Page 62 - Read Online
P. 62

Page 58                          Hunt et al. Extracell Vesicles Circ Nucleic Acids 2020;1:57-62  I  http://dx.doi.org/10.20517/evcna.2020.04

               Keywords: MicroRNA, microenvironment, adrenergic neurons, solid tumors, neurotrophic growth, neuron-tumor
               crosstalk




               Cancers are predominantly characterized by their loss of proliferative control. Genetic changes drive
               this increased proliferation, resulting in the formation, growth and spread of tumor cells throughout
               the body. Mutations that commonly drive tumor growth, however, also drive molecular changes within
               the tumor and the surrounding tissues. Encircling the solid tumors are collections of healthy cells that
               typically act to support cell types and tissue functions in the local region. These cells include structural
               fibroblasts, immune cells, neurons, blood vessels, and other cell types that in combination are known
               as the tumor microenvironment. In the context of cancer, these supporting cells can be manipulated to
               support the growth and spread of the tumor. These manipulative relationships become essential for the
                                                                                 [6]
                                                          [1,2]
                                                                                       [7,8]
               survival of the cancer and are found in prostate , gastric [3-5] , pancreatic , skin , glioma [9-11] , and a
               variety of other tumor types [12–15] . Subsequently, efforts to interrupt the relationships between solid tumors
                               [16]
                                                                          [17]
               and immune cells , as well as between tumors and blood vessels  have shown efficacy in stymying
               tumor growth, demonstrating the widespread therapeutic potential borne from understanding these
               tumor-microenvironment relationships. To date, the relationships between tumors and other members
               of the tumor microenvironment are not well understood. Additionally, the mechanisms by which these
                                                                                                    [18]
               relationships are formed and sustained are not well documented. Recent work from Amit et al.  has
               uncovered that tumors use extracellular vesicular signaling to drive nearby neuron survival, growth, spread,
               and subtype switching, which, in turn, drives tumor growth.

               This recent work focused on oral cavity squamous cell carcinoma (OCSCC), an aggressive tumor arising
               from mouth epithelial cells. To study these tumors in greater depth, the group leveraged laboratory mouse
               models of OCSCC. These include mice in which the tumor suppressor p53 was conditionally knocked
                                                                                            cre
               out of epithelial cells (Krt5 ; Tp53 flox/flox ). The control group for these mice lacked the Krt5  allele, leaving
                                      cre
               Tp53 intact in all cells. In these models, tumor initiation was induced via introduction of the carcinogen,
               4-nitroquinoline 1-oxide (4NQO) into the drinking water. Additionally, the group used patient-derived
                                         WT
                                                 null
               xenograft models in which p53  or p53  OCSCC cells were injected into the tongues of mice and allowed
               to grow. Finally, in cell culture dishes, dorsal root ganglia (DRG) were co-cultured with oral keratinocytes
                      WT
                              null
               and p53  or p53  OCSCC cells to model neural interactions with OCSCC tumors in vitro.
               Initially, the group found that in tumor samples derived from both their conditional p53 knockout
               mice, as well as their patient-derived xenograft models, loss of p53 coincided with increased adrenergic
               nerve density within the tumor. These findings were recapitulated in human OSCSS samples, indicating
               that tumor cells lacking p53 were driving increased adrenergic neuritogenesis. When testing this
                                                                             [18]
               same hypothesis using in vitro OCSCC-DRG co-cultures, Amit et al.  concordantly found increased
                                      null
                                                                WT
               DRG innervation of p53  OCSCC cells. When p53  OCSCC-DRG co-cultures were incubated
                                                                                  null
               with conditioned medium containing extracellular vesicles (EVs) from p53  OCSCC cells, the same
                                                                   null
               neuritogenic effect was observed. Additionally, when the p53  OCSCC cells co-cultured with DRGs were
                                                                                    null
               inhibited from releasing EVs, the effect was lost, thus demonstrating that the p53 -derived EVs and their
               contents were the driving force behind the described neuritogenic effect.
               Extracellular vesicles serve as important vehicles of small regulatory RNA species, which are known to be
               essential for proper neuronal development and function. By comparing the small RNAs found within EVs
                              WT
                                        null
               derived from p53  and p53  cell lines, Amit et al.  found that 17 microRNAs (miRNAs) were down-
                                                            [18]
                             null
               regulated in p53  cells. After narrowing this collection of miRNAs to the most down-regulated species,
                                                                                                        null
               they found that several of these miRNAs, including miR-34a, were also down-regulated in the p53
   57   58   59   60   61   62   63   64   65   66   67