undergoing malignant transformation in both NF2-related and sporadic meningiomas, whereas no TERT mutations were found in benign tumors.

      TERT activates Mutation.

    47. We recently showed that PI3K inhibition in merlin deficient mouse Schwann cells selectively decreased their proliferation.
    48. In primary rat Schwann cells, CD44 was shown to constitutively associate with the heterodimer receptor tyrosine kinase ErbB2 and ErbB3 and CD44 enhanced neuregulin induced ErbB2 activating phosphorylation.

      NRG activates ERBB2.

    49. Moreover, neuregulin survival signaling through the ErbB2 and ErbB3 receptor activates PI3K in rat Schwann cells through the activation of Akt and inhibition of Bad, a pro apoptotic Blc-2 family protein.

      NRG activates PI3K.

    50. In the canonical hippo pathway, mammalian Ste20 like kinases (Mst1/2; hippo homolog) phosphorylate large tumor suppressor kinases (LATS 1/2), which in turn phosphorylate and inactivate YAP and TAZ, blocking their role as TEAD and MEAD transcription factor co-activators.

      LATS activates TAZ.

    51. SOS is a GEF that activates Ras by catalyzing the nucleotide exchange.

      GEF activates RAS.

  2. Mar 2021
    1. Further, NF-kB inhibition by overexpression of IkB also results in S15 phosphorylation of mutant p53 via GADD45α mediated JNK1 activation ( xref ).

      RPS15 phosphorylates TP53.

    2. Chronic S15 phosphorylation of mutant p53 has been found in tumors where DNA damage signaling is constitutively activated ( xref , xref ).

      RPS15 phosphorylates TP53.

    3. On the contrary , Nanog suppresses p53 activity while Gli activated by Nanog inhibits p53 by activating Mdm2 to promote pluripotency .

      MDM2 inhibits TP53.

    4. For example , p53 repress CD133 by directly binding to its promoter and recruiting HDAC1 ( Figure 2 ) .

      TP53 inhibits PROM1.

    5. p53 loss upregulates CD133 which subsequently promotes CSC marker expression and confers stemness .

      TP53 inhibits PROM1.

    6. With the advent of reprogramming era , it was further highlighted that p53 loss promote dedifferentiation and reprogramming under favorable conditions .
    7. Furthermore, p53 loss was found to trigger dedifferentiation of mature hepatocytes to pluripotent cells by the activation of SC marker Nestin, which remains suppressed in wild-type p53 bearing cells (XREF_FIG).
    8. With the advent of reprogramming era, it was further highlighted that p53 loss promote dedifferentiation and reprogramming under favorable conditions.
    9. TP53 maintains homeostasis between self-renewal and differentiation depending on the cellular and developmental state and prevents the dedifferentiation and reprogramming of somatic cells to stem cells.
    10. Loss or gain-of-function mutations in TP53 induce dedifferentiation and proliferation of SCs with damaged DNA leading to the generation of CSCs.
    11. Also , upon DNA damage , p53 primarily promotes differentiation by suppression of Nanog .
    12. Association of p53 inactivation and loss of differentiation characteristics has also been reported in AML and lung cancer (XREF_FIG).
    13. Mutant p53 mediated repression of p63 function can also modulate the expression of certain miRNAs involved in invasion and metastasis such as let-7i, miR-155, miR-205, miR-130b, and miR-27a (XREF_FIG).

      Mutated TP53 inhibits TP63.

    14. Mutant p53 can itself disrupt the balance between stem cell proliferation and differentiation as well as sequester p63 or p73 thereby hindering apoptosis, augmenting proliferation, and driving chemoresistance and metastasis typical of cancer stem cells.

      Mutated TP53 inhibits TP63.

    15. Hence, loss of NUMB in breast cancer cells leads to decreased p53 levels and increased activity of NOTCH receptor which confers increased chemoresistance.

      NUMB increases the amount of TP53.

    16. It may form a complex with mutant p53 and MDM2 to block their ubiquitination mediated degradation or may form a complex with mutant p53 to prevent aggregation of mutant p53 by inhibiting MDM2 and CHIP in multiple cancer cell lines ( xref , xref ).

      TP53 binds MDM2.

    17. A recent study by Capaci et al. showed that mutant p53 can interact with HIF1α to induce miR-30d expression which promotes tubulo-vesiculation of Golgi apparatus leading to enhanced vesicular trafficking and secretion ( xref ) ( xref ).

      TP53 binds HIF1A.

    18. Binding of mutant p53 to ETS2 can promote expression of Pla2g16 or nucleotide synthesis genes required for invasion depending upon the cancer type ( xref ) ( xref , xref ).

      TP53 binds ETS2.

    19. Furthermore, the binding of mutant p53 to EGR1 promotes MYO10 expression which drives breast cancer cell invasion ( xref ) ( xref ).

      TP53 binds EGR1.

    20. Further, mutant p53 can interact with PELP1 to promote resistance to platinum-based drugs in triple negative breast cancer ( xref ).

      TP53 binds PELP1.

    21. A further study reported that mutant p53 enhance the association of mutant p53 and PARP on the replicating DNA ( xref ) ( xref ).

      TP53 binds PARP1.

    22. GOF mutant p53 can bind to TopBP1 and attenuate ATR checkpoint response during replication stress ( xref ) ( xref ).

      TP53 binds TOPBP1.

    23. The compound, RETRA disrupts mutant p53-p73 complex restoring p73-dependent transcription and apoptosis ( xref ) ( xref ).

      TP53 binds TP73.

    24. Short Interfering Mutant p53 Peptides (SIMP) can interact with different mutant p53 proteins and release p73, while peptides aptamers (PA) can inhibit mutant p53 transcription ( xref ) ( xref ).

      TP53 binds TP73.

    25. Mutant p53 and p63 complex can increase RAB coupling protein (RCP)-mediated recycling of cell surface growth promoting receptors.

      Mutated TP53 binds TP63.

    26. Zhou et al. showed that mutant p53 binds to novel interacting partner AMPKα in glucose starvation conditions and inhibits its activation by other kinases leading to increased aerobic glycolysis, lipid production, and cell growth ( xref ) ( xref ).

      AMPK_alpha binds TP53.

    27. This eliminates mitochondria-associated p53 which would otherwise be activated by PINK1 to mediate suppression of Nanog ( xref ) ( xref ).

      PINK1 activates TP53.

    28. This eliminates mitochondria associated p53 which would otherwise be activated by PINK1 to mediate suppression of Nanog (XREF_FIG).

      PINK1 activates TP53.

    29. Further, the human p53 isoform Delta133p53beta lacking the transactivation domain was observed to promote CSC features in breast cancer cell lines by expression of Sox2, Oct3/4, and Nanog in a Delta133p53beta dependent manner.
    30. Acetylation of p53 at K373 by CBP and p300 leads to dissociation of HDM2 and TRIM24 and subsequent activation of p53 which in turn transcriptionally activates p21, miR-34a, and miR-145 (XREF_FIG).

      TP53 activates MDM2.

    31. Additionally , p53 upregulates miR-34a that represses Notch ( Figure 2 ) and anti-apoptotic Bcl2 thereby promoting differentiation and apoptosis ( 82 ) .

      TP53 activates MIR34A.

    32. Further , induction of miR-34a by p53 functionally targets the CSC marker CD44 , thereby inhibiting prostate cancer regeneration and metastasis ( Figure 2 ) ( 74 ) .

      TP53 activates MIR34A.

    33. Acetylation of p53 at K373 by CBP/p300 leads to dissociation of HDM2 and TRIM24 and subsequent activation of p53 which in turn transcriptionally activates p21, miR-34a, and miR-145 ( xref ).

      TP53 activates CDKN1A.

    34. Acetylation of p53 at K373 by CBP and p300 leads to dissociation of HDM2 and TRIM24 and subsequent activation of p53 which in turn transcriptionally activates p21, miR-34a, and miR-145 (XREF_FIG).

      TP53 activates CDKN1A.

    35. A recent study by Alam et al. reveals GOF mutant p53 upregulates EFNB2 and activates ephrin B2 reverse signaling to impart enhanced chemoresistance to colorectal cancer cells ( xref ) ( xref ).

      TP53 activates EFN.

    36. Inactivation of p53 disrupts this balance and promotes pluripotency and somatic cell reprogramming .

      TP53 activates isoxaflutole.

    37. Inactivation of p53 disrupts this balance and promotes pluripotency and somatic cell reprogramming .

      TP53 activates isoxaflutole.

    38. Various transcription factors such as NF-Y, SREBPs, ETS, and EGFR1 play crucial role in mutant p53 driven invasion and metastasis.

      Mutated TP53 activates Neoplasm Metastasis.

    39. Mutant p53 implicate various context and tissue dependent mechanisms to promote cancer cell invasion and metastasis.

      Mutated TP53 activates Neoplasm Metastasis.

    40. Various transcription factors such as NF-Y, SREBPs, ETS, and EGFR1 play crucial role in mutant p53 driven invasion and metastasis.

      Mutated TP53 activates Neoplasm Invasiveness.

    41. Mutant p53 implicate various context and tissue dependent mechanisms to promote cancer cell invasion and metastasis.

      Mutated TP53 activates Neoplasm Invasiveness.

    42. This suggests that acetylation at K320 and K373 can alter the structure of mutant p53 and restore wild type p53 functions.

      Mutated TP53 activates TP53.

    43. Mutant p53 can also induce YAP and TAZ nuclear localization by interacting with SREBP and activating the mevalonate pathway.

      Mutated TP53 activates localization.

    44. Mutant p53 can also promote proliferation by inducing the REG-gamma proteosome pathway in association with p300 (XREF_FIG).

      Mutated TP53 activates cell population proliferation.

    45. GOF mutant p53 can modify the tumor microenvironment and has been found to support chronic inflammation.

      Mutated TP53 activates inflammatory response.

    46. While wild type p53 suppresses inflammatory response by inhibiting the production of cytokines and antagonizing NF-kB activity, mutant p53 on the other hand enhances NF-kB activity in response to TNF-alpha and promotes inflammation (XREF_FIG).

      Mutated TP53 activates inflammatory response.

    47. Several evidence demonstrate that mutant p53 promotes glycolysis and reprograms the cellular metabolism of cancer cells.

      Mutated TP53 activates glycolytic process.

    48. In absence of AMPK, mitochondrial stress augments aerobic glycolysis, also called " Warburg effect " in tumor cells, which is promoted by mutant p53.

      Mutated TP53 activates glycolytic process.

    49. Although these studies highlight that mutant p53 mediated EMT phenotype confer stemness in cancer cells, however, there is still a lot to explore in context of molecular mechanisms of mutant p53 driven stemness through activation of EMT genes.
    50. Gain-of function mutant p53 further promotes EMT and stemness phenotypes by activating genes regulating them.
    51. However, whether mutant p53 induced EMT trigger stemness properties in cancer cells, is still quite unexplored.
    52. The sustained activation of NF-kB signaling by mutant p53 not only elevate inflammatory response but also protects the cancer cells from cytotoxic effects of tumor microenvironment by activating pro survival pathways.

      Mutated TP53 activates NFkappaB.

    53. While wild type p53 suppresses inflammatory response by inhibiting the production of cytokines and antagonizing NF-kB activity, mutant p53 on the other hand enhances NF-kB activity in response to TNF-alpha and promotes inflammation (XREF_FIG).

      Mutated TP53 activates NFkappaB.

    54. Interaction of mutant p53 to SREBPs activates mevalonate pathway that promotes invasion in breast cancer cells (XREF_FIG).

      Mutated TP53 activates mevalonic acid.

    55. Mutant p53 can also induce YAP and TAZ nuclear localization by interacting with SREBP and activating the mevalonate pathway.

      Mutated TP53 activates mevalonic acid.

    56. Similarly, p53 activation by nutlin leads to transcriptional activation of p21 that cause cell cycle arrest and induces differentiation in human ESCs.
    57. Similarly , p53 activation by nutlin leads to transcriptional activation of p21 that cause cell cycle arrest and induces differentiation in human ESCs ( 35 ) .

      Nutlin activates TP53.

    58. Acetylation of p53 at K373 by CBP/p300 leads to dissociation of HDM2 and TRIM24 and subsequent activation of p53 which in turn transcriptionally activates p21, miR-34a, and miR-145 ( xref ).

      TP53 is acetylated on K373.

    1. Increased Expression of EZH2 Is Mediated by Higher Glycolysis and mTORC1 Activation in Lupus CD4 + T Cells.

      CD4 increases the amount of EZH2.

    2. Increased Expression of EZH2 Is Mediated by Higher Glycolysis and mTORC1 Activation in Lupus CD4

      CD4 increases the amount of EZH2.

    3. In summary, our findings suggest that EZH2 overexpression in SLE CD4 + T cells is induced by mTORC1 activation and increased glycolysis through effects on post-transcriptional regulation by miR-26a and miR-101 (XREF_FIG).

      mTORC1 increases the amount of EZH2.

    4. Increased Expression of EZH2 Is Mediated by Higher Glycolysis and mTORC1 Activation in Lupus CD4 + T Cells.

      mTORC1 increases the amount of EZH2.

    5. Indeed, inhibiting mTORC1 increased miR-26a and miR-101 and suppressed EZH2 expression in SLE CD4 + T cells.

      mTORC1 increases the amount of EZH2.

    6. Increased Expression of EZH2 Is Mediated by Higher Glycolysis and mTORC1 Activation in Lupus CD4

      mTORC1 increases the amount of EZH2.

    7. There are three potential mechanisms to explain these paradoxical findings : ( 1 ) Oxidative stress can induce EZH2 expression by additional mechanisms independent of our suggested mTORC1 / glycolysis / miRNA axis ; ( 2 ) A negative feedback homeostasis loop may exist to suppress EZH2 via miR-26a and miR-101 as EZH2 levels were significantly increased about 40 folds with oxidative stress ; and ( 3 ) miR-26a and miR-101 could be directly regulated by oxidative stress independent of mTORC1 / glycolysis .

      Oxidative Stress activates EZH2.

    8. These results suggest that oxidative stress upregulated EZH2 expression , via mechanisms that might be independent of post-transcriptional regulation by miR-26a and miR-101 .

      Oxidative Stress activates EZH2.

    9. EZH2 mediates abnormal CD4 + T cells adhesion in SLE by epigenetic dysregulation of the junctional adhesion molecule A (JAM-A) [XREF_BIBR].

      EZH2 activates CD4.

    10. The mechanisms underlying EZH2 upregulation in SLE CD4 + T cells remain unknown.

      EZH2 activates CD4.

    11. This is consistent with EZH2 mediated epigenetic changes in naive CD4 + T cells that were previously observed when SLE becomes more active [XREF_BIBR].

      EZH2 activates CD4.

    12. Increased disease activity in SLE patients is associated with a proinflammatory epigenetic shift in naive CD4 + T cells, likely mediated by EZH2.

      EZH2 activates CD4.

    13. EZH2 mediates abnormal CD4 + T cells adhesion in SLE by epigenetic dysregulation of the junctional adhesion molecule A (JAM-A) [XREF_BIBR].

      EZH2 activates cell adhesion.

    14. In summary, our findings suggest that EZH2 overexpression in SLE CD4 + T cells is induced by mTORC1 activation and increased glycolysis through effects on post-transcriptional regulation by miR-26a and miR-101 (XREF_FIG).
    15. Increased EZH2 is mediated by activation of mTORC1 and increased glycolysis in SLE CD4 + T cells.
    16. Conclusion : Increased EZH2 is mediated by activation of mTORC1 and increased glycolysis in SLE CD4 + T cells .

      EZH2 activates mTORC1.

    17. Increased EZH2 is mediated by activation of mTORC1 and increased glycolysis in SLE CD4 + T cells .

      EZH2 activates mTORC1.

    18. Taken together, these data suggest that increased mTORC1 activity in SLE CD4 + T cells might mediate upregulation of EZH2 through increasing glycolysis and the resulting suppression of miR-26a and miR-101.

      mTORC1 activates EZH2.

    19. Because mTORC1 is activated in SLE CD4 + T cells in part due to increased oxidative stress , and mTORC1 activation increases glycolysis , we hypothesized that mTORC1 mediates increased EZH2 expression .

      mTORC1 activates EZH2.

    20. Because mTORC1 is activated in SLE CD4 + T cells in part due to increased oxidative stress , and mTORC1 activation increases glycolysis , we hypothesized that mTORC1 mediates increased EZH2 expression .

      mTORC1 activates EZH2.

    21. Increased EZH2 is mediated by activation of mTORC1 and increased glycolysis in SLE CD4 + T cells.

      mTORC1 activates EZH2.

    1. Mechanically, they elucidated that YC-1 facilitates E3-ligase c-Cbl phosphorylation at T731 and T774, leading to the activation of c-Cbl and complex formation with EZH2, and then EZH2 ubiquitination degradation.

      EZH2 is ubiquitinated.

    2. A series of small molecules have been shown to facilitate EZH2-ubiquitination degradation.

      EZH2 is ubiquitinated.

    3. As we discussed, the PTM of the EZH2-ubiquitination pathway is an important negative regulator of EZH2.

      EZH2 is ubiquitinated.

    4. For instance, CDK1-mediated pT345-EZH2 and pT487-EZH2 facilitate EZH2 ubiquitination degradation in breast cancer cell, cervical cancer cell and lung cancer cell [ xref , xref , xref ]; JAK2 phosphorylates Y641-EZH2, leading to E3 ligase β-TrCP-mediated EZH2 degradation in lymphoma cell [ xref ]; and CDK5 phosphorylation of EZH2 at T261 residue results in the E3 ubiquitin ligase FBW7-mediated degradation of EZH2 in pancreatic cancer cell [ xref ].

      EZH2 is ubiquitinated.

    5. Mechanistically, YC-1 treatment promotes c-Cbl phosphorylation at T731 and T774, which results in c-Cbl-induced Src and ERK activation, leading to the formation of the c-Cbl-ERK-EZH2 complex and the consequent accumulation of EZH2 ubiquitination and proteasomal degradation.

      EZH2 is ubiquitinated.

    6. The first research of EZH2 ubiquitination was from the Aaron lab’s work in 2011 [ xref ].

      EZH2 is ubiquitinated.

    7. Ubiquitination, sumoylation, and deubiquitination of EZH2 in tumorigenesis and cancer metastasis.

      EZH2 is ubiquitinated.

    8. We showed that R342-EZH2 methylation inhibits TRAF6-mediated EZH2 ubiquitination [ xref ].

      EZH2 is ubiquitinated.

    9. Another recent study has reported that CDK5-mediated T261-EZH2 phosphorylation facilitates FBW7-mediated EZH2 ubiquitination and proteasome degradation in pancreatic cancer cells [ xref ].

      EZH2 is ubiquitinated.

    10. Moreover, researchers demonstrated that E3 ligase CHIP can mediate EZH2 ubiquitination degradation and subsequently derepress EZH2-silenced tumor suppressor genes by attenuating the H3K27me3 level in head and neck cancer cells [ xref ].

      EZH2 is ubiquitinated.

    11. They found that pT345-EZH2 and pT487-EZH2 facilitate EZH2 ubiquitination and hence its degradation by the proteasome pathway in human cervical cancer cells [ xref ].

      EZH2 is ubiquitinated.

    12. Our recent study has also confirmed that CDK1-mediated pT345-EZH2 and pT487-EZH2 facilitate EZH2 ubiquitination and subsequent degradation in breast cancer [ xref ].

      EZH2 is ubiquitinated.

    13. Moreover, a recent report has revealed that SYDM2 catalyzes EZH2-K307 di-methylation attenuating EZH2-ubiquitination degradation in breast cancer [ xref ].

      EZH2 is ubiquitinated.

    14. OGT-mediated O -GlcNAcylation of EZH2 attenuates EZH2 ubiquitination in breast cancer cell [ xref , xref ]; PCAF-mediated EZH2-K348 acetylation inhibits CDK1 catalyzing pT345-EZH2 and pT487-EZH2 and increases EZH2 stability in lung cancer [ xref ].

      EZH2 is ubiquitinated.

    15. SETD2-mediated mono-methylation of EZH2-K735 promotes EZH2 ubiquitination in prostate cancer [ xref ].

      EZH2 is ubiquitinated.

    16. They confirmed that circ-ADD3 binding with EZH2 facilitates CDK1-mediated EZH2 phosphorylation on T345 and T487, which results in EZH2 ubiquitination degradation in HCC cells.

      EZH2 is ubiquitinated.

    17. Sun et al. found that circ-ADD3, as a circular RNA, inhibits hepatocellular carcinoma (HCC) metastasis through facilitating EZH2 degradation through CDK1-mediated EZH2 ubiquitination [ xref ].

      EZH2 is ubiquitinated.

    18. Subsequently, the researchers designed a highly specific NEK2 inhibitor, CMP3a, which can promote EZH2 ubiquitination degradation and inhibit GBM tumor growth.

      EZH2 is ubiquitinated.

    19. Ubiquitination, sumoylation, and deubiquitination of EZH2 in tumorigenesis and cancer metastasis.

      EZH2 is sumoylated.

    20. A study showed that sumoylation of EZH2 is associated with EZH2 activity in U2OS cell (osteosarcoma cell line) [ xref ].

      EZH2 is sumoylated.

    21. Moreover, another study demonstrated that JAK3-mediated EZH2 tyrosine (Y) Y244 phosphorylation, which suppresses PRC2 complex formation, resulting in EZH2 oncogenic function independent of its HMTase activity in natural killer/T-cell lymphoma (NKTL) [ xref ].

      EZH2 is phosphorylated on Y244.

    22. In addition, p38 catalyzing EZH2 phosphorylation at T367 residue elevates its localized to cytoplasm and promotes breast cancer cells distant metastasis [ xref ].

      EZH2 is phosphorylated on T367.

    23. In addition, our recently studies discovered that PRMT1-mediated EZH2-R342 methylation attenuates CDK1-mediated EZH2-T345 and EZH2-T487 phosphorylation, which strengthens EZH2 stability [ xref , xref ].

      EZH2 is phosphorylated on T345.

    24. They confirmed that circ-ADD3 binding with EZH2 facilitates CDK1-mediated EZH2 phosphorylation on T345 and T487, which results in EZH2 ubiquitination degradation in HCC cells.

      EZH2 is phosphorylated on T345.

    25. Our recent research has illustrated that ANCR, a type of lncRNAs, promotes EZH2-T345 phosphorylation by associating with EZH2 [ xref ].

      EZH2 is phosphorylated on T345.

    26. For instance, CDK1-mediated pT345-EZH2 and pT487-EZH2 facilitate EZH2 ubiquitination degradation in breast cancer cell, cervical cancer cell and lung cancer cell [ xref , xref , xref ]; JAK2 phosphorylates Y641-EZH2, leading to E3 ligase β-TrCP-mediated EZH2 degradation in lymphoma cell [ xref ]; and CDK5 phosphorylation of EZH2 at T261 residue results in the E3 ubiquitin ligase FBW7-mediated degradation of EZH2 in pancreatic cancer cell [ xref ].

      CDK5 phosphorylates EZH2 on T261.

    27. For instance, CDK1 mediated pT345-EZH2 and pT487-EZH2 facilitate EZH2 ubiquitination degradation in breast cancer cell, cervical cancer cell and lung cancer cell [XREF_BIBR, XREF_BIBR, XREF_BIBR]; JAK2 phosphorylates Y641-EZH2, leading to E3 ligase beta-TrCP-mediated EZH2 degradation in lymphoma cell [XREF_BIBR]; and CDK5 phosphorylation of EZH2 at T261 residue results in the E3 ubiquitin ligase FBW7 mediated degradation of EZH2 in pancreatic cancer cell [XREF_BIBR].

      CDK5 phosphorylates EZH2 on T261.

    28. Talha et al. [ xref ] revealed that p38 phosphorylated EZH2 at T367 site facilitating its cytoplasmic localization and interacting with vinculin and other cytoskeletal regulators of cell migration and invasion.

      p38 phosphorylates EZH2.

    29. In 2018, Li et al. [ xref ] demonstrated that AMPK phosphorylates EZH2 at T311 residue to inhibit EZH2 binding with SUZ12, thereby attenuating the PRC2-dependent methylation of H3K27 and enhancing PRC2 target genes translation in ovarian and breast cancers.

      AMPK phosphorylates EZH2 on T311.

    30. In 2018, Li et al. [XREF_BIBR] demonstrated that AMPK phosphorylates EZH2 at T311 residue to inhibit EZH2 binding with SUZ12, thereby attenuating the PRC2 dependent methylation of H3K27 and enhancing PRC2 target genes translation in ovarian and breast cancers.

      AMPK phosphorylates EZH2 on T311.

    31. As early as 2005, Cha et al. [XREF_BIBR] showed phosphorylation of EZH2 at S21 (pS21-EZH2) by PI3K and AKT signaling in breast cancer cells.

      AKT leads to the phosphorylation of EZH2 on S21.

    32. SETD2-mediated mono-methylation of EZH2-K735 promotes EZH2 ubiquitination in prostate cancer [ xref ].

      EZH2 is methylated on K735.

    33. Furthermore, Kim et al. [ xref ] found that AKT-induced pS21-EZH2 elevates EZH2-mediated STAT3 methylation by increasing EZH2-STAT3 interaction in glioblastoma multiforme (GBM) stem-like cells.

      EZH2 methylates STAT3.

    34. In 2020, Yuan et al. [ xref ] reported that SETD2 methylates EZH2 at K735 promoting EZH2 degradation and impeding prostate cancer metastasis.

      SETD2 methylates EZH2.

    35. A study demonstrated that the phosphorylation of EZH2 at Y646 residue in human (Y641 in mouse) by JAK2 promotes the beta-TrCP-mediated EZH2 degradation and consequent regulation of H3K27me3 [XREF_BIBR].

      EZH2 inhibits EZH2.

    36. In 2020, Yuan et al. [XREF_BIBR] reported that SETD2 methylates EZH2 at K735 promoting EZH2 degradation and impeding prostate cancer metastasis.

      EZH2 inhibits EZH2.

    37. AKT-mediated pS21-EZH2 inhibits its methyltransferase activity by attenuating EZH2 associated with histone H3, which attenuates H3K27me3 level, increases EZH2 target genes expression, and facilitates breast cancer tumorigenesis.
    38. Moreover , Jin et al. [ 50 ] revealed that FBW7 decreases EZH2 activity and attenuates the motility of pancreatic cancer cells by mediating the degradation of the EZH2 ubiquitin proteasome pathway .

      FBXW7 inhibits EZH2.

    39. Moreover, Jin et al. [XREF_BIBR] revealed that FBW7 decreases EZH2 activity and attenuates the motility of pancreatic cancer cells by mediating the degradation of the EZH2 ubiquitin proteasome pathway.

      FBXW7 inhibits EZH2.

    40. Recently , a report has confirmed that Praja1 degrades EZH2 during skeletal myogenesis [ 38 ] .

      PJA1 inhibits EZH2.

    41. Aaron and his colleagues illustrated that Praja1 promotes EZH2 degradation through K48-linkage polyubiquitination and suppresses cells growth and migration in breast cancer [ 87 ] .

      PJA1 inhibits EZH2.

    42. Silvia et al. [XREF_BIBR] revealed that p38alpha promotes E3 ligase Praja1 mediated EZH2 degradation through the phosphorylation of T372-EZH2 (T367-EZH2 in mouse).

      PJA1 inhibits EZH2.

    43. This finding disclosed that Praja1 mediated EZH2 degradation is required for muscle satellite cells differentiation.

      PJA1 inhibits EZH2.

    44. Recently, a report has confirmed that Praja1 degrades EZH2 during skeletal myogenesis [XREF_BIBR].

      PJA1 inhibits EZH2.

    45. Aaron and his colleagues illustrated that Praja1 promotes EZH2 degradation through K48-linkage polyubiquitination and suppresses cells growth and migration in breast cancer [XREF_BIBR].

      PJA1 inhibits EZH2.

    46. They found that Ub E3 ligase Praja1 mediates EZH2 protein degradation through the ubiquitination-proteasome pathway in MCF7 cells (breast cancer cell line).

      PJA1 inhibits EZH2.

    47. A recent research has disclosed that sorafenib can prevent EZH2 expression by accelerating its ubiquitination-proteasome degradation in hepatoma cells [ 117 ] .

      sorafenib inhibits EZH2.

    48. Moreover, NSC745885, as a small molecular, is derived from natural anthraquinone emodin, which can downregulate EZH2 via proteasome mediated degradation [XREF_BIBR].

      emodin inhibits EZH2.

    49. Besides, Ma and his colleagues found that Ubiquitin specific protease 1 (USP1) directly interacts with and deubiquitinates EZH2.

      Protease deubiquitinates EZH2.

    50. For instance, EZH2 can promote the invasion and metastasis by suppressing E-cadherin transcriptional expression [XREF_BIBR, XREF_BIBR]; EZH2 can also increase tumorigenesis by silencing tumor suppressors [XREF_BIBR, XREF_BIBR, XREF_BIBR].

      EZH2 decreases the amount of CDH1.

    51. Reports on AKT-mediated pS21-EZH2 support the presumption that pS21-EZH2 mediated by AKT results in EZH2 promoting oncogenesis by several novel functions, which is independent on PRC2-mediated target gene transcriptional silencing.

      EZH2 binds PROS1.

    52. Interestingly, arsenic-induced pS21-EZH2 is mainly cytoplasmic localization.

      EZH2 binds PROS1.

    53. Instead of transcriptional repression EZH2 target gene expression, pS21-EZH2 serves as a transcriptional co-activator in castration-resistant prostate cancer through PI3K/AKT signaling [ xref ].

      EZH2 binds PROS1.

    54. AKT-mediated pS21-EZH2 inhibits its methyltransferase activity by attenuating EZH2 associated with histone H3, which attenuates H3K27me3 level, increases EZH2 target genes expression, and facilitates breast cancer tumorigenesis.

      EZH2 binds PROS1.

    55. As early as 2005, Cha et al. [ xref ] showed phosphorylation of EZH2 at S21 (pS21-EZH2) by PI3K/AKT signaling in breast cancer cells.

      EZH2 binds PROS1.

    56. For instance, AKT-mediated pS21-EZH2 can promote breast cancer tumorigenesis [ xref , xref ].

      EZH2 binds PROS1.

    57. They confirmed that the OGT-EZH2 axis inhibits tumor suppression by repressing the expression of several key tumor suppression genes in breast carcinoma.

      EZH2 binds OGT.

    58. They demonstrated that ZRANB1 can bind, deubiquitinate, and stabilize EZH2, which enhances breast cancer tumorigenesis and metastasis.

      ZRANB1 binds EZH2.

    59. They demonstrated that ZRANB1 can bind, deubiquitinate, and stabilize EZH2, which enhances breast cancer tumorigenesis and metastasis.

      ZRANB1 binds EZH2.

    60. We disclosed that ANCR-EZH2 interaction enhances CDK1 binding with EZH2 and increases the amount of pT345-EZH2, which results in EZH2 degradation and subsequently suppressing the oncogenesis and distant metastasis in breast cancer.

      CDK1 binds EZH2.

    61. We disclosed that ANCR-EZH2 interaction enhances CDK1 binding with EZH2 and increases the amount of pT345-EZH2, which results in EZH2 degradation and subsequently suppressing the oncogenesis and distant metastasis in breast cancer.

      CDK1 binds EZH2.

    62. Although AKT-mediated-EZH2-S21 phosphorylation reduces its affinity toward histone H3, it does not change its subcellular localization or its interaction with Polycomb group protein SUZ12 and EED subunits.

      SUZ12 binds EED and EED.

    63. A study revealed that Smurf2 can interact with EZH2 and mediate EZH2 ubiquitination-proteasome degradation.

      SMURF2 binds EZH2.

    64. A study revealed that Smurf2 can interact with EZH2 and mediate EZH2 ubiquitination-proteasome degradation.

      SMURF2 binds EZH2.

    65. We speculate that ANCR-EZH2 association may change the conformation of EZH2, which probably facilitates the recognition and binding of CDK1 on EZH2 to phosphorylate its T345 residue.

      UBE3A binds EZH2.

    66. We disclosed that ANCR-EZH2 interaction enhances CDK1 binding with EZH2 and increases the amount of pT345-EZH2, which results in EZH2 degradation and subsequently suppressing the oncogenesis and distant metastasis in breast cancer.

      UBE3A binds EZH2.

    67. Furthermore, Kim et al. [ xref ] found that AKT-induced pS21-EZH2 elevates EZH2-mediated STAT3 methylation by increasing EZH2-STAT3 interaction in glioblastoma multiforme (GBM) stem-like cells.

      STAT3 binds EZH2.

    68. It means that OGT mediated EZH2 GlcNAcylation have several different functions in breast cancer progression.Acetylation is a reversible and important PTM that regulates a series of cellular processes, including proliferation, apoptosis, migration, and metabolism, in cancer cells; it is achieved through the modulation of core histones or non histone proteins by histone acetyltransferases (HATs) or histone deacetylases (HDACs) [XREF_BIBR - XREF_BIBR].

      OGT activates EZH2.

    69. This report also found that OGT mediated O GlcNAcylation at S75 stabilizes EZH2 and subsequently facilitates the formation of H3K27me3 on PRC2 target genes.

      OGT activates EZH2.

    70. Professor Wong 's team first provided convincing evidence on OGT mediated EZH2 O GlcNAcylation at S75 in breast cancer [XREF_BIBR].

      OGT activates EZH2.