Page 122 - Read Online
P. 122

Page 306                                        Ponnusamy et al. Cancer Drug Resist 2019;2:297-312  I  http://dx.doi.org/10.20517/cdr.2018.11

               N-Cadherin expression . Lysine-specific demethylase-1 (LSD1) caused global reduction in H3K9Me2
                                   [73]
               and increase in H3K4Me3 and H3K36Me3 during TGF-β-mediated EMT and associated migration and
               chemoresistance . Other mechanisms of LSD1 as to EMT process have been reported while they are
                             [62]
               inconclusive for breast tumors . Moreover, EMT associated miRNAs and their modulation through
                                          [8]
               epigenetic regulation have been reviewed . These evidences further underwrite the epigenetic landscape
                                                   [8]
               regulating EMT phenotype to cause acquired resistance.
               Epigenetic dysregulation triggering formation of CSC phenotype, maintaining their self-renewal and
               apoptotic resistance to chemotherapy have been accruing. Inactivating mutations of epigenetic regulatory
               genes including DNMT3A, TET, and PRC2 complex genes aberrantly activate CSC pathways and associated
               resistance. Similarly, non-mutational epigenetic aberrations of genes KDM5B, G9a/EHMT2, EZH2, PRC1,
               BMI, BRD4, PRMT5, KDM1A/2A, MLL and SWI/SNF involved in dysregulation of CSC-self-renewal,
               stemness maintenance and drug sensitivity have been reviewed . Gene silencing mediated through
                                                                         [6]
               DNMTs, EZH2/BMI1 and HDAC1, and activation through MLL and CBP potentially induce aberrations
               associated with BCRP, E-cadherin, p16 and Wnt signaling in CSCs . Epigenetic alterations evident to
                                                                           [52]
               induce intratumoral heterogeneity through modulating microenvironment and associated signaling which
               supports CSCs including Wnt, TGF-β, and Notch signaling [6,23] . Transient drug-tolerant states generated by
               reversible poised chromatin state are regulated through histone demethylase RBP2/KDM5A/Jarid1A and
               Notch signaling, thus, help CSCs to evade cytotoxicity mainly by equipping them with complex resistance
               mechanisms .
                          [22]
               Similar to epigenetic regulating proteins, the TFs associated with EMT and CSC work coordinately to
               activate the downstream effects related to drug resistance. Cancer cells with stemness known to express
               high levels of metastatic markers such as Snail1, Twist 1 and FOXC2. Elevated FOXC2 in basal-like breast
               cancer cells activate EMT-CSC signaling and reduce drug sensitivity . Similarly, overexpression of Twist
                                                                          [74]
               selects cells with CD44-high/ CD24-low subpopulation that have increased MRP1 expression and activate
               β-catenin/Akt pathways to maintain stemness . Similarly, Stem cell factor BMI-1, which is also part of
                                                       [75]
               PRC1 complex reportedly involve in chemoresistance . Twist 1 directly activates the BMI1 expression and
                                                            [76]
               they together orchestrate the EMT and stemness induction and maintenance. BMI-1 also shown to regulate
               stem cell renewal and induce EMT through its co-ordination with Nanog and NF-κB pathway . Meanwhile
                                                                                              [77]
               loss of BMI-1 increased sensitivity of breast cancer cells to doxorubicin .
                                                                           [77]
               Epigenetic deregulation associated with signaling pathways involved in EMT-CSC co-ordination further
               implicate their roles in chemoresistance. For example, Wnt ligands and sequester proteins including Wnt5A,
               WIF-1, FZD and SFRP known to be modulated through promoter methylation and histone modifications in
               breast cancer . Aberrations in epigenetic regulations evaluating TGF-β, Hh and Notch signaling in CSCs
                          [39]
               are scarce. Hh signaling ligands SHH, PTCH1 and SOX17 frequently methylated in breast cancer initiating
               cells and Hh effector proteins KIF7 and SUF7 deregulated through histone modifications and miRNA
               while GLI1 expression is elevated due to loss of KMT-SETD7 methylase in breast cancer cells thus drive the
               aberrant Hh as well as associated NF-κB signaling . Epigenetic mechanisms regulating Notch signaling
                                                           [39]
               occur through HES, a  Notch effector and transcriptional repressor, which recruit  histone acetylases/
               deacetylases and through hypermethylation mediated silencing of DLL1 (Notch ligand). Taken together,
               all these evidences implicate the epigenetic aberrations dictating the activation of EMT-CSC pathways in
               acquired chemoresistance in breast cancer.


               Impact of treatment schedule on resistant development through acquisition of EMT and CSC-like
               phenotype and associated temporal epigenetic changes
               In support of the reports discussed in previous section, we are highlighting, in this section, the recent findings
               from our laboratory demonstrating the influence of treatment schedules on acquired chemoresistance in
   117   118   119   120   121   122   123   124   125   126   127