Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters










Publication year range
1.
Am J Cancer Res ; 11(10): 4668-4682, 2021.
Article in English | MEDLINE | ID: mdl-34765286

ABSTRACT

Despite the significant progress in cancer cure, the development of new approaches to cancer therapy is still of great importance since many deadly tumors remain untreatable. Boron neutron capture therapy (BNCT), proposed more than eighty years ago, is still considered a potentially advantageous approach. Irradiation of cells containing 10B isotopes with epithermal neutrons and the consequent decay of boron nuclei releases particles that deposit high energy along a very short path, inflicting heavy damage on the target cells but sparing the neighbouring tissue. Delivery and preferential accumulation of boron in cancer cells are the major obstacles that slow down the clinical use of BNCT. Since DNA damage caused by irradiation is the major reason for cell death, the incorporation of boron-containing nucleotides into the DNA of cancer cells may significantly increase the efficacy of BNCT. In this review, we discuss the current state of knowledge in the synthesis of boron-containing nucleosides and their application for BNCT with a special focus on their possible incorporation into genomic DNA.

2.
Nat Commun ; 12(1): 6959, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34845229

ABSTRACT

Efficient entry into S phase of the cell cycle is necessary for embryonic development and tissue homoeostasis. However, unscheduled S phase entry triggers DNA damage and promotes oncogenesis, underlining the requirement for strict control. Here, we identify the NUCKS1-SKP2-p21/p27 axis as a checkpoint pathway for the G1/S transition. In response to mitogenic stimulation, NUCKS1, a transcription factor, is recruited to chromatin to activate expression of SKP2, the F-box component of the SCFSKP2 ubiquitin ligase, leading to degradation of p21 and p27 and promoting progression into S phase. In contrast, DNA damage induces p53-dependent transcriptional repression of NUCKS1, leading to SKP2 downregulation, p21/p27 upregulation, and cell cycle arrest. We propose that the NUCKS1-SKP2-p21/p27 axis integrates mitogenic and DNA damage signalling to control S phase entry. The Cancer Genome Atlas (TCGA) data reveal that this mechanism is hijacked in many cancers, potentially allowing cancer cells to sustain uncontrolled proliferation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , S Phase/genetics , S-Phase Kinase-Associated Proteins/genetics , A549 Cells , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p27/metabolism , DNA Damage , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Osteoblasts/metabolism , Osteoblasts/pathology , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , S-Phase Kinase-Associated Proteins/antagonists & inhibitors , S-Phase Kinase-Associated Proteins/metabolism , Sf9 Cells , Signal Transduction , Spodoptera , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
3.
PLoS One ; 16(9): e0257473, 2021.
Article in English | MEDLINE | ID: mdl-34529719

ABSTRACT

The major human apurinic/apyrimidinic (AP) site endonuclease, APEX1, is a central player in the base excision DNA repair (BER) pathway and has a role in the regulation of DNA binding by transcription factors. In vertebrates, APEX1 knockouts are embryonic lethal, and only a handful of knockout cell lines are known. To facilitate studies of multiple functions of this protein in human cells, we have used the CRISPR/Cas9 system to knock out the APEX1 gene in a widely used non-cancer hypotriploid HEK 293FT cell line. Two stable knockout lines were obtained, one carrying two single-base deletion alleles and one single-base insertion allele in exon 3, another homozygous in the single-base insertion allele. Both mutations cause a frameshift that leads to premature translation termination before the start of the protein's catalytic domain. Both cell lines totally lacked the APEX1 protein and AP site-cleaving activity, and showed significantly lower levels of the APEX1 transcript. The APEX1-null cells were unable to support BER on uracil- or AP site-containing substrates. Phenotypically, they showed a moderately increased sensitivity to methyl methanesulfonate (MMS; ~2-fold lower EC50 compared with wild-type cells), and their background level of natural AP sites detected by the aldehyde-reactive probe was elevated ~1.5-2-fold. However, the knockout lines retained a nearly wild-type sensitivity to oxidizing agents hydrogen peroxide and potassium bromate. Interestingly, despite the increased MMS cytotoxicity, we observed no additional increase in AP sites in knockout cells upon MMS treatment, which could indicate their conversion into more toxic products in the absence of repair. Overall, the relatively mild cell phenotype in the absence of APEX1-dependent BER suggests that mammalian cells possess mechanisms of tolerance or alternative repair of AP sites. The knockout derivatives of the extensively characterized HEK 293FT cell line may provide a valuable tool for studies of APEX1 in DNA repair and beyond.


Subject(s)
DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , CRISPR-Cas Systems/genetics , Cell Cycle Checkpoints , DNA Repair/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Gene Editing , HEK293 Cells , Humans , Hydrogen Peroxide/chemistry , Methyl Methanesulfonate/pharmacology , Phenotype , RNA, Guide, Kinetoplastida/metabolism
4.
Nucleic Acids Res ; 48(22): 12483-12501, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33166394

ABSTRACT

Efficient S phase entry is essential for development, tissue repair, and immune defences. However, hyperactive or expedited S phase entry causes replication stress, DNA damage and oncogenesis, highlighting the need for strict regulation. Recent paradigm shifts and conflicting reports demonstrate the requirement for a discussion of the G1/S transition literature. Here, we review the recent studies, and propose a unified model for the S phase entry decision. In this model, competition between mitogen and DNA damage signalling over the course of the mother cell cycle constitutes the predominant control mechanism for S phase entry of daughter cells. Mitogens and DNA damage have distinct sensing periods, giving rise to three Commitment Points for S phase entry (CP1-3). S phase entry is mitogen-independent in the daughter G1 phase, but remains sensitive to DNA damage, such as single strand breaks, the most frequently-occurring lesions that uniquely threaten DNA replication. To control CP1-3, dedicated hubs integrate the antagonistic mitogenic and DNA damage signals, regulating the stoichiometric cyclin: CDK inhibitor ratio for ultrasensitive control of CDK4/6 and CDK2. This unified model for the G1/S cell cycle transition combines the findings of decades of study, and provides an updated foundation for cell cycle research.


Subject(s)
Cell Cycle Checkpoints/genetics , Cell Cycle/genetics , Cell Division/genetics , DNA Replication/genetics , DNA Damage/genetics , G1 Phase/genetics , Humans , S Phase/genetics , Signal Transduction/genetics
5.
FASEB J ; 34(8): 10443-10461, 2020 08.
Article in English | MEDLINE | ID: mdl-32539222

ABSTRACT

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is a key enzyme involved in energy metabolism. Recently, GAPDH has been suggested to have extraglycolytic functions in DNA repair, but the underlying mechanism for the GAPDH response to DNA damage remains unclear. Here, we demonstrate that the tyrosine kinase Src is activated under DNA damage stress and phosphorylates GAPDH at Tyr41. This phosphorylation of GAPDH is essential for its nuclear translocation and DNA repair function. Blocking the nuclear import of GAPDH by suppressing Src signaling or through a GAPDH Tyr41 mutation impairs its response to DNA damage. Nuclear GAPDH is recruited to DNA lesions and associates with DNA polymerase ß (Pol ß) to function in DNA repair. Nuclear GAPDH promotes Pol ß polymerase activity and increases base excision repair (BER) efficiency. Furthermore, GAPDH knockdown dramatically decreases BER efficiency and sensitizes cells to DNA damaging agents. Importantly, the knockdown of GAPDH in colon cancer SW480 cells and xenograft models effectively enhances their sensitivity to the chemotherapeutic drug 5-FU. In summary, our findings provide mechanistic insight into the new function of GAPDH in DNA repair and suggest a potential therapeutic target in chemotherapy.


Subject(s)
Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA Damage/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Phosphorylation/genetics , src-Family Kinases/metabolism , Active Transport, Cell Nucleus/genetics , Animals , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , DNA/genetics , DNA Polymerase beta/genetics , DNA Polymerase beta/metabolism , DNA Repair/genetics , Female , HEK293 Cells , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation/genetics , Protein Transport/genetics , Signal Transduction/genetics , src-Family Kinases/genetics
6.
NAR Cancer ; 2(2): zcaa004, 2020 Jun.
Article in English | MEDLINE | ID: mdl-34316684

ABSTRACT

Maintenance of genome stability suppresses cancer and other human diseases and is critical for organism survival. Inevitably, during a life span, multiple DNA lesions can arise due to the inherent instability of DNA molecules or due to endogenous or exogenous DNA damaging factors. To avoid malignant transformation of cells with damaged DNA, multiple mechanisms have evolved to repair DNA or to detect and eradicate cells accumulating unrepaired DNA damage. In this review, we discuss recent findings on the role of Sp1 (specificity factor 1) in the detection and elimination of cells accumulating persistent DNA strand breaks. We also discuss how this mechanism may contribute to the maintenance of physiological populations of healthy cells in an organism, thus preventing cancer formation, and the possible application of these findings in cancer therapy.

7.
DNA Repair (Amst) ; 86: 102740, 2020 02.
Article in English | MEDLINE | ID: mdl-31812125

ABSTRACT

Base excision repair (BER) is the major repair pathway that removes DNA single strand breaks (SSBs) arising spontaneously due to the inherent instability of DNA. Unrepaired SSBs promote cell-cycle delay, which facilitates DNA repair prior to replication. On the other hand, in response to persistent DNA strand breaks, ATM-dependent degradation of transcription factor Sp1 leads to downregulation of BER genes expression, further accumulation of SSBs and renders cells susceptible to elimination via apoptosis. In contrast, many cancer cells are not able to block replication and to downregulate the expression of Sp1 in response to DNA damage. However, knockdown of BER in cancer cells leads to the accumulation of DNA double strand breaks (DSBs), suggesting deficiency in non-homologous end joining (NHEJ) repair of DSBs. Here we investigated whether DNA repair deficiency caused by knockdown of the XRCC1 gene expression in proliferating cells results in downregulation of NHEJ genes expression. We find that knockdown of the XRCC1 gene expression does not cause degradation of Sp1, but leads to downregulation of Lig4/XRCC4 and Ku70/80 at the transcription and protein levels. We thus propose the existence of Sp1-independent backup mechanism that in response to BER deficiency downregulates NHEJ in proliferating cells.


Subject(s)
DNA End-Joining Repair , Down-Regulation , Sp1 Transcription Factor/metabolism , X-ray Repair Cross Complementing Protein 1/genetics , Cell Line , Cell Proliferation , DNA Breaks, Double-Stranded , DNA Repair , Gene Knockdown Techniques , Humans , Signal Transduction
8.
EMBO J ; 38(20): e101443, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31424118

ABSTRACT

Cyclins are central engines of cell cycle progression in conjunction with cyclin-dependent kinases (CDKs). Among the different cyclins controlling cell cycle progression, cyclin F does not partner with a CDK, but instead forms via its F-box domain an SCF (Skp1-Cul1-F-box)-type E3 ubiquitin ligase module. Although various substrates of cyclin F have been identified, the vulnerabilities of cells lacking cyclin F are not known. Thus, we assessed viability of cells lacking cyclin F upon challenging them with more than 180 different kinase inhibitors. The screen revealed a striking synthetic lethality between Chk1 inhibition and cyclin F loss. Chk1 inhibition in cells lacking cyclin F leads to DNA replication catastrophe. Replication catastrophe depends on accumulation of the transcription factor E2F1 in cyclin F-depleted cells. We find that SCF-cyclin F controls E2F1 ubiquitylation and degradation during the G2/M phase of the cell cycle and upon challenging cells with Chk1 inhibitors. Thus, Cyclin F restricts E2F1 activity during the cell cycle and upon checkpoint inhibition to prevent DNA replication stress. Our findings pave the way for patient selection in the clinical use of checkpoint inhibitors.


Subject(s)
Checkpoint Kinase 1/antagonists & inhibitors , Cyclins/metabolism , E2F1 Transcription Factor/metabolism , Protein Kinase Inhibitors/pharmacology , Proteolysis , SKP Cullin F-Box Protein Ligases/metabolism , Synthetic Lethal Mutations , Cell Cycle/drug effects , Checkpoint Kinase 1/genetics , Cyclins/genetics , DNA Replication , E2F1 Transcription Factor/genetics , HeLa Cells , Humans , Phosphorylation , Protein Binding , SKP Cullin F-Box Protein Ligases/genetics , Ubiquitination
9.
FASEB J ; 33(11): 11668-11681, 2019 11.
Article in English | MEDLINE | ID: mdl-31348687

ABSTRACT

Multiple DNA repair pathways may be involved in the removal of the same DNA lesion caused by endogenous or exogenous agents. Although distinct DNA repair machinery fulfill overlapping roles in the repair of DNA lesions, the mechanisms coordinating different pathways have not been investigated in detail. Here, we show that Ku70, a core protein of nonhomologous end-joining (NHEJ) repair pathway, can directly interact with DNA polymerase-ß (Pol-ß), a central player in the DNA base excision repair (BER), and this physical complex not only promotes the polymerase activity of Pol-ß and BER efficiency but also enhances the classic NHEJ repair. Moreover, we find that DNA damages caused by methyl methanesulfonate (MMS) or etoposide promote the formation of Ku70-Pol-ß complexes at the repair foci. Furthermore, suppression of endogenous Ku70 expression by small interfering RNA reduces BER efficiency and leads to higher sensitivity to MMS and accumulation of the DNA strand breaks. Similarly, Pol-ß knockdown impairs total-NHEJ capacity but only has a slight influence on alternative NHEJ. These results suggest that Pol-ß and Ku70 coordinate 2-way crosstalk between the BER and NHEJ pathways.-Xia, W., Ci, S., Li, M., Wang, M., Dianov, G. L., Ma, Z., Li, L., Hua, K., Alagamuthu, K. K., Qing, L., Luo, L., Edick, A. M., Liu, L., Hu, Z., He, L., Pan, F., Guo, Z. Two-way crosstalk between BER and c-NHEJ repair pathway is mediated by Pol-ß and Ku70.


Subject(s)
DNA Damage/genetics , DNA Repair/genetics , DNA Replication/genetics , Ku Autoantigen/metabolism , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Polymerase beta/genetics , DNA-Binding Proteins/metabolism , Humans
10.
Cell Oncol (Dordr) ; 41(5): 527-539, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30088263

ABSTRACT

BACKGROUND: To deliver efficacious personalised cancer treatment, it is essential to characterise the cellular metabolism as well as the genetic stability of individual tumours. In this study, we describe a new axis between DNA repair and detoxification of aldehyde derivatives with important implications for patient prognosis and treatment. METHODS: Western blot and qPCR analyses were performed in relevant non-transformed and cancer cell lines from lung and liver tissue origin in combination with bioinformatics data mining of The Cancer Genome Atlas database from lung and hepatocellular cancer patients. RESULTS: Using both biochemical and bioinformatics approaches, we revealed an association between the levels of expression of the aldehyde detoxifying enzyme aldehyde dehydrogenase 2 (ALDH2) and the key DNA base excision repair protein XRCC1. Across cancer types, we found that if one of the corresponding genes exhibits a low expression level, the level of the other gene is increased. Surprisingly, we found that low ALDH2 expression levels associated with high XRCC1 expression levels are indicative for a poor overall survival, particularly in lung and liver cancer patients. In addition, we found that Mithramycin A, a XRCC1 expression inhibitor, efficiently kills cancer cells expressing low levels of ALDH2. CONCLUSIONS: Our data suggest that lung and liver cancers require efficient single-strand break repair for their growth in order to benefit from a low aldehyde detoxification metabolism. We also propose that the ratio of XRCC1 and ALDH2 levels may serve as a useful prognostic tool in these cancer types.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , X-ray Repair Cross Complementing Protein 1/metabolism , Aldehyde Dehydrogenase, Mitochondrial/genetics , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , DNA Damage/genetics , DNA Damage/physiology , Humans , Liver Neoplasms/genetics , Lung Neoplasms/genetics , Plicamycin/analogs & derivatives , Plicamycin/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , X-ray Repair Cross Complementing Protein 1/antagonists & inhibitors , X-ray Repair Cross Complementing Protein 1/genetics
11.
Sci Rep ; 8(1): 6010, 2018 Apr 12.
Article in English | MEDLINE | ID: mdl-29651030

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

12.
Oncotarget ; 9(17): 13666-13681, 2018 Mar 02.
Article in English | MEDLINE | ID: mdl-29568385

ABSTRACT

Cancer-associated fibroblasts (CAFs) are an emerging target for cancer therapy as they promote tumour growth and metastatic potential. However, CAF targeting is complicated by the lack of knowledge-based strategies aiming to selectively eliminate these cells. There is a growing body of evidence suggesting that a pro-inflammatory microenvironment (e.g. ROS and cytokines) promotes CAF formation during tumorigenesis, although the exact mechanisms involved remain unclear. In this study, we reveal that a prolonged pro-inflammatory stimulation causes a de facto deficiency in base excision repair, generating unrepaired DNA strand breaks and thereby triggering an ATF4-dependent reprogramming of normal fibroblasts into CAF-like cells. Based on the phenotype of in vitro-generated CAFs, we demonstrate that midostaurin, a clinically relevant compound, selectively eliminates CAF-like cells deficient in base excision repair and prevents their stimulatory role in cancer cell growth and migration.

13.
Nucleic Acids Res ; 46(4): 1834-1846, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29294106

ABSTRACT

ATM (ataxia-telangiectasia mutated) is a central molecule for DNA quality control. Its activation by DNA damage promotes cell-cycle delay, which facilitates DNA repair prior to replication. On the other hand, persistent DNA damage has been implicated in ATM-dependent cell death via apoptosis; however, the mechanisms underlying this process remain elusive. Here we find that, in response to persistent DNA strand breaks, ATM phosphorylates transcription factor Sp1 and initiates its degradation. We show that Sp1 controls expression of the key base excision repair gene XRCC1, essential for DNA strand break repair. Therefore, degradation of Sp1 leads to a vicious cycle that involves suppression of DNA repair and further aggravation of the load of DNA damage. This activates transcription of pro-apoptotic genes and renders cells susceptible to elimination via both apoptosis and natural killer cells. These findings constitute a previously unrecognized 'gatekeeper' function of ATM as a detector of cells with persistent DNA damage.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Repair , Sp1 Transcription Factor/metabolism , Apoptosis , Cells, Cultured , DNA Damage , Down-Regulation , Humans , Killer Cells, Natural/physiology , Male , Phosphorylation , Serine/metabolism , Sp1 Transcription Factor/chemistry , X-ray Repair Cross Complementing Protein 1/biosynthesis , X-ray Repair Cross Complementing Protein 1/genetics
14.
Sci Rep ; 7(1): 15050, 2017 11 08.
Article in English | MEDLINE | ID: mdl-29118367

ABSTRACT

Mutations in the HECT, UBA and WWE domain-containing 1 (HUWE1) E3 ubiquitin ligase cause neurodevelopmental disorder X-linked intellectual disability (XLID). HUWE1 regulates essential processes such as genome integrity maintenance. Alterations in the genome integrity and accumulation of mutations have been tightly associated with the onset of neurodevelopmental disorders. Though HUWE1 mutations are clearly implicated in XLID and HUWE1 regulatory functions well explored, currently much is unknown about the molecular basis of HUWE1-promoted XLID. Here we showed that the HUWE1 expression is altered and mutation frequency increased in three different XLID individual (HUWE1 p.R2981H, p.R4187C and HUWE1 duplication) cell lines. The effect was most prominent in HUWE1 p.R4187C XLID cells and was accompanied with decreased DNA repair capacity and hypersensitivity to oxidative stress. Analysis of HUWE1 substrates revealed XLID-specific down-regulation of oxidative stress response DNA polymerase (Pol) λ caused by hyperactive HUWE1 p.R4187C. The subsequent restoration of Polλ levels counteracted the oxidative hypersensitivity. The observed alterations in the genome integrity maintenance may be particularly relevant in the cortical progenitor zones of human brain, as suggested by HUWE1 immunofluorescence analysis of cerebral organoids. These results provide evidence that impairments of the fundamental cellular processes, like genome integrity maintenance, characterize HUWE1-promoted XLID.


Subject(s)
Genes, X-Linked , Intellectual Disability/genetics , Oxidative Stress , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Cell Line , DNA Polymerase beta/metabolism , DNA Repair/genetics , Genomic Instability/genetics , Humans , Intellectual Disability/pathology , Mutation
15.
Nucleic Acids Res ; 45(17): 10042-10055, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28973444

ABSTRACT

Ataxia telangiectasia (A-T) is a syndrome associated with loss of ATM protein function. Neurodegeneration and cancer predisposition, both hallmarks of A-T, are likely to emerge as a consequence of the persistent oxidative stress and DNA damage observed in this disease. Surprisingly however, despite these severe features, a lack of functional ATM is still compatible with early life, suggesting that adaptation mechanisms contributing to cell survival must be in place. Here we address this gap in our knowledge by analysing the process of human fibroblast adaptation to the lack of ATM. We identify profound rearrangement in cellular proteostasis occurring very early on after loss of ATM in order to counter protein damage originating from oxidative stress. Change in proteostasis, however, is not without repercussions. Modulating protein turnover in ATM-depleted cells also has an adverse effect on the DNA base excision repair pathway, the major DNA repair system that deals with oxidative DNA damage. As a consequence, the burden of unrepaired endogenous DNA lesions intensifies, progressively leading to genomic instability. Our study provides a glimpse at the cellular consequences of loss of ATM and highlights a previously overlooked role for proteostasis in maintaining cell survival in the absence of ATM function.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/deficiency , DNA Repair/physiology , Ataxia Telangiectasia/enzymology , Ataxia Telangiectasia/pathology , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Survival , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/enzymology , Humans , Molecular Chaperones/metabolism , Oxidation-Reduction , Oxidative Stress , Proteasome Endopeptidase Complex/metabolism , Protein Biosynthesis , Proteostasis Deficiencies , RNA Interference , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism , Unfolded Protein Response
16.
Curr Pharm Des ; 23(32): 4758-4772, 2017.
Article in English | MEDLINE | ID: mdl-28699540

ABSTRACT

Our current understanding of cancer suggests that every tumour has individual features. Approaches to cancer treatment require thorough comprehension of the mechanisms triggering genomic instability and protecting cancer cells from therapeutic treatments. Base excision repair (BER) is a frontline DNA repair system that is responsible for maintaining genome integrity. The BER pathway prevents the occurrence of disease, including cancer, by constantly repairing DNA base lesions and DNA single strand breaks caused by endogenous and exogenous mutagens. BER is an important DNA repair system for cancer cell survival, as it can affect both chemoand radio-resistance of tumours. Variations in BER capacity are likely responsible for a number of cases of sporadic cancer and may also modulate cancer sensitivity and resistance to therapeutic treatments. For these reasons, it is broadly accepted that targeting BER enzymes might be a promising approach to personalised anti-cancer therapy. However, recent advances in both treatment strategies and the comprehension of cancer development call for a better understanding of the consequences of BER inhibition. Indeed, the impact on both the tumour microenvironment and healthy tissues is still unclear. This review will summarise the current status of the approaches exploiting BER targeting, describing the most promising small molecule inhibitors and synthetic lethality strategies, as well as potential limitations of these approaches.


Subject(s)
DNA Repair/genetics , Neoplasms/pathology , Tumor Microenvironment/genetics , Animals , Cell Survival/genetics , DNA Breaks, Single-Stranded , DNA Damage/genetics , Genomic Instability/genetics , Humans , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/therapy
17.
Int J Mol Sci ; 17(6)2016 Jun 01.
Article in English | MEDLINE | ID: mdl-27258260

ABSTRACT

Schizophrenia and autism spectrum disorder (ASD) are multi-factorial and multi-symptomatic psychiatric disorders, each affecting 0.5%-1% of the population worldwide. Both are characterized by impairments in cognitive functions, emotions and behaviour, and they undermine basic human processes of perception and judgment. Despite decades of extensive research, the aetiologies of schizophrenia and ASD are still poorly understood and remain a significant challenge to clinicians and scientists alike. Adding to this unsatisfactory situation, patients with schizophrenia or ASD often develop a variety of peripheral and systemic disturbances, one prominent example of which is cancer, which shows a direct (but sometimes inverse) comorbidity in people affected with schizophrenia and ASD. Cancer is a disease characterized by uncontrolled proliferation of cells, the molecular origin of which derives from mutations of a cell's DNA sequence. To counteract such mutations and repair damaged DNA, cells are equipped with intricate DNA repair pathways. Oxidative stress, oxidative DNA damage, and deficient repair of oxidative DNA lesions repair have been proposed to contribute to the development of schizophrenia and ASD. In this article, we summarize the current evidence of cancer comorbidity in these brain disorders and discuss the putative roles of oxidative stress, DNA damage and DNA repair in the aetiopathology of schizophrenia and ASD.


Subject(s)
Autistic Disorder/genetics , DNA Damage , DNA Repair , Schizophrenia/genetics , Animals , Autism Spectrum Disorder/diagnosis , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/genetics , Autistic Disorder/diagnosis , Autistic Disorder/etiology , Autistic Disorder/metabolism , Comorbidity , Humans , Neoplasms/etiology , Neoplasms/metabolism , Oxidative Stress , Risk , Schizophrenia/diagnosis , Schizophrenia/etiology , Schizophrenia/metabolism
18.
Nucleic Acids Res ; 44(7): 3165-75, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-26773055

ABSTRACT

DNA constantly undergoes chemical modification due to endogenous and exogenous mutagens. The DNA base excision repair (BER) pathway is the frontline mechanism handling the majority of these lesions, and primarily involves a DNA incision and subsequent resealing step. It is imperative that these processes are extremely well-coordinated as unrepaired DNA single strand breaks (SSBs) can be converted to DNA double strand breaks during replication thus triggering genomic instability. However, the mechanism(s) governing the BER process are poorly understood. Here we show that accumulation of unrepaired SSBs triggers a p53/Sp1-dependent downregulation of APE1, the endonuclease responsible for the DNA incision during BER. Importantly, we demonstrate that impaired p53 function, a characteristic of many cancers, leads to a failure of the BER coordination mechanism, overexpression of APE1, accumulation of DNA strand breaks and results in genomic instability. Our data provide evidence for a previously unrecognized mechanism for coordination of BER by p53, and its dysfunction in p53-inactivated cells.


Subject(s)
DNA Repair , Genomic Instability , Tumor Suppressor Protein p53/metabolism , Cells, Cultured , DNA Breaks, Single-Stranded , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Down-Regulation , Humans , Sp1 Transcription Factor/metabolism
19.
Cancer Cell ; 28(5): 557-568, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26602815

ABSTRACT

Histone H3K36 trimethylation (H3K36me3) is frequently lost in multiple cancer types, identifying it as an important therapeutic target. Here we identify a synthetic lethal interaction in which H3K36me3-deficient cancers are acutely sensitive to WEE1 inhibition. We show that RRM2, a ribonucleotide reductase subunit, is the target of this synthetic lethal interaction. RRM2 is regulated by two pathways here: first, H3K36me3 facilitates RRM2 expression through transcription initiation factor recruitment; second, WEE1 inhibition degrades RRM2 through untimely CDK activation. Therefore, WEE1 inhibition in H3K36me3-deficient cells results in RRM2 reduction, critical dNTP depletion, S-phase arrest, and apoptosis. Accordingly, this synthetic lethality is suppressed by increasing RRM2 expression or inhibiting RRM2 degradation. Finally, we demonstrate that WEE1 inhibitor AZD1775 regresses H3K36me3-deficient tumor xenografts.


Subject(s)
Cell Cycle Proteins/metabolism , Histones/metabolism , Neoplasms/metabolism , Nuclear Proteins/metabolism , Nucleotides/metabolism , Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Gene Expression Regulation, Neoplastic/drug effects , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Humans , Lysine/genetics , Lysine/metabolism , Methylation/drug effects , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Neoplasms/genetics , Neoplasms/prevention & control , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nucleotides/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrimidinones , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleoside Diphosphate Reductase/metabolism , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Xenograft Model Antitumor Assays
20.
Nucleic Acids Res ; 43(7): 3667-79, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25800737

ABSTRACT

Genetic instability, provoked by exogenous mutagens, is well linked to initiation of cancer. However, even in unstressed cells, DNA undergoes a plethora of spontaneous alterations provoked by its inherent chemical instability and the intracellular milieu. Base excision repair (BER) is the major cellular pathway responsible for repair of these lesions, and as deficiency in BER activity results in DNA damage it has been proposed that it may trigger the development of sporadic cancers. Nevertheless, experimental evidence for this model remains inconsistent and elusive. Here, we performed a proteomic analysis of BER deficient human cells using stable isotope labelling with amino acids in cell culture (SILAC), and demonstrate that BER deficiency, which induces genetic instability, results in dramatic changes in gene expression, resembling changes found in many cancers. We observed profound alterations in tissue homeostasis, serine biosynthesis, and one-carbon- and amino acid metabolism, all of which have been identified as cancer cell 'hallmarks'. For the first time, this study describes gene expression changes characteristic for cells deficient in repair of endogenous DNA lesions by BER. These expression changes resemble those observed in cancer cells, suggesting that genetically unstable BER deficient cells may be a source of pre-cancerous cells.


Subject(s)
DNA Repair , Genomic Instability , Neoplasms/genetics , Amino Acids/biosynthesis , Amino Acids/metabolism , Base Sequence , Cell Line , Comet Assay , DNA Primers , DNA-Binding Proteins/genetics , Energy Metabolism , Humans , Real-Time Polymerase Chain Reaction , X-ray Repair Cross Complementing Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...