Page 20 - Read Online
P. 20

of cells in gliomas with properties of stem cells, known as   also contribute to radioresistance in classical-GBM.  Thus,
                                                                                                       [43]
            glioma-initiating cells (GICs). These cells express neural   inhibiting TGF-β has been proposed as a treatment since
            stem cell markers such as NESTIN, OLIG2, sex determining   it induces radiosensitivity  in gliomas through decreasing
            region  Y-box 2 (SOX2) and fucosyltransferase  4. [20,24,25]    the expression of miR-1 and -125a. [23,39,44]  Additionally,
            The  classification  of  GICs  is  based  on  the  expression  of   miR-221/222  downregulate  PTEN, leading  to activation
            prominin-1 (CD133); the CD133  GICs are more invasive   of proteins that promote cell proliferation or prevent cell
                                       +
            than those that do not express the antigen, and constitute   death, such as: AKT, B-cell lymphoma 2 (Bcl-2), Cyclin-D,
            3-29% of the glioma mass. [26,27]  Also, GICs from secondary   matrix metallopeptidase 2 and 9, [41,45,46]  Interestingly,
            cultures conserve the characteristics of the original tumor,   the  phosphorylation  of AKT is the  main  mechanism  for
            even after several passages.  Self-renewal, aggressiveness   developing radioresistance, regardless of the activity  of
                                  [28]
            and stemness of GICs are associated with the expression of   its negative regulator PTEN. [40,41]  In summary, molecular
            Cyclin E and proteins of the family of inhibitor of DNA-  analysis could help to reconsider whether conventional
            binding/differentiation  proteins, increased  activity  of   treatments are suitable, or therapeutic modifications should
            several signaling pathways including: transforming growth   be adapted to the requirements of the patient.
            factor (TGF)-β; protein kinase A and jagged-NOTCH; [26,29]
            as well as the activation of the receptors for PDGF, epidermal   GROWTH-ARREST-SPECIFIC 1
            growth factor (EGF) and fibroblast growth factor (FGF).
                                                         [26]
            Surprisingly, CD133  GICs  also contribute to malignant   The balance  between proliferation or growth arrest is
                             +
            transformation  of the adjacent  normal glial  cells through   regulated  by several extrinsic  and intrinsic  factors. Cells
            paracrine activity of PDGF-α and stimulate angiogenesis by   can exit the cell cycle  and enter in the non-proliferative
            activation of the NOTCH signaling pathway on endothelial   phase,  known  as  the  G0  phase.  Particularly,  in  the  this
            tissue. [26,28,30]                                 phase six genes named growth-arrest-specific (Gas) genes
                                                               are expressed, from 1 to 6.  The gas1 transcript is the
                                                                                      [47]
            It is noteworthy that CD133  GICs are predisposed to   most abundant in NIH3T3 cells arrested in the G0 phase by
                                    +
            become resistant to chemo- and radiotherapy with respect   deprivation of serum or high cell density. [47-49]  Gas1 induces
            to non-GICs cells, implicating the activation of extracellular   growth arrest by inhibiting DNA synthesis in NIH3T3 cells
            signal-regulated  kinases  1  and  2  (ERK1/2). [20,31,32]  In this   when it is ectopically expressed. [49]
            context,  the use of classical  therapies  against  gliomas,
            facilitate  the selection  of multi-resistant  GICs, leading   TRANSCRIPTIONAL AND
            to the formation  of more aggressive tumors, resistant  to   TRANSLATIONAL REGULATION
            chemo- and radiotherapy.  Moreover, in vitro and in vivo
                                [33]
            studies have shown that with the adequate stimulus, GICs   Human and mouse gas1 genes are located in the long arms
                                                                                                            [52]
            differentiate to either neuronal or astrocytic cells, however   of chromosome 9 (9q21.3-q22) [50,51]  and chromosome 13
            many of these responses are deregulated in gliomas. [31]  respectively, with 77.04% of homology  between  them.
                                                               Gas1 is an intronless gene, suggesting that it probably
            Currently, the analysis of protein expression and of the   originated from a retrotransposon. [53]
            transcriptome, including micro-RNAs (miRNAs), has helped
            to uncover molecular markers involved in the susceptibility   There are few studies about the regulation of the gas1 gene,
            or resistance to treatments. Indeed, differential expression   however  it  has  been  reported  that  Menin  and  Myb-like
            patterns  of miRNAs have been  reported  in high-grade   (coded by the men1 and dmp1 genes respectively), induce the
            gliomas.  Even now, the association of miRNAs with the   transcriptional repression of gas1. [54,55]  Also, c-Myc and Src
                   [34]
            methylation of the MGMT promoter is still controversial   repress gas1 transcription, since they facilitate re-entering to the
            since miRNAs are not considered as direct  epigenetic   cell cycle. [56,57]  c-Myc protein requires the Myc-Box2 domain
            regulators. [21,35]  However, overexpression of miR-222,   to be present on the N-terminus to repress the transcription of
            -145 and -132 is related with TMZ resistance coupled with   gas1. Furthermore, the basic Helix-loop-Helix leucine zipper
            MGMT promoter methylation.  Additionally,  miR-181b   domain located at the C-terminus of c-Myc is also necessary
                                     [35]
            and -181c are downregulated in patients that responded to   to induce the transcriptional repression of  gas1, perhaps
            radiotherapy and concomitant TMZ.  However, sensibility   together  with  an  accessory  protein  not  yet  identified. [56,58]
                                        [36]
            to chemotherapy is not only dependent of the presence of   Both c-Myc and Src are key components for the proliferation,
            MGMT. [37,38]                                      growth, and survival of glioma cells. The expression of c-Myc
                                                               closely correlates both with cellular dedifferentiation and the
            Screening of molecular changes after radiotherapy showed   grade  of malignancy, [59-61]  since  its  activity  induces  the
            overexpression  of miR-1,  -125a,  -144, -150, -151-5p,   transcripcion of cyclin D1 and repression of the p21 WAF1/
            -221/22, -425 and -1285. [39-42]   The ectopic expression of   CIP1  cyclin-dependent kinase inhibitor.  Interestingly, the
                                                                                             [62]
            miR-1,  -125a, -150 and  -425 increases  cell  survival  and   histone  chaperone,  Facilitate  Chromatin  Transcription
            confers radioresistance through the induction of the cell-  protein complex (FACT) increases the transcription of Myc,
            cycle.  On the other hand, TGF-α and -β, and the EGFR   a recent report showed that the downregulation or inhibition
                [39]

                        Journal of Cancer Metastasis and Treatment ¦ Volume 2 ¦ March 11, 2016 ¦           103
   15   16   17   18   19   20   21   22   23   24   25