SCC15 aggressively migrated to SCC4 or SCC9 and pushed these cells to undergo retrograde migration (Supplementary Fig

SCC15 aggressively migrated to SCC4 or SCC9 and pushed these cells to undergo retrograde migration (Supplementary Fig.?5cCf; Supplementary Video 7). requests to the authors. Spreadsheets and Supplementary Videos 1C12 and Supplementary Videos 13C42 for Supplementary Figs. 2 and 5 are deposited at Figshare (10.6084/m9.figshare.14560782). Legends for all videos is shown in the Description of Additional Supplementary Files. Abstract Metastatic cancer cells are (+)-MK 801 Maleate frequently deficient in WWOX protein or express dysfunctional WWOX (designated WWOXd). Here, we determined that functional WWOX-expressing (WWOXf) cells migrate collectively and expel the individually migrating WWOXd cells. For return, WWOXd cells induces apoptosis of WWOXf cells from a remote distance. Survival of WWOXd from the cell-to-cell encounter is due to activation of the survival IB/ERK/WWOX signaling. Mechanistically, cell surface epitope WWOX286-299 (repl) in WWOXf repels the invading WWOXd to undergo retrograde migration. However, when epitope WWOX7-21 (gre) is exposed, WWOXf greets WWOXd to migrate forward for merge. WWOX binds membrane type II TGF receptor (TRII), and TRII IgG-pretreated Mouse monoclonal to TIP60 WWOXf greet WWOXd to migrate forward and merge with each other. In contrast, TRII IgG-pretreated WWOXd loses recognition by WWOXf, and WWOXf mediates apoptosis of WWOXd. The observatons suggest that?normal cells can be activated to attack metastatic cancer cells.?WWOXd cells are less efficient in generating Ca2+ influx and undergo non-apoptotic explosion in response to UV irradiation in room temperature. WWOXf cells exhibit bubbling cell death and Ca2+ influx effectively caused by UV or apoptotic stress. Together, membrane WWOX/TRII complex is needed for cell-to-cell recognition, maintaining the efficacy of Ca2+ influx, and control of cell invasiveness. gene encodes a 46-kDa tumor suppressor protein WW domain-containing oxidoreductase, designated WWOX, FOR, or WOX11C6. WWOX participates in the regulation of many signal pathways (e.g., p53, TGF-, Wnt, Hippo, YAP/TAZ, Hyal-2, etc.) for cell growth and differentiation and organ development7C13. Alterations in these pathways due to WWOX dysfunction may facilitate disease progression7C13. Aberrant WWOX phosphorylation also enhances the progression of cancer and Alzheimers disease (AD)9,14C16. For example, transition of?proapoptotic pY33-WWOX to prosurvival pS14-WWOX promotes the progression of both cancer15,16 and AD17. Human newborns deficient in WWOX suffer severe neural diseases, metabolic disorders, and early death, but fail to form tumors spontaneously14,18C23. When functional WWOX starts to downregulate in the brain of middle-aged healthy individuals, this may lead to slow aggregation of a cascade of brain proteins that eventually form tau tangles and amyloid beta plaques in old ages14,24C28. WWOX has recently been established as one of the AD (+)-MK 801 Maleate risk factors29. (+)-MK 801 Maleate As a partner of WWOX binding proteins, p53 may functionally counteract with WWOX that leads to neural inflammation and aggregate formation of tau, amyloid beta and other proteins in vivo as (+)-MK 801 Maleate shown in the AD pathologies30. gene is located on a common fragile site on chromosome ch16q23.3-24.1, alterations of this gene is associated with cancer development1C6. Loss of heterozygosity (LOH) of human gene may go as high as 30C50%. Human gene is rarely mutated. Majority of metastatic cancer cells are deficient in WWOX protein due to epigenetic modification1C6 and translational blockade of mRNA31. Tumor suppressor WWOX is anchored, in part, in the cell membrane by Hyal-2, as determined by immunoelectron microscopy7,13,32. Also, membrane WWOX may undergo self-polymerization7,32. Proapoptotic pY33-WWOX binds Hyal-2 and Smad47,32. When the signaling complex WWOX/Hyal-2/Smad4 is transiently overexpressed, TGF- or hyaluronan induces internalization of the signaling complex to relocate?the nucleus for causing cell death7,32. Most recently, we have identified two cell surface-exposed WWOX epitopes, namely WWOX7-21 and WWOX286-29916,33. Synthetic WWOX7-21 peptide potently enhances ceritinib-mediated breast 4T1 cell death33. Also, WWOX7-21 peptide significantly enhances ceritinib-mediated explosion and death of 4T1 stem cell spheres33. The proapoptotic effect of WWOX7-21 peptide is due to (1) reduction of ERK phosphorylation, (2) significant upregulation of pY33-WWOX, (3) rapid increases in Ca2+ influx, and (4) disruption of the IB/WWOX/ERK prosurvival signaling33. In stark contrast, pS14-WWOX7-21 peptide strongly supports cancer growth in vivo and blocks ceritinib-mediated apoptosis in vitro33. Overall, endogenous pY33-WWOX is proapoptotic and acts as a tumor suppressor4,31,34C36. In contrast, pS14-WWOX strongly supports lymphocytic cell differentiation9,37 but enhances the progression of cancer15,16 and AD17. A majority of metastatic cancer cells are devoid of (+)-MK 801 Maleate WWOX expression or possess dysfunctional WWOX protein1C6. These cells tend to have an enhanced mobility and increased sensitivity to microenvironment38. For example, when WWOX-deficient or -dysfunctional cells (hereby designated WWOXd) encounter functional WWOX-expressing (WWOXf) cells, WWOXd cells migrate forward.