16 research outputs found

    Mitochondrial dysfunction induced by a SH2 domain-Targeting STAT3 inhibitor leads to metabolic synthetic lethality in cancer cells

    Get PDF
    In addition to its canonical role in nuclear transcription, signal transducer and activator of transcription 3 (STAT3) is emerging as an important regulator of mitochondrial function. Here, we demonstrate that a novel inhibitor that binds with high affinity to the STAT3 SH2 domain triggers a complex cascade of events initiated by interference with mitochondrial STAT3 (mSTAT3). The mSTAT3\u2013drug interaction leads to mitochondrial dysfunction, accumulation of proteotoxic STAT3 aggregates, and cell death. The cytotoxic effects depend directly on the drug\u2019s ability to interfere with mSTAT3 and mitochondrial function, as demonstrated by site-directed mutagenesis and use of STAT3 knockout and mitochondria-depleted cells. Importantly, the lethal consequences of mSTAT3 inhibition are enhanced by glucose starvation and by increased reliance of cancer cells and tumor-initiating cells on mitochondria, resulting in potent activity in cell cultures and tumor xenografts in mice. These findings can be exploited for eliciting synthetic lethality in metabolically stressed cancer cells using highaffinity STAT3 inhibitors. Thus, this study provides insights on the role of mSTAT3 in cancer cells and a conceptual framework for developing more effective cancer therapies

    Atypical E2Fs either Counteract or Cooperate with RB during Tumorigenesis Depending on Tissue Context

    Get PDF
    Simple Summary In virtually all human malignancies, the CDK-RB-E2F pathway is dysregulated resulting in the activation of the E2F transcriptional network. Rb and atypical E2Fs are the most important negative regulators of E2F-dependent transcription during tumorigenesis. However, it is unknown whether they cooporate or act independently in tumor development. Here we show that combined loss of RB and atypical E2Fs in mice enhances tumorigenesis in the liver, while in the pituitary gland, we observe inhibition of tumorigenesis. These findings suggest that the interaction between RB and atypical E2Fs in controlling tumorigenesis occurs in a tissue cell-type specific manner. E2F-transcription factors activate many genes involved in cell cycle progression, DNA repair, and apoptosis. Hence, E2F-dependent transcription must be tightly regulated to prevent tumorigenesis, and therefore metazoan cells possess multiple E2F regulation mechanisms. The best-known is the Retinoblastoma protein (RB), which is mutated in many cancers. Atypical E2Fs (E2F7 and -8) can repress E2F-target gene expression independently of RB and are rarely mutated in cancer. Therefore, they may act as emergency brakes in RB-mutated cells to suppress tumor growth. Currently, it is unknown if and how RB and atypical E2Fs functionally interact in vivo. Here, we demonstrate that mice with liver-specific combinatorial deletion of Rb and E2f7/8 have reduced life-spans compared to E2f7/8 or Rb deletion alone. This was associated with increased proliferation and enhanced malignant progression of liver tumors. Hence, atypical repressor E2Fs and RB cooperatively act as tumor suppressors in hepatocytes. In contrast, loss of either E2f7 or E2f8 largely prevented the formation of pituitary tumors in Rb+/- mice. To test whether atypical E2Fs can also function as oncogenes independent of RB loss, we induced long-term overexpression of E2f7 or E2f8 in mice. E2F7 and -8 overexpression increased the incidence of tumors in the lungs, but not in other tissues. Collectively, these data show that atypical E2Fs can promote but also inhibit tumorigenesis depending on tissue type and RB status. We propose that the complex interactions between atypical E2Fs and RB on maintenance of genetic stability underlie this context-dependency

    Mitochondrial dysfunction induced by a SH2 domain-targeting STAT3 inhibitor leads to metabolic synthetic lethality in cancer cells

    Get PDF
    In addition to its canonical role in nuclear transcription, signal transducer and activator of transcription 3 (STAT3) is emerging as an important regulator of mitochondrial function. Here, we demonstrate that a novel inhibitor that binds with high affinity to the STAT3 SH2 domain triggers a complex cascade of events initiated by interference with mitochondrial STAT3 (mSTAT3). The mSTAT3–drug interaction leads to mitochondrial dysfunction, accumulation of proteotoxic STAT3 aggregates, and cell death. The cytotoxic effects depend directly on the drug’s ability to interfere with mSTAT3 and mitochondrial function, as demonstrated by site-directed mutagenesis and use of STAT3 knockout and mitochondria- depleted cells. Importantly, the lethal consequences of mSTAT3 inhibition are enhanced by glucose starvation and by increased reliance of cancer cells and tumor-initiating cells on mitochondria, resulting in potent activity in cell cultures and tumor xenografts in mice. These findings can be exploited for eliciting synthetic lethality in metabolically stressed cancer cells using high-affinity STAT3 inhibitors. Thus, this study provides insights on the role of mSTAT3 in cancer cells and a conceptual framework for developing more effective cancer therapies

    Surgical resection and radiofrequency ablation initiate cancer in cytokeratin-19(+)- liver cells deficient for p53 and Rb

    Get PDF
    The long term prognosis of liver cancer patients remains unsatisfactory because of cancer recurrence after surgical interventions, particularly in patients with viral infections. Since hepatitis B and C viral proteins lead to inactivation of the tumor suppressors p53 and Retinoblastoma (Rb), we hypothesize that surgery in the context of p53/Rb inactivation initiate de novo tumorigenesis. We, therefore, generated transgenic mice with hepatocyte and cholangiocyte/liver progenitor cell (LPC)-specific deletion of p53 and Rb, by interbreeding conditional p53/Rb knockout mice with either Albumin-cre or Cytokeratin-19-cre transgenic mice. We show that liver cancer develops at the necrotic injury site after surgical resection or radiofrequency ablation in p53/Rb deficient livers. Cancer initiation occurs as a result of specific migration, expansion and transformation of cytokeratin-19+-liver (CK-19+) cells. At the injury site migrating CK-19+ cells formed small bile ducts and adjacent cells strongly expressed the transforming growth factor β (TGFβ). Isolated cytokeratin-19+ cells deficient for p53/Rb were resistant against hypoxia and TGFβ-mediated growth inhibition. CK-19+ specific deletion of p53/Rb verified that carcinomas at the injury site originates from cholangiocytes or liver progenitor cells. These findings suggest that human liver patients with hepatitis B and C viral infection or with mutations for p53 and Rb are at high risk to develop tumors at the surgical intervention site

    Novel functions for atypical E2Fs, E2F7 and E2F8, in polyploidization and liver cancer

    No full text
    Atypical E2F transcription factors, E2F7 and E2F8, function as transcriptional repressors of E2F target genes and are crucial for controlling the cell proliferation. In this thesis, we reveal that these two factors are crucial for liver cell polyploidization, embryonic development and prevention of liver cancer in mice. In chapter 2, we show that atypical E2Fs, especially E2F8, play an important role in promoting polyploidization in the liver. Loss of E2F7/8 resulted in dramatic reduction in binucleation and polyploidization of hepatocytes. In contrast, single loss of E2F1 enhanced polyploidization in hepatocytes. Interestingly, loss of E2f1 in E2f7/8-deficient liver led to partial rescue of polyploidization defect observed upon loss of E2f7/8. Moreover, we identified a transcriptional network program regulated by repressor E2F8 and activator E2F1 that controls polyploidization in liver. Together, these data suggest opposing functions for classical activator E2Fs and atypical repressor E2Fs in regulating polyploidization in liver. Surprisingly, contrary to long-term theory, we discovered that loss of polyploidization has no impact on liver differentiation and regeneration. We demonstrate in chapter 4 that E2F7 and E2F8 are required for murine embryonic development. While mice mutant for either E2F7 or E2F8 developed normally with no obvious developmental defects, global deletion of both E2F7 and 8 resulted in embryonic lethality owing to vascular and cell survival defects such as dilated blood vessels associated with multifocal hemorrhages and massive apoptosis. Interestingly, loss of either E2F1 or p53 suppressed the massive apoptosis observed in E2f7/8 double knockout embryos, however, the triple knockout embryos still carried the vascular defects and died around same embryonic age, suggesting complex mechanisms underlying the embryonic lethality. As described in chapter 5, we have revealed that E2f7/8 function as tumor suppressor genes and that they can cooperate with the known tumor suppressor gene Rb in preventing liver cancer. Using liver-specific knockout mice, we found that deletion of E2f7/8 results in spontaneous development of hepatocellular carcinoma in mice. Furthermore, additional deletion of Rb along with E2f7/8 resulted in development of tumors earlier than in E2f7/8 deficient livers, indicating that loss of Rb accelerates tumorigenesis

    Novel functions for atypical E2Fs, E2F7 and E2F8, in polyploidization and liver cancer

    No full text
    Atypical E2F transcription factors, E2F7 and E2F8, function as transcriptional repressors of E2F target genes and are crucial for controlling the cell proliferation. In this thesis, we reveal that these two factors are crucial for liver cell polyploidization, embryonic development and prevention of liver cancer in mice. In chapter 2, we show that atypical E2Fs, especially E2F8, play an important role in promoting polyploidization in the liver. Loss of E2F7/8 resulted in dramatic reduction in binucleation and polyploidization of hepatocytes. In contrast, single loss of E2F1 enhanced polyploidization in hepatocytes. Interestingly, loss of E2f1 in E2f7/8-deficient liver led to partial rescue of polyploidization defect observed upon loss of E2f7/8. Moreover, we identified a transcriptional network program regulated by repressor E2F8 and activator E2F1 that controls polyploidization in liver. Together, these data suggest opposing functions for classical activator E2Fs and atypical repressor E2Fs in regulating polyploidization in liver. Surprisingly, contrary to long-term theory, we discovered that loss of polyploidization has no impact on liver differentiation and regeneration. We demonstrate in chapter 4 that E2F7 and E2F8 are required for murine embryonic development. While mice mutant for either E2F7 or E2F8 developed normally with no obvious developmental defects, global deletion of both E2F7 and 8 resulted in embryonic lethality owing to vascular and cell survival defects such as dilated blood vessels associated with multifocal hemorrhages and massive apoptosis. Interestingly, loss of either E2F1 or p53 suppressed the massive apoptosis observed in E2f7/8 double knockout embryos, however, the triple knockout embryos still carried the vascular defects and died around same embryonic age, suggesting complex mechanisms underlying the embryonic lethality. As described in chapter 5, we have revealed that E2f7/8 function as tumor suppressor genes and that they can cooperate with the known tumor suppressor gene Rb in preventing liver cancer. Using liver-specific knockout mice, we found that deletion of E2f7/8 results in spontaneous development of hepatocellular carcinoma in mice. Furthermore, additional deletion of Rb along with E2f7/8 resulted in development of tumors earlier than in E2f7/8 deficient livers, indicating that loss of Rb accelerates tumorigenesis

    Mitochondrial Plasticity Promotes Resistance to Sorafenib and Vulnerability to STAT3 Inhibition in Human Hepatocellular Carcinoma

    No full text
    The multi-kinase inhibitor sorafenib is a primary treatment modality for advanced-stage hepatocellular carcinoma (HCC). However, the therapeutic benefits are short-lived due to innate and acquired resistance. Here, we examined how HCC cells respond to sorafenib and adapt to continuous and prolonged exposure to the drug. Sorafenib-adapted HCC cells show a profound reprogramming of mitochondria function and marked activation of genes required for mitochondrial protein translation and biogenesis. Mitochondrial ribosomal proteins and components of translation and import machinery are increased in sorafenib-resistant cells and sorafenib-refractory HCC patients show similar alterations. Sorafenib-adapted cells also exhibited increased serine 727 phosphorylated (pSer727) STAT3, the prevalent form in mitochondria, suggesting that STAT3 might be an actionable target to counteract resistance. Consistently, a small-molecule STAT3 inhibitor reduces pSer727, reverts mitochondrial alterations, and enhances the response to sorafenib in resistant cells. These results sustain the importance of mitochondria plasticity in response to sorafenib and identify a clinically actionable strategy for improving the treatment efficacy in HCC patients

    Atypical E2Fs either Counteract or Cooperate with RB during Tumorigenesis Depending on Tissue Context

    Get PDF
    E2F-transcription factors activate many genes involved in cell cycle progression, DNA repair, and apoptosis. Hence, E2F-dependent transcription must be tightly regulated to prevent tumorigenesis, and therefore metazoan cells possess multiple E2F regulation mechanisms. The best-known is the Retinoblastoma protein (RB), which is mutated in many cancers. Atypical E2Fs (E2F7 and -8) can repress E2F-target gene expression independently of RB and are rarely mutated in cancer. Therefore, they may act as emergency brakes in RB-mutated cells to suppress tumor growth. Currently, it is unknown if and how RB and atypical E2Fs functionally interact in vivo. Here, we demonstrate that mice with liver-specific combinatorial deletion of Rb and E2f7/8 have reduced life-spans compared to E2f7/8 or Rb deletion alone. This was associated with increased proliferation and enhanced malignant progression of liver tumors. Hence, atypical repressor E2Fs and RB cooperatively act as tumor suppressors in hepatocytes. In contrast, loss of either E2f7 or E2f8 largely prevented the formation of pituitary tumors in Rb+/- mice. To test whether atypical E2Fs can also function as oncogenes independent of RB loss, we induced long-term overexpression of E2f7 or E2f8 in mice. E2F7 and -8 overexpression increased the incidence of tumors in the lungs, but not in other tissues. Collectively, these data show that atypical E2Fs can promote but also inhibit tumorigenesis depending on tissue type and RB status. We propose that the complex interactions between atypical E2Fs and RB on maintenance of genetic stability underlie this context-dependency

    E2F8 is essential for polyploidization in mammalian cells

    No full text
    Polyploidization is observed in all mammalian species and is a characteristic feature of hepatocytes, but its molecular mechanism and biological significance are unknown. Hepatocyte polyploidization in rodents occurs through incomplete cytokinesis, starts after weaning and increases with age. Here, we show in mice that atypical E2F8 is induced after weaning and required for hepatocyte binucleation and polyploidization. A deficiency in E2f8 led to an increase in the expression level of E2F target genes promoting cytokinesis and thereby preventing polyploidization. In contrast, loss of E2f1 enhanced polyploidization and suppressed the polyploidization defect of hepatocytes deficient for atypical E2Fs. In addition, E2F8 and E2F1 were found on the same subset of target promoters. Contrary to the long-standing hypothesis that polyploidization indicates terminal differentiation and senescence, we show that prevention of polyploidization through inactivation of atypical E2Fs has, surprisingly, no impact on liver differentiation, zonation, metabolism and regeneration. Together, these results identify E2F8 as a repressor and E2F1 as an activator of a transcriptional network controlling polyploidization in mammalian cells
    corecore