Page 30 - Read Online
P. 30

Page 6 of 14                                  Lozano-Rosas et al. Hepatoma Res 2018;4:19  I  http://dx.doi.org/10.20517/2394-5079.2018.48

               change in the levels of an enzyme called MAT that catalyzes the formation of SAM has been implicated also.
               MAT is encoded by two genes, Mat1a and Mat2a. In HCC, the liver decreases the amount of MAT1A and
               increases MAT2A through epigenetic mechanisms alone; this switch is responsible for the decreasing level
               of SAM, favoring the development of this pathology [61,62] .

               Due to the above, it is necessary not only to consider the epigenetic modifications but also the generation of
               intermediaries by mitochondria that allow for the appropriate epigenetics control of both the mtDNA and
               the nDNA.

               Altered mitoepigenetics in HCC
               As afore mentioned, NRF1 and PGC1-α act on nuclear genes encoding respiratory subunits from ETC and
               are involved in the transcription and replication machinery. An up-regulation of this protein in HCC has
               been demonstrated [8,63] . An increased PGC1α level has been suggested to be an important inducer for the
               accumulation of dysfunctional mitochondria . Although the role of pgc1-α and nrf1 genes methylation has
                                                     [8]
               not been studied in HCC, this is an interesting area to be investigated. A meta-analysis of DNA methylation
               in HCC revealed a correlation between several aberrant methylated genes and the risk of HCC, among them
               p53 was hypermethylated in HCC tumor tissue compared to the adjacent tissue. It is important to consider
               that, in turn, this gene is implicated in the transcription and translocation of mtDNMT1 to mitochondria,
               and, in this way, besides of its role as tumor suppressor it could be modulating the methylation status of
               mitochondrial genes .
                                 [64]
               Under conditions of oxidative stress, which may be a factor for the development of HCC, the transcriptional
               and mitochondrial DNA replication machinery is altered. Consequently, the ETC loses its functionality
               and favors the accumulation of reactive oxygen species (ROS). In addition, the mtDNA may suffer injuries
               because of the accumulation ROS [1,65] . On the other hand, the mitochondrial damage observed in cancer cells
               can have consequences on the expression of nuclear genes. There are studies that indicate that the removal
               of mtDNA responds to changes in the nuclear genome [56,66] .


               A genome-wide mapping of DNA methylation and hydroxymethylation in a study on HBV-related HCC
               revealed that the metabolic pathways that include glycolysis, gluconeogenesis, oxidative phosphorylation,
               and TCA contained the largest number of (hydroxy) methylation-altered genes, indicating the crucial roles of
               metabolic processes that implicate mitochondria in the progression of HCC, which, in turn, are regulated by
               epigenetic mechanisms. The authors propose that some of the identified (hydroxy) methylation-altered genes
               may serve as biomarkers for the diagnosis and prognosis of HCC . Among the 5-mC and 5-hmC altered
                                                                       [67]
               genes related to OXPHOS were the following: NADH dehydrogenase [ubiquinone] 1 subunit C2 (NDUFC2),
               NADH dehydrogenase [ubiquinone] flavoprotein 1 (NDUFV1), NADH: ubiquinone oxidoreductase subunit
               S6 (NDUFS6) from complex 1 and succinate dehydrogenase complex flavoprotein subunit A (SDHA)
               from complex II. Among the TCA genes: succinyl-CoA ligase [GDP-forming] subunit beta (SUCLG2) and
               pyruvate carboxylase.

               Also, in HBV-induced hepatic carcinogenesis, protein X (HBx), encoded by the virus, has been proposed
               as an epigenetic regulator for tumor suppressor genes, by hypermethylation. It has been suggested that,
               in hepatomas, NQO1 (NAD(P)H: quinone oxidoreductase 1), which is a cytosolic protein that catalyzes
               two-electron reduction, can be deregulated by induction of HBx, generating mitochondrial damage and
               increasing oxidant stress in cells through hypermethylation of the NQO1 promoter .
                                                                                      [68]
               Specifically  in  mitochondria,  some  epigenetic  modifications  have  been  described  in  HCC,  such  as
               hypermethylation of the Mrps12 (mitochondrial ribosomal protein S12), Mgrap (mitochondria-localized
               glutamic acid-rich protein), and  Tmem70 (transmembrane protein 70) genes . The TMEM70 protein,
                                                                                   [69]
   25   26   27   28   29   30   31   32   33   34   35