Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
DNA Repair (Amst) ; 100: 103070, 2021 04.
Article in English | MEDLINE | ID: mdl-33618126

ABSTRACT

The classical small Rho GTPase (Rho, Rac, and Cdc42) protein family is mainly responsible for regulating cell motility and polarity, membrane trafficking, cell cycle control, and gene transcription. Cumulative recent evidence supports important roles for these proteins in the maintenance of genomic stability. Indeed, DNA damage response (DDR) and repair mechanisms are some of the prime biological processes that underlie several disease phenotypes, including genetic disorders, cancer, senescence, and premature aging. Many reports guided by different experimental approaches and molecular hypotheses have demonstrated that, to some extent, direct modulation of Rho GTPase activity, their downstream effectors, or actin cytoskeleton regulation contribute to these cellular events. Although much attention has been paid to this family in the context of canonical actin cytoskeleton remodeling, here we provide a contextualized review of the interplay between Rho GTPase signaling pathways and the DDR and DNA repair signaling components. Interesting questions yet to be addressed relate to the spatiotemporal dynamics of this collective response and whether it correlates with different subcellular pools of Rho GTPases. We highlight the direct and indirect targets, some of which still lack experimental validation data, likely associated with Rho GTPase activation that provides compelling evidence for further investigation in DNA damage-associated events and with potential therapeutic applications in translational medicine.


Subject(s)
Actin Cytoskeleton/metabolism , DNA Damage , DNA Repair , Genomic Instability , Signal Transduction , rho GTP-Binding Proteins/metabolism , Animals , Humans
2.
Front Cell Dev Biol ; 8: 816, 2020.
Article in English | MEDLINE | ID: mdl-33015036

ABSTRACT

Typical Rho GTPases include the enzymes RhoA, Rac1, and Cdc42 that act as molecular switches to regulate essential cellular processes in eukaryotic cells such as actomyosin dynamics, cell cycle, adhesion, death and differentiation. Recently, it has been shown that different conditions modulate the activity of these enzymes, but their functions still need to be better understood. Here we examine the interplay between RhoA and the NER (Nucleotide Excision Repair) pathway in human cells exposed to UVA, UVB or UVC radiation. The results show high levels and accumulation of UV-induced DNA lesions (strand breaks and cyclobutane pyrimidine dimers, CPDs) in different cells with RhoA loss of function (LoF), either by stable overexpression of negative dominant RhoA (RhoA-N19 mutant), by inhibition with C3 toxin or by transient silencing with siRNA. Cells under RhoA LoF showed reduced levels of γH2AX, p-Chk1 (Ser345) and p-p53 (Ser15) that reflected causally in their accumulation in G1/S phases, in low survival rates and in reduced cell proliferation, also in accordance with the energy of applied UV light. Even NER-deficient cells (XPA, XPC) or DNA translesion synthesis (TLS)-deficient cells (XPV) showed substantial hypersensitivity to UV effects when previously submitted to RhoA LoF. In contrast, analyses of apoptosis, necrosis, autophagy and senescence revealed that all cells displaying normal levels of active RhoA (RhoA-GTP) are more resistant to UV-promoted cell death. This work reaffirms the role of RhoA protein signaling in protecting cells from damage caused by UV radiation and demonstrates relevant communicating mechanisms between actin cytoskeleton and genomic stability.

3.
J Photochem Photobiol B ; 209: 111947, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32652466

ABSTRACT

Actin cytoskeleton remodeling is the major motor of cytoskeleton dynamics driving tumor cell adhesion, migration and invasion. The typical RhoA, RhoB and RhoC GTPases are the main regulators of actin cytoskeleton dynamics. The C3 exoenzyme transferase from Clostridium botulinum is a toxin that causes the specific ADP-ribosylation of Rho-like proteins, leading to its inactivation. Here, we examine what effects the Rho GTPase inhibition and the consequent actin cytoskeleton instability would have on the emergence of DNA damage and on the recovery of genomic stability of malignant melanoma cells, as well as on their survival. Therefore, the MeWo cell line, here assumed as a melanoma cell line model for the expression of genes involved in the regulation of the actin cytoskeleton, was transiently transfected with the C3 toxin and subsequently exposed to UV-radiation. Phalloidin staining of the stress fibers revealed that actin cytoskeleton integrity was strongly disrupted by the C3 toxin in association with reduced melanoma cells survival, and further enhanced the deleterious effects of UV light. MeWo cells with actin cytoskeleton previously perturbed by the C3 toxin still showed higher levels and accumulation of UV-damaged DNA (strand breaks and cyclobutane pyrimidine dimers, CPDs). The interplay between reduced cell survival and impaired DNA repair upon actin cytoskeleton disruption can be explained by constitutive ERK1/2 activation and an inefficient phosphorylation of DDR proteins (γH2AX, CHK1 and p53) caused by C3 toxin treatment. Altogether, these results support the general idea that actin network help to protect the genome of human cells from damage caused by UV light through unknown molecular mechanisms that tie the cytoskeleton to processes of genomic stability maintenance.


Subject(s)
ADP Ribose Transferases/metabolism , Botulinum Toxins/metabolism , Cell Survival/radiation effects , Genomic Instability , Melanoma/metabolism , Skin Neoplasms/metabolism , Ultraviolet Rays , Actin Cytoskeleton/metabolism , Cell Line, Tumor , Humans , Melanoma/genetics , Melanoma/pathology , Mutation , Skin Neoplasms/genetics , Skin Neoplasms/pathology
4.
Methods Mol Biol ; 1821: 339-355, 2018.
Article in English | MEDLINE | ID: mdl-30062423

ABSTRACT

Actin polymerization, actomyosin ring contraction, and stress fiber formation are examples of relevant actions of the RhoA/B/C pathway as GTPases that regulate the cytoskeleton. However, open questions that remain to be addressed are whether this pathway and/or downstream components protect against or facilitate the formation of DNA double-strand breaks, the most lethal form of DNA damage in cells. Genotoxic drugs are radiomimetic and/or chemotherapeutic agents that are currently used for cancer treatments and are associated with specific methodologies; thus, these compounds should represent good tools to answer these questions. In this chapter, we describe two methods, the alkaline comet assay and homologous/nonhomologous recombination assays, to investigate the mechanism by which the Rho pathway modulates the repair of DNA breaks in tumor epithelial cell lines.


Subject(s)
Comet Assay/methods , DNA Breaks, Double-Stranded , DNA, Neoplasm/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Recombinational DNA Repair , rho GTP-Binding Proteins/metabolism , HeLa Cells , Humans , Neoplasms, Glandular and Epithelial/pathology
5.
Biochim Biophys Acta Gen Subj ; 1861(7): 1879-1894, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28389334

ABSTRACT

BACKGROUND: Radiotherapy causes the regression of many human tumors by increasing DNA damage, and the novel molecular mechanisms underlying the genomic instability leading to cancer progression and metastasis must be elucidated. Atypical dual-specificity phosphatase 3 (DUSP3) has been shown to down-regulate mitogen-activated protein kinases (MAPKs) to control the proliferation and apoptosis of human cancer cells. We have recently identified novel molecular targets of DUSP3 that function in DNA damage response and repair; however, whether DUSP3 affects these processes remains unknown. METHODS: Tumor cell lines in which DUSP3 activity was suppressed by pharmacological inhibitors or a targeted siRNA were exposed to gamma radiation, and proliferation, survival, DNA strand breaks and recombination repair pathways were sequentially analyzed. RESULTS: The combination of reduced DUSP3 activity and gamma irradiation resulted in decreased cellular proliferation and survival and increased cellular senescence compared with the effects of radiation exposure alone. Gamma radiation-induced DNA damage was increased by the loss of DUSP3 activity and correlated with increased levels of phospho-H2AX protein and numbers of ionizing radiation-induced γ-H2AX foci, which were reflected in diminished efficiencies of homologous recombination (HR) and non-homologous end-joining (NHEJ) repair. Similar results were obtained in ATM-deficient cells, in which reduced DUSP3 activity increased radiosensitivity, independent of increased MAPK phosphorylation. CONCLUSION: The loss of DUSP3 activity markedly increases gamma radiation-induced DNA strand breaks, suggesting a potential novel role for DUSP3 in DNA repair. GENERAL SIGNIFICANCE: The radioresistance of tumor cells is effectively reduced by a combination of approaches through the inhibition of DUSPs.


Subject(s)
DNA Repair , Dual Specificity Phosphatase 3/physiology , Neoplasms/radiotherapy , Radiation Tolerance , Ataxia Telangiectasia Mutated Proteins/physiology , Cell Line, Tumor , DNA Damage , Dual Specificity Phosphatase 3/antagonists & inhibitors , Gamma Rays , Histones/analysis , Humans , Mitogen-Activated Protein Kinases/metabolism
6.
Oxid Med Cell Longev ; 2016: 6012642, 2016.
Article in English | MEDLINE | ID: mdl-26649141

ABSTRACT

Radiotherapy with γ-radiation is widely used in cancer treatment to induce DNA damage reducing cell proliferation and to kill tumor cells. Although RhoA GTPase overexpression/hyperactivation is observed in many malignancies, the effect of RhoA activity modulation on cancer radiosensitivity has not been previously investigated. Here, we generated stable HeLa cell clones expressing either the dominant negative RhoA-N19 or the constitutively active RhoA-V14 and compared the responses of these cell lines with those of parental HeLa cells, after treatment with low doses of γ-radiation. HeLa-RhoA-N19 and HeLa-RhoA-V14 clones displayed reduced proliferation and survival compared to parental cells after radiation and became arrested at cell cycle stages correlated with increased cellular senescence and apoptosis. Also, Chk1/Chk2 and histone H2A phosphorylation data, as well as comet assays, suggest that the levels of DNA damage and DNA repair activation and efficiency in HeLa cell lines are correlated with active RhoA. In agreement with these results, RhoA inhibition by C3 toxin expression drastically affected homologous recombination (HR) and nonhomologous end joining (NHEJ). These data suggest that modulation of RhoA GTPase activity impairs DNA damage repair, increasing HeLa cell radiosensitivity.


Subject(s)
DNA Repair/radiation effects , Gamma Rays , Neoplasm Proteins/metabolism , Uterine Cervical Neoplasms/metabolism , rhoA GTP-Binding Protein/metabolism , ADP Ribose Transferases/pharmacology , Botulinum Toxins/pharmacology , Checkpoint Kinase 1 , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , DNA Repair/drug effects , DNA Repair/genetics , Female , HeLa Cells , Humans , Mutation , Neoplasm Proteins/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , rhoA GTP-Binding Protein/genetics
7.
J Cell Biochem ; 116(9): 2086-97, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25780896

ABSTRACT

Cell division control protein 42 (CDC42) homolog is a small Rho GTPase enzyme that participates in such processes as cell cycle progression, migration, polarity, adhesion, and transcription. Recent studies suggest that CDC42 is a potent tumor suppressor in different tissues and is related to aging processes. Although DNA damage is crucial in aging, a potential role for CDC42 in genotoxic stress remains to be explored. Migration, survival/proliferation and DNA damage/repair experiments were performed to demonstrate CDC42 involvement in the recovery of HeLa cells exposed to ultraviolet radiation-induced stress. Sub-lines of HeLa cells ectopically expressing the constitutively active CDC42-V12 mutant were generated to examine whether different CDC42-GTP backgrounds might reflect different sensitivities to UV radiation. Our results show that CDC42 constitutive activation does not interfere with HeLa cell migration after UV radiation. However, the minor DNA damage exhibited by the CDC42-V12 mutant exposed to UV radiation most likely results in cell cycle arrest at the G2/M checkpoint and reduced proliferation and survival. HeLa cells and Mock clones, which express endogenous wild-type CDC42 and show normal activity, are more resistant to UV radiation. None of these effects are altered by pharmacological CDC42 inhibition. Finally, the phosphorylation status of the DNA damage response proteins γ-H2AX and p-Chk1 was found to be delayed and attenuated, respectively, in CDC42-V12 clones. In conclusion, the sensitivity of HeLa cells to ultraviolet radiation increases with CDC42 over-activation due to inadequate DNA repair signaling, culminating in G2/M cell accumulation, which is translated into reduced cellular proliferation and survival.


Subject(s)
Cell Proliferation/radiation effects , DNA Repair , Ultraviolet Rays/adverse effects , cdc42 GTP-Binding Protein/metabolism , Cell Cycle Checkpoints/radiation effects , Cell Movement/radiation effects , Cell Survival/radiation effects , HeLa Cells , Humans , Mutation , Radiation Tolerance , cdc42 GTP-Binding Protein/genetics
8.
Mol Cell Biochem ; 404(1-2): 281-97, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25758356

ABSTRACT

Rac1 GTPase controls essential cellular functions related to the cytoskeleton, such as motility and adhesion. Rac1 is overexpressed in many tumor cells, including breast cancers, where it is also involved in the proliferation and checkpoint control necessary for the cell's recovery after exposure to ionizing radiation. However, its role in DNA damage and repair remains obscure in other tumor cells and under different genotoxic conditions. Here, we compare HeLa cells with mutants exogenously expressing a dominant-negative Rac1 (HeLa-Rac1-N17) by their responses to DNA damage induced by gamma or UV radiation. In HeLa cells, these treatments led to increased levels of active Rac1 (Rac1-GTP) and of stress fibers, with a diminished ability to migrate compared to untreated cells. However, the reduction of Rac1-GTP in Rac1-N17-deficient clones resulted in much higher levels of polymerized stress fibers accompanied by a strong impairment of cell migration, even after both radiation treatments. With regard to proliferation and genomic stability, dominant-negative Rac1 cells were more sensitive to gamma and UV radiation, exhibiting reduced proliferation and survival consistent with increased DNA damage and delayed or reduced DNA repair observed in this Rac1-deficient clone. The DNA damage response, as indicated by pH2AX and pChk1 levels, was increased in HeLa cells but was not effectively triggered in the Rac1-N17 clone after radiation treatment, which is likely the main cause of DNA damage accumulation. These data suggest that Rac1 GTPase plays an important role in signaling and contributes to the sensitivity of cervical cancer cells under UV or gamma radiation treatments.


Subject(s)
Cell Proliferation/genetics , DNA Repair/genetics , Signal Transduction/genetics , rac1 GTP-Binding Protein/genetics , Cell Proliferation/radiation effects , Cell Survival/genetics , Cell Survival/radiation effects , Gamma Rays , HeLa Cells , Humans , Signal Transduction/radiation effects , Ultraviolet Rays , rac1 GTP-Binding Protein/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...