Article

The Enhancement of Radiation Sensitivity in Nasopharyngeal Carcinoma Cells via Activation of the Rac1/NADPH Signaling Pathway

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract

We reported in an earlier study that using mass spectrometry and bioinformatic analysis demonstrated Rac1 proteins might be mostly mitochondrial targets in the radiosensitization process of nasopharyngeal carcinoma CNE-1 cells. The goal of our current study was to reveal the relationship between Rac1/NADPH pathway and radiosensitization in CNE-1 cells using Rac1 activator, phorbol 12-myristate 13-acetate (PMA) and Rac1 inhibitor NSC23766. The Rac1-GTP expression was determined using a pulldown assay, the Rac1 location using a immunofluorescence with a laser scanning confocal microscope, the NADPH oxidase activity with NBT assay and the reactive oxygen species with DCFH-DA probe. The apoptosis rate was analyzed by flow cytometry, and the expressions of p67(phox) and NFκB-p105/p50 were analyzed by Western blot. After treatment with PMA and 2 Gy radiation (compared to the control), Rac1-GTP was activated and translocated to the cell membrane. NADPH oxidase activity, reactive oxygen species of intracellular concentration and the apoptosis rate increased significantly. The expression of p67(phox) and NFκB-p50 protein also increased. However, in the cells treated with NSC23766 alone or NSC23766 combined with 2 Gy irradiation, the results were just the opposite. Overall, these results indicate that the Rac1 protein may be the key target involved in the radiosensitization of nasopharyngeal carcinoma cells. The activated Rac1/NADPH pathway combined with radiation can increase the radiosensitivity of nasopharyngeal carcinoma cells, and the Rac1/NADPH pathway may be the signaling pathway involved in the radiosensitization process.

No full-text available

Request Full-text Paper PDF

To read the full-text of this research,
you can request a copy directly from the authors.

... In addition, marked activation of Rac1 is detected in IR-exposed breast cancer cells, cervical carcinoma cells, and breast cancer cells [29,32,33]. In human nasopharyngeal carcinoma cells, Rac1 is detected more abundantly in the active GTP-bound form and with translocation to cell membrane under the treatment with IR [38]. Intriguingly, the activation of Rac1 under radiotherapy can be regarded as a protective mechanism that increases radioresistance, or conversely the downstream signaling of IR-mediated apoptotic pathways which enhances radiation-induced cytotoxicity. ...
... On the contrary, Rac1 has also been identified as an enhancer of radiosensitivity in nasopharyngeal carcinoma cells [38,39] and NSCLC cells [36,37]. In nasopharyngeal carcinoma cells, the enhanced activity of Rac1 is correlated with increased radiosensitivity [38,39]. ...
... On the contrary, Rac1 has also been identified as an enhancer of radiosensitivity in nasopharyngeal carcinoma cells [38,39] and NSCLC cells [36,37]. In nasopharyngeal carcinoma cells, the enhanced activity of Rac1 is correlated with increased radiosensitivity [38,39]. In addition, Rac1-N17 suppresses radiation-induced apoptosis in NSCLC cells [36]. ...
Article
Full-text available
Despite treatment advances, radioresistance and metastasis markedly impair the benefits of radiotherapy to patients with malignancies. Functioning as molecular switches, Rho guanosine triphosphatases (GTPases) have well-recognized roles in regulating various downstream signaling pathways in a wide range of cancers. In recent years, accumulating evidence indicates the involvement of Rho GTPases in cancer radiotherapeutic efficacy and metastasis, as well as radiation-induced metastasis. The functions of Rho GTPases in radiotherapeutic efficacy are divergent and context-dependent, thereby a comprehensive integration of their roles and correlated mechanisms is urgently needed. This review integrates current evidence supporting the roles of Rho GTPases in mediating radiotherapeutic efficacy and the underlying mechanisms. In addition, their correlations with metastasis and radiation-induced metastasis are discussed. Under the prudent application of Rho GTPase inhibitors based on critical evaluations of biological contexts, targeting Rho GTPases can be a promising strategy in overcoming radioresistance and simultaneously reducing the metastatic potential of tumor cells.
... These data suggest that overexpression of Rac1 increased the sensitivity of tumors to radiation in NPC xenograft tumor model in nude mice. The results are consistent with the findings of Wang et al [11]. ...
Article
Purpose: To study the effect of overexpression of Rac1 on the radiosensitivity of transplanted nasopharyngeal carcinoma (NPC) in nude mice, and the mechanism involved. Methods: Forty Lucky SPF-grade male thymus-free nude mice were used. Mice were divided into 4 groups: overexpression control, overexpression Rac1, Rac1 inhibition, and inhibition control groups, each with 10 mice. The prepared cell lines were treated with 6 MV x-ray. Before and after radiation, the growth of tumors in each group was monitored. Histomorphological images of nude mice tumors were obtained using hematoxylin and eosin (H&E) stains. Protein expressions of p67, P47, and Rac1 were evaluated bu Western blotting. Results: Transplanted tumor growth slowed down after 20 days. Growth rate was significantly higher in Rac1 and Rac1 overexpression groups than in overexpression and inhibition control groups (p < 0.05). Overexpression of Rac1 resulted in more cell necrosis, incomplete cellular structure, severe nuclear fragmentation, nuclear pyknosis, and cytotoxic red staining in endoscopic tumor tissues (p < 0.05). There were significantly lower expression levels of p67, P47, and Rac1 in Rac1 invasion group than in invasion control and overexpression control groups, while the expression levels of p67 and P47 were significantly higher in overexpression Rac1 group and IR overexpression Rac1 group than in inhibition control (p < 0.05). However, concentrations of p67 and P47 were significantly higher in overexpression Rac1 and IR overexpression Rac1 groups than in inhibition control mice. Conclusion: Rac1 increases the radiosensitivity of NPC xenografts in nude mice via a mechanism related to the regulation of expressions of proteins associated with Rac1/NADPH signaling pathway. Thus, Rac1 is potentially a new target for radio-sensitization of NPC.
... Thus, ionizing radiation can induces ROS productions by activating the cellular Rac1/NADPH signaling pathway 30,31 . The redox states of cells are balanced by pro-oxidants and antioxidants generated during host enzymatic redox reactions. ...
Article
Full-text available
Radiotherapy potentially offers protection from recurrence of tumor that also causes normal tissue damage and creates major concern. Another important factor is long-term immune suppression in patients treated with radiotherapy. Therefore, crucial need for the survival of surrounding normal cells of tumor by radiation‑protecting agents is the prime focus of this study. Aqueous extract (AE) and ethanolic extract (EE), Tanacetum parthenium extracts100 mg/kg each and parthenolide (PAR) 4mg/kg body weight were orally administered prior to sub-lethal radiation dose exposure. Mice were used for the evaluation of radiation-mediated chromosomal aberrations in bone marrow cells and DNA break by comet assay in the blood lymphocytes of mice. The pro-inflammatory levels were determined by cytokine estimations namely interleukin‑2, interferongamma and tumor necrosis factor-alpha performed in the liver homogenate using ELISA kits. Thus the results demonstrated ameliorating, radio-mitigating and immune-stimulatory efficacy of AE, EE and PAR against radiation ‑induced DNA damage and immunosuppression by regulating cytokine.
... Thus, ionizing radiation can induces ROS productions by activating the cellular Rac1/NADPH signaling pathway 30,31 . The redox states of cells are balanced by pro-oxidants and antioxidants generated during host enzymatic redox reactions. ...
Article
Full-text available
Radiotherapy potentially offers protection from recurrence of tumor that also causes normal tissue damage and creates major concern. Another important factor is long-term immune suppression in patients treated with radiotherapy. Therefore, crucial need for the survival of surrounding normal cells of tumor by radiation-protecting agents is the prime focus of this study. Aqueous extract (AE) and ethanolic extract (EE), Tanacetum parthenium extracts100 mg/kg each and parthenolide (PAR) 4mg/kg body weight were orally administered prior to sub-lethal radiation dose exposure. Mice were used for the evaluation of radiation-mediated chromosomal aberrations in bone marrow cells and DNA break by comet assay in the blood lymphocytes of mice. The pro-inflammatory levels were determined by cytokine estimations namely interleukin-2, interferon-gamma and tumor necrosis factor-alpha performed in the liver homogenate using ELISA kits. Thus the results demonstrated ameliorating, radio-mitigating and immune-stimulatory efficacy of AE, EE and PAR against radiation-induced DNA damage and immunosuppression by regulating cytokine.
... Activated Rac1/NADPH pathway combined with radiation can enhance radiosensitivity of NPC cells. RAC1/NADPH pathway may be a signal pathway involved in radiosensitization [48]. The results of Qi et al. showed that RAC1 was highly expressed in NPC cells. ...
Article
Full-text available
Nasopharyngeal carcinoma (NPC) is one of the common malignant tumors in China, which occurs on the top and sidewalls of the nasopharyngeal cavity. The incidence of malignant tumors of the ear, nose and throat is the highest. However, little is known about the growth of the cells. Therefore, this study constructed a multi-regulator-driven NPC cell growth-related module, aiming to explore the mechanism of functional pathways regulating the proliferation of NPC cells in an all-round way. Firstly, differential expression analysis, co-expression analysis, enrichment analysis and connectivity analysis were synthesized to identify the intrinsic genes of expression disorder module. Subsequently, we analyzed the module by crosstalk, and observed the interaction between modules intuitively. Finally, based on hypergeometric test, the significance of multi-regulators on the regulation of potential modules is calculated. We obtained 17 cell growth-related expression disorder modules by 2148 gene modules focusing. These modules are mainly involved in the growth cycle of NPC cells, including cell proliferation, migration and apoptosis. At the same time, they mainly affect the proliferation and apoptosis of NPC cells through PI3K-AKT signaling pathway, NF-kappa B signaling pathway and Wnt signaling pathway. Based on the growth-related modules of NPC cells, we have obtained a series of non-coding RNAs (ncRNAs) including microRNA-92a-3p, microRNA-19a-3p and microRNA-130a-3p, play an important role in regulating the growth of NPC cells. Similarly, we also predicted transcription factors (involving E2F1, NFKB1, SP1, etc.) that may play a key role in cell growth-related modules. This study is based on cell growth-related expression disorder module to explore the regulatory role of its functional pathway on cell proliferation mechanism, which will help researchers to have a deeper understanding of the potential pathogenesis of NPC.
... Rac1 and its effectors can activate mitogenic pathways, including the MAPK cascades, leading to changes in cell cycle progression (Olson et al., 1995;Yang et al., 2008). Rac1 has been consistently involved in the activation of pro-survival pathways in cancer cells, such as NF-κB, thereby counterbalancing the apoptotic responses triggered by chemotherapeutic agents and radiation therapy (Friedland et al., 2007;Han et al., 2013;Wang et al., 2013Wang et al., , 2016Hein et al., 2016). Regarding cellular metabolism, Rac1 has been widely associated with macropinocytosis, a mechanism of endocytic uptake of extracellular fluid that assists in meeting the increased demand of nutrients required for cancer cells to proliferation (Erami et al., 2017;Ramirez et al., 2019). ...
Article
Full-text available
Lung cancer is the leading cause of cancer-related deaths worldwide, with non-small cell lung cancer (NSCLC) representing ∼85% of new diagnoses. The disease is often detected in an advanced metastatic stage, with poor prognosis and clinical outcome. In order to escape from the primary tumor, cancer cells acquire highly motile and invasive phenotypes that involve the dynamic reorganization of the actin cytoskeleton. These processes are tightly regulated by Rac1, a small G-protein that participates in the formation of actin-rich membrane protrusions required for cancer cell motility and for the secretion of extracellular matrix (ECM)-degrading proteases. In this perspective article we focus on the mechanisms leading to aberrant Rac1 signaling in NSCLC progression and metastasis, highlighting the role of Rac Guanine nucleotide Exchange Factors (GEFs). A plausible scenario is that specific Rac-GEFs activate discrete intracellular pools of Rac1, leading to unique functional responses in the context of specific oncogenic drivers, such as mutant EGFR or mutant KRAS. The identification of dysregulated Rac signaling regulators may serve to predict critical biomarkers for metastatic disease in lung cancer patients, ultimately aiding in refining patient prognosis and decision-making in the clinical setting.
... However, there were studies reported that Rac1 agonist (PMA) could improve the radiation sensitivity of nasopharyngeal carcinoma mainly by excessively increasing the ROS level after radiotherapy [33,34]. Our results were not contradictory to their studies. ...
Article
Aims: To investigate whether Rac1 inhibition can alleviate radiation-induced intestinal injury (RIII), meanwhile exist no protection on tumors. Materials and methods: Rac1 inhibition was achieved by its specific inhibitor, NSC23766. Mice were pretreated with different intraperitoneal injections, which were normal saline for NS group (N = 9), and 2.5 mg/kg and 5 mg/kg of NSC23766 for Low-Dose group (N = 9) and High-Dose group (N = 9), respectively. After total body irritation (10Gy), small intestinal tissues were collected for Hematoxylin-Eosin (H&E) staining and Terminal-deoxynucleotidyl Transferase Mediated dUTP Nick End Labeling (TUNEL). Intestinal epithelial and tumor cell lines, namely MODE-k and CT26, were used to further study the role of Rac1 inhibition on radiation damage. Flow cytometry was used to detect changes in reactive oxygen species production, cell cycles and mitochondrial membrane potential, the latter was also checked by fluorescence microscope. Changes of protein-expression associated with apoptosis and cell cycles were detected by Western blotting to explain the possible molecular mechanism. Key findings: Height of intestine villi and depth of crypt were higher (P < 0.01) and apoptosis ratio lower (P < 0.01) in High-Dose group compared with those in NS group. After radiation, Rac1 inhibition pre-treatment improved the vitality (P < 0.01) and reduced the apoptosis (P < 0.01) in MODE-k while yielded opposite results in CT-26, and reduced ROS production of MODE-k (P < 0.01) while had little effect on that of CT-26. Rac1 inhibition differently affected the cell cycles of normal cells and that of tumor cells. Significance: Inhibition of Rac1 could alleviate RIII, meanwhile assist the killing effect of radiation on tumor cells.
... Radiation-induced ROS production can derive from mitochondria which increases with oxygen tension [14]. Radiation can also induce ROS by activating the cytosolic Rac1/NADPH oxidase system [15,16]. Cellular redox conditions are regulated by the balance between anti-oxidants and pro-oxidants performed by a host of redox enzymatic reactions. ...
Article
Full-text available
Effective radiotherapy for cancer has relied on the promise of maximally eradicating tumor cells while minimally killing normal cells. Technological advancement has provided state-of-the-art instrumentation that enables delivery of radiotherapy with great precision to tumor lesions with substantial reduced injury to normal tissues. Moreover, better understanding of radiobiology, particularly the mechanisms of radiation sensitivity and resistance in tumor lesions and toxicity in normal tissues, has improved the treatment efficacy of radiotherapy. Previous mechanism-based studies have identified many cellular targets that can affect radiation sensitivity, notably reactive oxygen species, DNA-damaging response signals, and tumor microenvironments. Several radiation sensitizers and protectors have been developed and clinically evaluated; however, many of these results are inconclusive, indicating that improvement remains needed. In this era of personalized medicine in which patients' genetic variations, transcriptome and proteomics, tumor metabolism and microenvironment, and tumor immunity are available. These new developments have provided opportunity for new target discovery. Several radiotherapy sensitivity-associated "gene signatures" have been reported although clinical validations are needed. Recently, several immune modifiers have been shown to associate with improved radiotherapy in preclinical models and in early clinical trials. Combination of radiotherapy and immunocheckpoint blockade has shown promising results especially in targeting metastatic tumors through abscopal response. In this article, we succinctly review recent advancements in the areas of mechanism-driven targets and exploitation of new targets from current radio-oncogenomic and radiation-immunotherapeutic approaches that bear clinical implications for improving the treatment efficacy of radiotherapy.
... Besides heavy ion radiation, X-ray radiation could also significantly activate NADPH oxidase and enhance the production of ROS, leading to X-radiation-induced growth inhibition of nasopharyngeal carcinoma cells. 28 It has been observed that Nox4 expression continues to increase at all time points from 1 day to 6 months post X-ray radiation in mice lung tissue. 29 Similarly, low-dose g irradiation is capable of upregulating NADPH oxidase in central nervous system in mice. ...
Article
Full-text available
Increased oxidative stress plays an important role in heavy ion radiation–induced cell death. The mechanism involved in the generation of elevated reactive oxygen species (ROS) is not fully illustrated. Here we show that NADPH oxidase activation is closely related to heavy ion radiation–induced cell death via excessive ROS generation. Cell death and cellular ROS can be greatly reduced in irradiated cancer cells with the preincubation of diphenyleneiodium, an inhibitor of NADPH oxidase. Most of the NADPH oxidase (NOX) family proteins (NOX1, NOX2, NOX3, NOX4, and NOX5) showed increased expression after heavy ion irradiation. Meanwhile, the cytoplasmic subunit p47phox was translocated to the cell membrane and localized with NOX2 to form reactive NADPH oxidase. Our data suggest for the first time that ROS generation, as mediated by NADPH oxidase activation, could be an important contributor to heavy ion irradiation–induced cell death.
... PMA can activate the NADPH oxidase activity and NSC23766 inhibits its activity. No significant cytotoxicity to SKOV3-PM4 cells was found when the concentration of PMA and NSC23766 was 100 nM and 12.5 µM, respectively (21). Considering that the inhibitory rate of SKOV3-PM4 cells treated with rhein alone was <20%, the concentration of rhein was chosen as ≤11.3 µM. ...
Article
Matrix metalloproteinases (MMPs) are a family of calcium-dependent zinc-containing endopeptidases, which play an integral role in migration and invasion of ovarian cancer. Rac1 proteins might mostly influence cell migration and invasion by generating endogenous reactive oxygen species. Therefore, inhibiting MMPs and regulating the Rac1/ROS/MAPK/AP-1 pathway may be a new therapeutic strategy for ovarian cancer. In this study, we found that rhein could suppress the invasion and migration of SKOV3-PM4 cells with characteristics of directional highly lymphatic metastasis. Phorbol 12-myristate 13-acetate (PMA), which is a Rac1 activator, significantly enhanced the expression levels of MMP-2, -3, -9 and -19 proteins, whereas the results of rhein and Rac1 inhibitor NSC23766 were just the opposite. The inhibitory effects of rhein were associated with the upregulation of tissue inhibitors of metalloproteinase (TIMP)-1, TIMP-2 and NM23-H1. Subsequent mechanism studies revealed that rhein reduce the production of reactive oxygen species (ROS) and lower NADPH oxidase activity. Furthermore, rhein significantly inhibited JNK, AP-1 phosphorylation in the cells treated with PMA. The results obtained from the cells treated with NSC23766 alone or NSC23766 combined with rhein, were consistent with rhein treatment alone. Taken together, these results indicate that rhein may be a potential inhibitor of Rac1 and can inhibit the migration and invasion of SKOV3-PM4 cells through modulating matrix metalloproteinases and RAC1/ROS/MAPK/AP-1 signaling pathway-associated proteins.
Article
Radiotherapy has a high cure rate for early nasopharyngeal carcinoma(NPC). However, the radiation resistance of poorly differentiated NPC cells impacts the effectiveness of treatment of early-stage NPC patients. Here, we explored the relationship between Ras-related C3 botulinum toxin substrate 1(Rac1) expression and NPC radiosensitivity. In vitro and in vivo studies revealed that upregulation of Rac1, when combined with X-ray treatment, increased growth inhibition and induced remarkable morphological changes and apoptosis in CNE2 cells. Furthermore, rupturing of the cell and nuclear membranes, degeneration of the cristae and significant swelling of the mitochondria were observed, which were consistent with the high apoptotic rate. The Rac1(+) cells exhibited approximately 50% more migration compared with that of the NC and Rac1(-)cells. The overexpression of Rac1 can increase the radiation sensitivity of NPC CNE2 cells, and the mechanism may be closely related to the oxidative damage of mitochondria. Rac1 might be a potential target for radiosensitization in poorly differentiated NPC.
Article
Arsenic trioxide (ATO), a highly effective drug in treating acute promyelocytic leukemia with low toxicity, demonstrates a significant effect on lung cancer. The anti-cancer mechanisms of ATO include inhibition of cancer stem-like cells, induction of apoptosis, anti-angiogenesis, sensitization of chemotherapy and radiotherapy, anti-cancer effects of hypoxia, and immunoregulation properties. In addition, some studies have reported that different lung cancers respond differently to ATO. It was concluded on numerous studies that the rational combination of administration and encapsulation of ATO have promising potentials in increasing drug efficacy and decreasing adverse drug effects. We reviewed the efficacy of ATO in the treatment of lung cancer in recent years to provide some views for further study.
Article
Radiation resistance of nasopharyngeal carcinoma (NPC) is a joint effect caused by complex molecular mechanisms. The development of multi-target radiotherapy sensitization agents offered a promising method for the treatment of NPC. In this work, the probability of Rhein to be a multi-target radiotherapy sensitization agent was explored through computer aid virtual screening by inverse docking study. In order to validate the accuracy of the computational results, radiotherapy sensitization of Rhein to NPC cells and its effects on the expression of target proteins were evaluated separately by CCK8 assay and Western blotting analysis. Our result demonstrated that Rhein possessed strong binding affinity with RAC1 and HSP90. No cytotoxic concentration of Rhein had radiosensitization effect on nasopharyngeal carcinoma CNE1 cells. After treatment with Rhein and 2Gy radiation, the expression of RAC1 upregulated and the expression of HSP90 down-regulated in cells. Based on the above data, Rhein is likely to become an attractive lead compound for the future design of multi-target radiotherapy sensitization agents.
Article
Full-text available
The superoxide-generating NADPH oxidase of phagocytes consists of the membrane-associated cytochrome b 558 (a heterodimer of Nox2 and p22(phox)) and 4 cytosolic components: p47(phox), p67(phox), p40(phox), and the small GTPase, Rac, in complex with RhoGDI. Superoxide is produced by the NADPH-driven reduction of molecular oxygen, via a redox gradient located in Nox2. Electron flow in Nox2 is initiated by interaction with cytosolic components, which translocate to the membrane, p67(phox) playing the central role. The participation of Rac is expressed in the following sequence: (1) Translocation of the RacGDP-RhoGDI complex to the membrane; (2) Dissociation of RacGDP from RhoGDI; (3) GDP to GTP exchange on Rac, mediated by a guanine nucleotide exchange factor; (4) Binding of RacGTP to p67(phox); (5) Induction of a conformational change in p67(phox), promoting interaction with Nox2. The particular involvement of Rac in NADPH oxidase assembly serves as a paradigm for signaling by Rho GTPases, in general.
Article
Full-text available
Epidermal growth factor (EGF) may increase cell motility, an event implicated in cancer cell invasion and metastasis. However, the underlying mechanisms for EGF-induced cell motility remain elusive. In this study, we found that EGF treatment could activate Ras-related C3 botulinum toxin substrate 1 (Rac1), PI3K/Akt and p21-actived kinase (PAK1) along with cell migration. Ectopic expression of PAK1 K299R, a dominant negative PAK1 mutant, could largely abolish EGF-induced cell migration. Blocking PI3K/Akt signalling with LY294002 or Akt siRNA remarkably inhibited both EGF-induced PAK1 activation and cell migration. Furthermore, expression of dominant-negative Rac1 (T17N) could largely block EGF-induced PI3K/Akt-PAK1 activation and cell migration. Interestingly, EGF could induce a significant production of ROS, and N-acetyl-L-cysteine, a scavenger of ROS which abolished the EGF-induced ROS generation, cell migration, as well as activation of PI3K/Akt and PAK, but not Rac1. Our study demonstrated that EGF-induced cell migration involves a cascade of signalling events, including activation of Rac1, generation of ROS and subsequent activation of PI3K/Akt and PAK1.
Article
Full-text available
The present study was performed to determine whether genistein could inhibit in vivo LPS-induced alveolar macrophage TNF production and thus reduce the alveolar neutrophil influx following LPS. In vitro incubation with genistein completely inhibited LPS-induced TNF production by alveolar macrophages (AM) from BALB/c mice. Subsequently mice were pretreated with intraperitoneal genistein or vehicle, then received nasal LPS to induce an alveolitis. Genistein was then administered every eight hours for five days following LPS. At 24 hours after LPS, the bronchoalveolar lavage (BAL) TNF and ex vivo TNF production from AM, were lower in the genistein treated animals. As well, total BAL white blood cell (WBC) count was reduced in the genistein as compared to the vehicle-only group. The percent neutrophils and the resolution of neutrophils were similar between genistein and vehicle groups. Therefore, genistein was able to decrease AM TNF production, and was associated with a decrease in BAL WBC count post-LPS.
Article
Full-text available
RhoG is a member of the Rac-like subgroup of Rho GTPases and has been linked to a variety of different cellular functions. Nevertheless, many aspects of RhoG upstream and downstream signaling remain unclear; in particular, few extracellular stimuli that modulate RhoG activity have been identified. Here, we describe that stimulation of epithelial cells with epidermal growth factor leads to strong and rapid activation of RhoG. Importantly, this rapid activation was not observed with other growth factors tested. The kinetics of RhoG activation after epidermal growth factor (EGF) stimulation parallel the previously described Rac1 activation. However, we show that both GTPases are activated independently of one another. Kinase inhibition studies indicate that the rapid activation of RhoG and Rac1 after EGF treatment requires the activity of the EGF receptor kinase, but neither phosphatidylinositol 3-kinase nor Src kinases. By using nucleotide-free RhoG pull-down assays and small interfering RNA-mediated knockdown studies, we further show that guanine-nucleotide exchange factors (GEFs) of the Vav family mediate EGF-induced rapid activation of RhoG. In addition, we found that in certain cell types the recently described RhoG GEF PLEKHG6 can also contribute to the rapid activation of RhoG after EGF stimulation. Finally, we present results that show that RhoG has functions in EGF-stimulated cell migration and in regulating EGF receptor internalization.
Article
Full-text available
The Rho family of small GTPases are critical elements involved in the regulation of signal transduction cascades from extracellular stimuli to the cell nucleus, including the JNK/SAPK signaling pathway, the c-fos serum response factor, and the p70 S6 kinase. Here we report a novel signaling pathway activated by the Rho proteins that may be responsible for their biological activities, including cytoskeleton organization, transformation, apoptosis, and metastasis. The human RhoA, CDC42, and Rac-1 proteins efficiently induce the transcriptional activity of nuclear factor kappaB (NF-kappaB) by a mechanism that involves phosphorylation of Ikappa Balpha and translocation of p50/p50 and p50/p65 dimers to the nucleus, but independent of the Ras GTPase and the Raf-1 kinase. We also show that activation of NF-kappaB by TNFalpha depends on CDC42 and RhoA, but not Rac-1 proteins, because this activity is drastically inhibited by their respective dominant-negative mutants. In contrast, activation of NF-kappaB by UV light was not affected by Rho, CDC42, or Rac-1 dominant-negative mutants. Thus, members of the Rho family of GTPases are involved specifically in the regulation of NF-kappaB-dependent transcription.
Article
Full-text available
Members of the Rho family of small GTPases activate the nuclear factor κB (NF-κB) (Perona, R., Montaner, S., Saniger, L., Sánchez-Pérez, I., Bravo, R., and Lacal, J. C. (1997), Genes & Dev. 11, 463–475). We have investigated whether different members of the family of exchange factors specific for Rho proteins (Dbl family) could activate the transcription factor NF-κB and have explored both their specificity under in vivo conditions and the mechanisms involved. Activated forms of Dbl, Ost, and Vav proteins induce NF-κB activation. While the activation induced by the Vav oncogen was efficiently inhibited by a dominant negative mutant of Rac1, the corresponding mutant of Cdc42Hs was able to block selectively NF-κB activation mediated by Dbl. Finally, mutants of RhoA and Cdc42Hs, but not that of Rac1, inhibited the activation of NF-κB by Ost. Thus, under in vivo conditions, different members of the Dbl family are related to specific Rho GTPases for the regulation of NF-κB. Activation of NF-κB by Rho or Ras proteins is mutually independent. However, there is a link between the NF-κB and the c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) cascades since a dominant negative mutant of MEKK1 is able to inhibit NF-κB activation induced by Rac1 and Cdc42Hs proteins, but not by RhoA. These results indicate that, in mammalian cells, multiple pathways coexist for the activation of NF-κB, some of which are mediated by specific members of the Ras and Rho families of small GTPases.
Article
Full-text available
Reactive oxygen species (ROS) have been increasingly recognized as important components of cell signaling in addition to their well-established roles in host defense. The formation of ROS in phagocytic and nonphagocytic cells involves membrane-localized and Rac guanosine triphosphatase (GTPase)-regulated reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase(s). We discuss here the current molecular models for Rac GTPase action in the control of the phagocytic leukocyte NADPH oxidase. As a mechanistically detailed example of Rac GTPase signaling, the NADPH oxidase provides a potential paradigm for signaling by Rho family GTPases in general.
Article
Full-text available
Damage recognition by repair/checkpoint factors is the critical first step of the DNA damage response. DNA double strand breaks (DSBs) activate checkpoint signaling and are repaired by nonhomologous end-joining (NHEJ) and homologous recombination (HR) pathways. However, in vivo kinetics of the individual factor responses and the mechanism of pathway choice are not well understood. We report cell cycle and time course analyses of checkpoint activation by ataxia-telangiectasia mutated and damage site recruitment of the repair factors in response to laser-induced DSBs. We found that MRN acts as a DNA damage marker, continuously localizing at unrepaired damage sites. Damage recognition by NHEJ factors precedes that of HR factors. HR factor recruitment is not influenced by NHEJ factor assembly and occurs throughout interphase. Damage site retention of NHEJ factors is transient, whereas HR factors persist at unrepaired lesions, revealing unique roles of the two pathways in mammalian cells.
Article
Full-text available
Rac plays a pivotal role in the assembly of the superoxide-generating NADPH oxidase of phagocytes. In resting cells, Rac is found in the cytosol in complex with Rho GDP dissociation inhibitor (RhoGDI). NADPH oxidase assembly involves dissociation of the Rac·RhoGDI complex and translocation of Rac to the membrane. We reported that liposomes containing high concentrations of monovalent anionic phospholipids cause Rac·RhoGDI complex dissociation ( Ugolev, Y., Molshanski-Mor, S., Weinbaum, C., and Pick, E. (2006) J. Biol. Chem. 281, 19204-19219 ). We now designed an in vitro model mimicking membrane phospholipid remodeling during phagocyte stimulation in vivo. We showed that liposomes of “resting cell membrane” composition (less than 20 mol % monovalent anionic phospholipids), supplemented with 1 mol % of polyvalent anionic phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P3) in conjunction with constitutively active forms of the guanine nucleotide exchange factors (GEFs) for Rac, Trio, or Tiam1 and a non-hydrolyzable GTP analogue, cause dissociation of Rac1(GDP)·RhoGDI complexes, GDP to GTP exchange on Rac1, and binding of Rac1(GTP) to the liposomes. Complexes were not dissociated in the absence of GEF and GTP, and optimal dissociation required the presence of PtdIns(3,4,5)P3 in the liposomes. Dissociation of Rac1(GDP)·RhoGDI complexes was correlated with the affinity of particular GEF constructs, via the N-terminal pleckstrin homology domain, for PtdIns(3,4,5)P3 and involved GEF-mediated GDP to GTP exchange on Rac1. Phagocyte membranes enriched in PtdIns(3,4,5)P3 responded by NADPH oxidase activation upon exposure in vitro to Rac1(GDP)·RhoGDI complexes, p67phox, GTP, and Rac GEF constructs with affinity for PtdIns(3,4,5)P3 at a level superior to that of native membranes.
Article
Radiotherapy plays an important role in treatment of cancers with low toxicity to the surrounding normal tissues. However, it still fails to eradicate hypoxic tumors due to the occurrence of radioresistance. Therefore, the search for new radiation sensitizers is of great significance. Platinum (Pt) complexes have been identified as potential radiation sensitizers to increase the sensitivity of cancer cells to radiotherapy. In the present study, we have synthesized four Pt complexes containing (2 - benzimidazole [4, 5-f] - [1, 10] phenanthroline) ligand and found that they could effectively enhance the X-ray-induced growth inhibition against A375 human melanoma cells through induction of G2/M cell cycle arrest. In contrast, they showed much lower cytotoxicity toward human normal cells. The complexes also dramatically inhibited the TrxR activity and caused intracellular ROS overproduction, due to the Auger electron effect of heavy metal element under X- ray radiation. Excessive ROS triggered DNA damage and activated downstream signaling pathways, including the phosphorylation of p53 and p38MAPK, and down-regulation of phosphorylated AKT and ERK, finally resulted in increase of radiosensitivity and inhibition of tumor reproduction. Taken together, our results suggest that the synthetic Pt complexes could be further developed as sensitizers of X-ray radiotherapy.
Article
In our preliminary study, 1, 8-dihydroxy-3-acetyl-6-methyl-9, 10 anthraquinone (GXHSWAQ-1), synthesised according to the basic structure of emodin, exhibited a 1.58-fold radiosensitisation on nasopharyngeal carcinoma CNE-1 cells. This study demonstrated that its radiosensitisation activity was achieved by altering the mitochondrial structure: swollen volume, fragmented crista, and decreasing transmembrane potential (P<0.01). Using isobaric tag for relative and absolute quantitation (iTRAQ) technology, 1396 proteins were identified, and the differentially expressed proteins were involved in metabolism, cell proliferation, angiogenesis, DNA repair process according to the biological process clustering results. Bioinformatic analysis showed that CDH1, RAC1, CDC42 proteins might be mostly mitochondrial targets in the radiosensitisation process. Western blotting analyses verified the differential expression of these proteins.
Article
Inherent and acquired resistance of cancer cells is increasingly recognized as a significant impediment to effective radiation cancer treatment. As important intracellular factors, aberrant tumor transmembrane signal transduction pathways which include the prosurvival cascades (PI3K/Akt, MAPK/ERK and JAK/STAT) and proapoptosis pathways (Wnt, p53 and TNF-α/NF-κB) have been proved to be one of the crucial determinant of the probability of cell sensitivity to radiation in malignant lesions. Increasing researches imply that targeting the abnormal pathways that can regulate the activity of the DNA damage response and further influence the response of tumor cells to radiation may be optimal treatment of radiation sensitization. Preclinical and clinical evidences suggest that agents targeted aberrant tumor signals can effectively improve the therapeutic effect of ionizing radiation. So, in this review, we discuss the intricate interplay between tumor responses to radiation with the aberrant signal pathways and the potential druggable targets within the pathways to sensitize tumors without significant collateral damage to normal tissues; allied application of novel targeting compounds in manipulating the aberrant signal of tumor cells in the rational clinical treatments are also summarized. This article is protected by copyright. All rights reserved.
Article
Rotenone, a widely used pesticide, reproduces parkinsonism in rodents and associates with increased risk for Parkinson disease. We previously reported that rotenone increased superoxide production by stimulating the microglial phagocyte NADPH oxidase (PHOX). This study identified a novel mechanism by which rotenone activates PHOX. Ligand-binding assay revealed that rotenone directly bound to membrane gp91(phox), the catalytic subunit of PHOX; such binding was inhibited by diphenyleneiodonium, a PHOX inhibitor with a binding site on gp91(phox). Functional studies showed that both membrane and cytosolic subunits were required for rotenone-induced superoxide production in cell-free systems, intact phagocytes, and COS7 cells transfected with membrane subunits (gp91(phox)/p22(phox)) and cytosolic subunits (p67(phox) and p47(phox)). Rotenone-elicited extracellular superoxide release in p47(phox)-deficient macrophages suggested that rotenone enabled activation of PHOX through a p47(phox)-independent mechanism. Increased membrane translocation of p67(phox), elevated binding of p67(phox) to rotenone-treated membrane fractions, and coimmunoprecipitation of p67(phox) and gp91(phox) in rotenone-treated wild-type and p47(phox)-deficient macrophages indicated that p67(phox) played a critical role in rotenone-induced PHOX activation via its direct interaction with gp91(phox). Rac1, a Rho-like small GTPase, enhanced p67(phox)-gp91(phox) interaction; Rac1 inhibition decreased rotenone-elicited superoxide release. In conclusion, rotenone directly interacted with gp91(phox); such an interaction triggered membrane translocation of p67(phox), leading to PHOX activation and superoxide production.
Article
The need to both understand and minimize the side effects of brain irradiation is heightened by the ever-increasing number of patients with brain metastases that require treatment with whole brain irradiation (WBI); some 200,000 cancer patients/year receive partial or WBI. At the present time, there are no successful treatments for radiation-induced brain injury, nor are there any known effective preventive strategies. Data support a role for chronic oxidative stress in radiation-induced late effects. However, the pathogenic mechanism(s) involved remains unknown. One candidate source of reactive oxygen species (ROS) is nicotinamide adenosine dinucleotide phosphate (NADPH) oxidase, which converts molecular oxygen (O(2)) to the superoxide anion (O(2)(-)) on activation. We hypothesize that brain irradiation leads to activation of NADPH oxidase. We report that irradiating rat brain microvascular endothelial cells in vitro leads to increased (i) intracellular ROS generation, (ii) activation of the transcription factor NFkappaB, (iii) expression of ICAM-1 and PAI-1, and (iv) expression of Nox4, p22(phox), and p47(phox). Pharmacologic and genetic inhibition of NADPH oxidase blocked the radiation-mediated upregulation of intracellular ROS, activation of NFkappaB, and upregulation of ICAM-1 and PAI-1. These results suggest that activation of NADPH oxidase may play a role in radiation-induced oxidative stress.
Article
Resveratrol (RV), a natural plant polyphenol widely present in foods such as grapes, wine, and peanuts, has an ability to inhibit various stages of carcinogenesis in vitro and in vivo. In this report, we explored the roles of intrinsic and extrinsic apoptotic pathways during RV-induced apoptosis in human lung adenocarcinoma (ASTC-a-1) cells. After exposure of cells to different concentrations of RV, we found that RV induced concentration-dependent apoptosis. Fluorometric substrates assay and western blotting (WB) analysis showed that caspase-8 was not activated, which was further verified by monitoring the cleavage of Bid to tBid using fluorescence resonance energy transfer (FRET) microscopy imaging inside single living cells, indicating that extrinsic apoptotic pathway was not involved in RV-induced apoptosis. In addition, inhibition of caspases-3 or -9 but not caspase-8 using the specific inhibitors of caspases modestly but significantly attenuated RV-induced apoptosis. Moreover, flow cytometry (FCM) analysis showed that RV treatment induced time-dependent loss of mitochondrial membrane potential (∆ψ(m)), in combination with the activation of caspases-3 and -9; we therefore concluded that RV-induced apoptosis involved the intrinsic apoptotic pathway. It is noteworthy that RV treatment induced translocation of AIF from mitochondria to nucleus in a time dependent manner, and that knockdown of AIF remarkably attenuated RV-induced apoptosis. Collectively, our findings demonstrate that RV induces caspase-8-independent apoptosis via AIF and to a lesser extent caspase-9-dependent mitochondrial pathway in ASTC-a-1 cells.
Article
Lysophosphatidic acid (LPA) is produced by tumor cells and is present in the ascites fluid of ovarian cancer patients. To determine the role of endogenous LPA in the ovarian cancer cell line SKOV3, we treated cells with the LPA receptor antagonist VPC32183 and found that it inhibited cell growth and induced apoptosis. Exogenous LPA further stimulated ERK and Akt phosphorylation and NF-κB activity. To determine if reactive oxygen species (ROS), which have been implicated as second messengers in cell signaling, were also involved in LPA signaling, we treated cells with the NADPH oxidase inhibitor diphenyleneiodonium (DPI), and antioxidants N-acetyl cysteine, EUK-134 and curcumin, and showed that all blocked LPA-dependent NF-κB activity and cell proliferation. DPI and EUK-134 also inhibited Akt and ERK phosphorylation. LPA was shown to stimulate dichlorofluorescein fluorescence, though not in the presence of DPI, apocynin (an inhibitor of NADPH oxidase), VPC32183, or PEG-catalase. Akt phosphorylation was also inhibited by PEG-catalase and apocynin. These data indicate that NADPH oxidase is a major source of ROS and H(2)O(2) is critical for LPA-mediated signaling. Thus, LPA acts as a growth factor and prevents apoptosis in SKOV3 cells by signaling through redox-dependent activation of ERK, Akt, and NF-κB-dependent signaling pathways.
Article
Nuclear factor kappaB (NF-kappaB) transcription factors have a key role in many physiological processes such as innate and adaptive immune responses, cell proliferation, cell death, and inflammation. It has become clear that aberrant regulation of NF-kappaB and the signalling pathways that control its activity are involved in cancer development and progression, as well as in resistance to chemotherapy and radiotherapy. This article discusses recent evidence from cancer genetics and cancer genome studies that support the involvement of NF-kappaB in human cancer, particularly in multiple myeloma. The therapeutic potential and benefit of targeting NF-kappaB in cancer, and the possible complications and pitfalls of such an approach, are explored.
Article
Reactive oxygen species (ROS) are generated in response to growth factors, cytokines, G protein-coupled receptor agonists, or shear stress, and function as signaling molecules in nonphagocytes. However, it is poorly understood how freely diffusible ROS can activate specific signaling, so-called "redox signaling." NADPH oxidases are a major source of ROS and now recognized to have specific subcellular localizations, and this targeting to specific compartments is required for localized ROS production. One important mechanism may involve the interaction of oxidase subunits with various targeting proteins localized in lamellipodial leading edge and focal adhesions/complexes. ROS are believed to inactivate protein tyrosine phosphatases, thereby establishing a positive-feedback system that promotes activation of specific redox signaling pathways involved in various functions. Additionally, ROS production may be localized through interactions of NADPH oxidase with signaling platforms associated with caveolae/lipid rafts, endosomes, and nucleus. These indicate that the specificity of ROS-mediated signal transduction may be modulated by the localization of Nox isoforms and their regulatory subunits within specific subcellular compartments. This review summarizes the recent progress on compartmentalization of redox signaling via activation of NADPH oxidase, which is implicated in cell biology and pathophysiologies.
Article
The survival curves of 694 human cell lines irradiated in exponentially growing phase in vitro were collected from the literature. Among them, 271 were derived from tumors, 423 were nontransformed fibroblasts and other normal cell strains from healthy people or people with some genetic disorders. Seventy-six different cell types are identified, and a specific radiosensitivity could be associated with each, using D and surviving fraction at 2 Gy. Technical factors such as culture medium, feeder cells, and scoring method were found to affect intrinsic radiosensitivity. In particular, the cell type is not a discriminating factor when cells are studied in agar. Results obtained with cells irradiated in agar must be used cautiously, depending on how the cells were prepared for the experiments. The use of feeder cells narrows the range of radiosensitivity of human cells. For cells irradiated as monolayer, it was possible to build a scale of radiosensitivity according to cell type, ranging, in terms of D from 0.6 Gy for the most sensitive cell lines to more than 4 Gy for the most resistant. Considering that, in most cases, we could estimate the variation of radiosensitivity within each cell type, our classification among cell types can be used by researchers to place their results in the context of the literature.
Article
Reactive oxygen species (ROS) are constantly generated and eliminated in the biological system, and play important roles in a variety of normal biochemical functions and abnormal pathological processes. Growing evidence suggests that cancer cells exhibit increased intrinsic ROS stress, due in part to oncogenic stimulation, increased metabolic activity, and mitochondrial malfunction. Since the mitochondrial respiratory chain (electron transport complexes) is a major source of ROS generation in the cells, the vulnerability of the mitochondrial DNA to ROS-mediated damage appears to be a mechanism to amplify ROS stress in cancer cells. The escalated ROS generation in cancer cells serves as an endogenous source of DNA-damaging agents that promote genetic instability and development of drug resistance. Malfunction of mitochondria also alters cellular apoptotic response to anticancer agents. Despite the negative impacts of increased ROS in cancer cells, it is possible to exploit this biochemical feature and develop novel therapeutic strategies to preferentially kill cancer cells through ROS-mediated mechanisms. This article reviews ROS stress in cancer cells, its underlying mechanisms and relationship with mitochondrial malfunction and alteration in drug sensitivity, and suggests new therapeutic strategies that take advantage of increased ROS in cancer cells to enhance therapeutic activity and selectivity.
Article
The high incidence of overexpression of some members of the Rho family of GTPases in human tumors suggests that (1) these proteins are involved in cancer onset, and (2) they are potential candidates for a therapeutic intervention. In recent years, the characterization of downstream effectors to Rho GTPases has provided crucial clues on the general cellular effects that permit aberrant proliferation and adhesiveness of tumor cells. The activation of many of these effector proteins in turn results in the modulation of the activity of several transcription factors that play an important role at various levels of Rho signaling. The precise mechanisms by which Rho GTPases participate in carcinogenesis are still not fully understood. However, it is becoming more evident that the specific role of Rho overexpression in tumor initiation, progression and metastasis, as well as the nature and cause of such overexpression in specific human tumors (i.e., transient or stable; tumor environment-regulated; genetic or epigenetic) may be linked to the activation of specific signaling pathways that result in transcriptional regulation. In this review, we summarize the functions of Rho proteins in the regulation of several transcription factors and their relationship to tumor biology.
Article
Hepatocellular carcinoma (HCC) is the third deadliest and fifth most common human cancer worldwide. Hepatitis C virus (HCV) and hepatitis B virus (HBV) infections along with alcohol and aflatoxin B1 intake are widely recognized etiological agents in HCCs. It is anticipated that HCCs will constitute a major health problem in the next two decades because of the rising incidence of HCV infections in the US. The poor survival rate achieved by current surgical procedures and chemotherapy treatment has prompted the scientific community to gain a better understanding of the molecular events involved in hepatocarcinogenesis in order to define new targets for more effective treatment. Recent findings from several laboratories have implicated constitutive activation of the transcription factor NF-kappaB as one of the early key events involved in neoplastic progression of the liver. Data is summarized here from recently published studies illustrating a crucial role of NF-kappaB in bridging the action of growth factors and inflammation to hepatic oncogenesis. Although additional work is needed to fully understand the precise role of NF-kappaB in the regulation of the various transitions of HCC development, these new findings raise the intriguing possibility that pharmacologic inhibition of NF-kappaB in the liver could selectively eradicate malignant liver cells without affecting normal liver homeostasis.
Article
The role for reactive oxygen species (ROS) in cellular (patho)physiology, in particular in signal transduction, is increasingly recognized. The family of NADPH oxidases (NOXes) plays an important role in the production of ROS in response to receptor agonists such as growth factors or inflammatory cytokines that signal through the Rho-like small GTPases Rac1 or Rac2. The phagocyte oxidase (gp91phox/NOX2) is the best characterized family member, and its mode of activation is relatively well understood. Recent work has uncovered novel and increasingly complex modes of control of the NOX2-related proteins. Some of these, including NOX2, have been implicated in various aspects of (cardio)vascular disease, including vascular smooth muscle and endothelial cell hypertrophy and proliferation, inflammation, and atherosclerosis. This review focuses on the role of the Rac1 and Rac2 GTPases in the activation of the various NOX family members.
Article
Phagocytic leukocytes generate reactive oxygen species important for the killing of invading microorganisms. The source of these oxidants is the NADPH oxidase, a tightly controlled multicomponent enzyme made up of a membrane-associated catalytic moiety and cytosolic regulatory components that must assemble to form the active oxidase. The phagocyte NADPH oxidase was the first mammalian system shown to be directly regulated by a Rac GTPase. We review here our understanding of NADPH oxidase regulation by Rac, as well as the regulation of Rac itself, in phagocytic leukocytes. Rather than viewing Rac as a "cog" in the NADPH oxidase machinery, we argue for a view of Rac GTPases as critical "molecular switches" regulating the formation of ROS by phagocytic leukocytes under physiologic and pathologic conditions.
Article
For a long time, superoxide generation by an NADPH oxidase was considered as an oddity only found in professional phagocytes. Over the last years, six homologs of the cytochrome subunit of the phagocyte NADPH oxidase were found: NOX1, NOX3, NOX4, NOX5, DUOX1, and DUOX2. Together with the phagocyte NADPH oxidase itself (NOX2/gp91(phox)), the homologs are now referred to as the NOX family of NADPH oxidases. These enzymes share the capacity to transport electrons across the plasma membrane and to generate superoxide and other downstream reactive oxygen species (ROS). Activation mechanisms and tissue distribution of the different members of the family are markedly different. The physiological functions of NOX family enzymes include host defense, posttranlational processing of proteins, cellular signaling, regulation of gene expression, and cell differentiation. NOX enzymes also contribute to a wide range of pathological processes. NOX deficiency may lead to immunosuppresion, lack of otoconogenesis, or hypothyroidism. Increased NOX activity also contributes to a large number or pathologies, in particular cardiovascular diseases and neurodegeneration. This review summarizes the current state of knowledge of the functions of NOX enzymes in physiology and pathology.