Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Nitric Oxide ; 128: 25-36, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35970264

ABSTRACT

Photodynamic therapy (PDT) is a therapeutic modality based on the simultaneous action of three elements: photosensitizer, light and oxygen. This triad generates singlet oxygen and reactive oxygen species that can reduce the mass of a tumor. PDT is also able to stimulate iNOS, the enzyme that generates nitric oxide (NO). The role of NO in PDT-treated cancer cells has been investigated in several studies. They showed that low iNOS/NO levels stimulate signaling pathways that promote tumor survival, while high iNOS/NO levels arrest tumor growth. There is increasing evidence that ROS/RNS control both proliferation and migration of cells in the vicinity of PDT-treated tumor cells (so-called bystander cells). In this work, we addressed the question of how NO, which is generated by weak PDT, affects bystander cells. We used a conditioned medium: medium of PDT-treated tumor cells containing the stressors produced by the cells was added to untreated cells mimicking the neighboring bystander cells to investigate whether the conditioned medium affects cell proliferation. We found that low-level NO in prostate cancer cells affects the bystander tumor cells in a manner that depends on their malignancy grade.


Subject(s)
Photochemotherapy , Prostatic Neoplasms , Bystander Effect , Cell Line, Tumor , Cell Survival , Culture Media, Conditioned/pharmacology , Humans , Male , Nitric Oxide/metabolism , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Prostatic Neoplasms/drug therapy , Reactive Oxygen Species/metabolism
2.
Nucleic Acids Res ; 48(16): 9336-9345, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32432667

ABSTRACT

KRAS is one of the most mutated oncogenes and still considered an undruggable target. An alternative strategy would consist in targeting its gene rather than the protein, specifically the formation of G-quadruplexes (G4) in its promoter. G4 are secondary structures implicated in biological processes, which can be formed among G-rich DNA (or RNA) sequences. Here we have studied the major conformations of the commonly known KRAS 32R, or simply 32R, a 32 residue sequence within the KRAS Nuclease Hypersensitive Element (NHE) region. We have determined the structure of the two major stable conformers that 32R can adopt and which display slow equilibrium (>ms) with each other. By using different biophysical methods, we found that the nucleotides G9, G25, G28 and G32 are particularly implicated in the exchange between these two conformations. We also showed that a triad at the 3' end further stabilizes one of the G4 conformations, while the second conformer remains more flexible and less stable.


Subject(s)
DNA/genetics , G-Quadruplexes , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Binding Sites/genetics , Circular Dichroism , Humans , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/therapy , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors
3.
Int J Mol Sci ; 22(3)2021 Jan 24.
Article in English | MEDLINE | ID: mdl-33498912

ABSTRACT

The promoter of the Kirsten ras (KRAS) proto-oncogene contains, upstream of the transcription start site, a quadruplex-forming motif called 32R with regulatory functions. As guanine under oxidative stress can be oxidized to 8-oxoguanine (8OG), we investigated the capacity of glycosylases 8-oxoguanine glycosylase (OGG1) and endonuclease VIII-like 1 (Neil1) to excise 8OG from 32R, either in duplex or G-quadruplex (G4) conformation. We found that OGG1 efficiently excised 8OG from oxidized 32R in duplex but not in G4 conformation. By contrast, glycosylase Neil1 showed more activity on the G4 than the duplex conformation. We also found that the excising activity of Neil1 on folded 32R depended on G4 topology. Our data suggest that Neil1, besides being involved in base excision repair pathway (BER), could play a role on KRAS transcription.


Subject(s)
DNA Damage , DNA Glycosylases/metabolism , DNA Repair , G-Quadruplexes , Transcription, Genetic , Cell Line, Tumor , DNA/metabolism , Guanine/analogs & derivatives , Guanine/metabolism , Humans , Oxidative Stress , Promoter Regions, Genetic , Proto-Oncogene Mas , Proto-Oncogene Proteins p21(ras)/genetics
4.
Nucleic Acids Res ; 46(2): 661-676, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29165690

ABSTRACT

KRAS is one of the most mutated genes in human cancer. It is controlled by a G4 motif located upstream of the transcription start site. In this paper, we demonstrate that 8-oxoguanine (8-oxoG), being more abundant in G4 than in non-G4 regions, is a new player in the regulation of this oncogene. We designed oligonucleotides mimicking the KRAS G4-motif and found that 8-oxoG impacts folding and stability of the G-quadruplex. Dimethylsulphate-footprinting showed that the G-run carrying 8-oxoG is excluded from the G-tetrads and replaced by a redundant G-run in the KRAS G4-motif. Chromatin immunoprecipitation revealed that the base-excision repair protein OGG1 is recruited to the KRAS promoter when the level of 8-oxoG in the G4 region is raised by H2O2. Polyacrylamide gel electrophoresis evidenced that OGG1 removes 8-oxoG from the G4-motif in duplex, but when folded it binds to the G-quadruplex in a non-productive way. We also found that 8-oxoG enhances the recruitment to the KRAS promoter of MAZ and hnRNP A1, two nuclear factors essential for transcription. All this suggests that 8-oxoG in the promoter G4 region could have an epigenetic potential for the control of gene expression.


Subject(s)
G-Quadruplexes , Guanine/analogs & derivatives , Proto-Oncogene Proteins p21(ras)/chemistry , Transcription Initiation Site , Transcription, Genetic , Cell Line, Tumor , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Guanine/chemistry , Guanine/metabolism , HEK293 Cells , Heterogeneous Nuclear Ribonucleoprotein A1/genetics , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Humans , Oxidation-Reduction , Promoter Regions, Genetic/genetics , Protein Binding , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Int J Mol Sci ; 21(17)2020 Aug 28.
Article in English | MEDLINE | ID: mdl-32872305

ABSTRACT

In pancreatic Panc-1 cancer cells, an increase of oxidative stress enhances the level of 7,8-dihydro-8-oxoguanine (8OG) more in the KRAS promoter region containing G4 motifs than in non-G4 motif G-rich genomic regions. We found that H2O2 stimulates the recruitment to the KRAS promoter of poly [ADP-ribose] polymerase 1 (PARP-1), which efficiently binds to local G4 structures. Upon binding to G4 DNA, PARP-1 undergoes auto PARylation and thus becomes negatively charged. In our view this should favor the recruitment to the KRAS promoter of MAZ and hnRNP A1, as these two nuclear factors, because of their isoelectric points >7, are cationic in nature under physiological conditions. This is indeed supported by pulldown assays which showed that PARP-1, MAZ, and hnRNP A1 form a multiprotein complex with an oligonucleotide mimicking the KRAS G4 structure. Our data suggest that an increase of oxidative stress in Panc-1 cells activates a ROS-G4-PARP-1 axis that stimulates the transcription of KRAS. This mechanism is confirmed by the finding that when PARP-1 is silenced by siRNA or auto PARylation is inhibited by Veliparib, the expression of KRAS is downregulated. When Panc-1 cells are treated with H2O2 instead, a strong up-regulation of KRAS transcription is observed.


Subject(s)
Hydrogen Peroxide/pharmacology , Pancreatic Neoplasms/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Up-Regulation , Benzimidazoles/pharmacology , Cell Line, Tumor , DNA-Binding Proteins/metabolism , G-Quadruplexes , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Guanine/analogs & derivatives , Guanine/metabolism , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Humans , Oxidative Stress , Pancreatic Neoplasms/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins p21(ras)/chemistry , Transcription Factors/metabolism
6.
Biomacromolecules ; 20(7): 2530-2544, 2019 07 08.
Article in English | MEDLINE | ID: mdl-31241900

ABSTRACT

Nowadays, active targeting of nanotherapeutics is a challenging issue. Here, we propose a rational design of a ternary nanoassembly (SAP) composed of nonionic amphiphilic ß-cyclodextrins (amphiphilic CD) incorporating pheophorbide (Pheo) as a phototherapeutic and an adamantanyl-folic acid conjugate (Ada-FA) to target tumor cells overexpressing α-folate receptor (FR-α(+)). Dynamic light scattering and ζ-potential pointed out the presence of nanoassemblies bearing a negative surface charge (ζ = -51 mV). Morphology of SAP was investigated by atomic force microscopy and microphotoluminescence, indicating the presence of highly emissive near-spherical assemblies of about 280 nm in size. Complementary spectroscopic techniques such as ROESY-NMR, UV/vis and steady-state fluorescence revealed that the folic acid protrudes out of amphiphilic CD rims, prone for recognition with FR-α. Pheo was strongly loaded in the nanoassembly mostly in monomeric form, thus generating singlet oxygen (1O2) and consequentely showing phototherapeutic action. SAP remained stable until 2 weeks in aqueous solutions. Stability studies in biologically relevant media pointed out the ability of SAP to interact with serum proteins by means of the oligoethylenglycole fringe, without destabilization. Release experiments demonstrated the sustained release of Pheo from SAP in environments mimiking physiological conditions (∼20% within 1 week), plausibly suggesting low Pheo leaking and high integrity of the assembly within 24 h, time spent on average to reach the target sites. Cellular uptake of SAP was confirmed by confocal microscopy, pointing out that SAP was internalized into the tumoral cells expressing FR-α more efficiently than SP. SAP showed improved phototoxicity in human breast MCF-7 cancer cells FR-α(+) (IC50 = 270 nM) with respect to human prostate carcinoma PC3 cells (IC50 = 700 nM) that express a low level of that receptor (FR-α(-)). Finally, an improved phototoxicity in FR-α(+) MCF-7 cells (IC50 = 270 nM) was assessed after treatment with SAP vs SP (IC50 = 600 nM) which was designed without Ada-FA as a targeting unit.


Subject(s)
Cyclodextrins , Drug Delivery Systems , Folic Acid , Neoplasms , Photochemotherapy , Cyclodextrins/chemistry , Cyclodextrins/pharmacology , Folic Acid/chemistry , Folic Acid/pharmacology , Humans , MCF-7 Cells , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , PC-3 Cells
7.
RNA Biol ; 15(10): 1273-1285, 2018.
Article in English | MEDLINE | ID: mdl-30306823

ABSTRACT

Datasets reporting microRNA expression profiles in normal and cancer cells show that miR-216b is aberrantly downregulated in pancreatic ductal adenocarcinoma (PDAC). We found that KRAS, whose mutant G12D allele drives the pathogenesis of PDAC, is a target of miR-216b. To suppress oncogenic KRAS in PDAC cells, we designed single-stranded (ss) miR-216b mimics with unlocked nucleic acid (UNA) modifications to enhance their nuclease resistance. We prepared variants of ss-miR-216b mimics with and without a 5' phosphate group. Both variants strongly suppressed oncogenic KRAS in PDAC cells and inhibited colony formation in pancreatic cancer cells. We observed that the designed ss-miR-216b mimics engaged AGO2 to promote the silencing of KRAS. We also tested a new delivery strategy based on the use of palmityl-oleyl-phosphatidylcholine (POPC) liposomes functionalized with ss-miR-216b conjugated with two palmityl chains and a lipid-modified cell penetrating peptide (TAT). These versatile nanoparticles suppressed oncogenic KRAS in PDAC cells.


Subject(s)
Argonaute Proteins/genetics , Carcinoma, Pancreatic Ductal/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins p21(ras)/genetics , 3' Untranslated Regions/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/therapy , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Lipids/chemistry , Lipids/pharmacology , Liposomes/chemistry , Liposomes/pharmacology , MicroRNAs/chemistry , MicroRNAs/pharmacology , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Pancreas/metabolism , Pancreas/pathology , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors
8.
Biochim Biophys Acta ; 1859(4): 663-74, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26855080

ABSTRACT

It is now well established that in the human genome the canonical double helix coexists with folded G-quadruplex structures that are known to have important biological functions. In this review we summarize the current knowledge on quadruplex formation in the promoters of the ras genes that are mutated in about 30% of all human cancers. We describe the nuclear proteins that recognize these unusual DNA structures and discuss their function in transcription. We also examine the formation of G-quadruplexes in the 5'-untranslated region of the ras transcripts and conclude this review by reporting strategies that use either ras G-quadruplexes or proteins recognizing the ras G-quadruplexes as targets of anticancer small molecules.


Subject(s)
G-Quadruplexes , Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Transcription, Genetic , 5' Untranslated Regions/genetics , Genome, Human , Humans , Neoplasms/therapy , Promoter Regions, Genetic , Small Molecule Libraries/therapeutic use
9.
Invest New Drugs ; 35(1): 115-123, 2017 02.
Article in English | MEDLINE | ID: mdl-27726093

ABSTRACT

Photodynamic therapy (PDT) has drawn considerable attention for its efficacy against certain types of cancers. It shows however limits in the case of deep cancers, favoring tumor recurrence under suboptimal conditions. More insight into the molecular mechanisms of PDT-induced cytotoxicity and cytoprotection is essential to extend and strengthen this therapeutic modality. As PDT induces iNOS/NO in both tumor and microenvironment, we examined the role of nitric oxide (NO) in cytotoxicity and cytoprotection. Our findings show that NO mediates its cellular effects by acting on the NF-κB/YY1/RKIP loop, which controls cell growth and apoptosis. The cytoprotective effect of PDT-induced NO is observed at low NO levels, which activate the pro-survival/anti-apoptotic NF-κB and YY1, while inhibiting the anti-survival/pro-apoptotic and metastasis suppressor RKIP. In contrast, high PDT-induced NO levels inhibit NF-κB and YY1 and induce RKIP, resulting in significant anti-tumor activity. These findings reveal a critical role played by NO in PDT and suggest that the use of bifunctional PDT agents composed of a photosensitizer and a NO-donor could enhance the photo-treatment effect. A successful application of NO in anticancer therapy requires control of its concentration in the target tissue. To address this issue we propose as PDT agent, a bimolecular conjugate called DR2, composed of a photosensitizer (Pheophorbide a) and a non-steroidal anti-androgen molecule capable of releasing NO under the exclusive control of light. The mechanism of action of DR2 in prostate cancer cells is reported and discussed.


Subject(s)
Chlorophyll/analogs & derivatives , Nitric Oxide Donors/pharmacology , Photochemotherapy , Photosensitizing Agents/pharmacology , Prostatic Neoplasms/drug therapy , Triazenes/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Chlorophyll/pharmacology , Humans , Male , NF-kappa B/metabolism , Nitric Oxide/metabolism , Phosphatidylethanolamine Binding Protein/metabolism , Prostatic Neoplasms/metabolism , YY1 Transcription Factor/metabolism
10.
Biochim Biophys Acta Gen Subj ; 1861(5 Pt B): 1389-1398, 2017 May.
Article in English | MEDLINE | ID: mdl-27888145

ABSTRACT

KRAS is one of the most mutated genes in human cancer. Its crucial role in the tumourigenesis of pancreatic ductal adenocarcinoma (PDAC) has been widely demonstrated. As this deadly cancer does not sufficiently respond to conventional chemotherapies, it is important to increase our knowledge of pancreatic cancer biology, in particular how oncogenic KRAS is regulated. The promoter of KRAS contains a GA-element composed of runs of guanines that fold into a G4 structure. This unusual DNA conformation is recognized by several nuclear proteins, including MAZ and hnRNP A1. Recent data have revealed that KRAS is interconnected to ILK and hnRNP A1 in a circuitry that enables pancreatic cancer cells to maintain an aggressive phenotype. The present review illustrates recent advances on how KRAS is regulated in pancreatic cancer cells, focusing on the formation of G4 structures in the KRAS promoter and their interaction with hnRNP A1. The newly discovered KRAS-ILK-hnRNP A1 regulatory loop is discussed, emphasizing its potential as a therapeutic target for PDAC-specific molecules. This article is part of a Special Issue entitled "G-quadruplex" Guest Editor: Dr. Concetta Giancola and Dr. Daniela Montesarchio.


Subject(s)
DNA, Neoplasm/genetics , G-Quadruplexes , Guanosine/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Transcription, Genetic , Binding Sites , DNA, Neoplasm/chemistry , DNA, Neoplasm/metabolism , Gene Expression Regulation, Neoplastic , Guanosine/chemistry , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/chemistry , Humans , Ligands , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Polymorphism, Genetic , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins p21(ras)/metabolism , Structure-Activity Relationship , Transcriptional Activation
11.
Nucleic Acids Res ; 42(13): 8379-88, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25013182

ABSTRACT

The HRAS promoter contains immediately upstream of the transcription start site two neighboring G-elements, each capable of folding into a G-quadruplex structure. We have previously found that these G-quadruplexes bind to the zinc-finger transcription factors MAZ and Sp1. In the present study we have examined the interaction between the HRAS promoter and MAZ, demonstrating for the first time that the protein unfolds the G-quadruplex structures. We also demonstrate that MAZ-GST, in the presence of the complementary strands, promotes a rapid transformation of the two HRAS quadruplexes into duplexes. By a mutational analysis of the HRAS G-elements, we dissected the MAZ-binding sites from the quadruplex-forming motifs, finding that the two neighboring G-quadruplexes bring about a dramatic repression of transcription, in a synergistic manner. We also discovered that the two G-quadruplexes are strong targets for small anticancer molecules. We found that a cell-penetrating anthratiophenedione (ATPD-1), which binds tightly to the G-quadruplexes (ΔT > 15°C), promotes the total extinction of HRAS transcription. In contrast, when one of the two G-quadruplexes was abrogated by point mutations, ATPD-1 repressed transcription by only 50%. Our study provides relevant information for the rationale design of targeted therapy drugs specific for the HRAS oncogene.


Subject(s)
DNA-Binding Proteins/metabolism , G-Quadruplexes , Gene Expression Regulation, Neoplastic , Promoter Regions, Genetic , Proto-Oncogene Proteins p21(ras)/genetics , Transcription Factors/metabolism , Cell Line, Tumor , DNA/chemistry , DNA/metabolism , Gene Silencing , Humans , Transcriptional Activation , Urinary Bladder Neoplasms/genetics
12.
Nucleic Acids Res ; 41(7): 4049-64, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23471001

ABSTRACT

KRAS mutations are primary genetic lesions leading to pancreatic cancer. The promoter of human KRAS contains a nuclease-hypersensitive element (NHE) that can fold in G4-DNA structures binding to nuclear proteins, including MAZ (myc-associated zinc-finger). Here, we report that MAZ activates KRAS transcription. To knockdown oncogenic KRAS in pancreatic cancer cells, we designed oligonucleotides that mimic one of the G-quadruplexes formed by NHE (G4-decoys). To increase their nuclease resistance, two locked nucleic acid (LNA) modifications were introduced at the 3'-end, whereas to enhance the folding and stability, two polycyclic aromatic hydrocarbon units (TINA or AMANY) were inserted internally, to cap the quadruplex. The most active G4-decoy (2998), which had two para-TINAs, strongly suppressed KRAS expression in Panc-1 cells. It also repressed their metabolic activity (IC50 = 520 nM), and it inhibited cell growth and colony formation by activating apoptosis. We finally injected 2998 and control oligonucleotides 5153, 5154 (2 nmol/mouse) intratumorally in SCID mice bearing a Panc-1 xenograft. After three treatments, 2998 reduced tumor xenograft growth by 64% compared with control and increased the Kaplan-Meier median survival time by 70%. Together, our data show that MAZ-specific G4-decoys mimicking a KRAS quadruplex are promising for pancreatic cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , DNA-Binding Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins/antagonists & inhibitors , Transcription Factors/metabolism , ras Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis , Binding Sites , Binding, Competitive , Cell Line, Tumor , DNA/chemistry , G-Quadruplexes , Humans , Mice , Mice, SCID , Oligonucleotides/chemistry , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , Transcriptional Activation , ras Proteins/genetics , ras Proteins/metabolism
13.
Mol Cancer ; 13: 75, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24684778

ABSTRACT

BACKGROUND: Porphyrin TMPyP4 (P4) and its C14H28-alkyl derivative (C14) are G-quadruplex binders and singlet oxygen (1O2) generators. In contrast, TMPyP2 (P2) produces 1O2 but it is not a G-quadruplex binder. As their photosensitizing activity is currently undefined, we report in this study their efficacy against a melanoma skin tumour and describe an in vitro mechanistic study which gives insights into their anticancer activity. METHODS: Uptake and antiproliferative activity of photoactivated P2, P4 and C14 have been investigated in murine melanoma B78-H1 cells by FACS, clonogenic and migration assays. Apoptosis was investigated by PARP-1 cleavage and annexin-propidium iodide assays. Biodistribution and in vivo anticancer activity were tested in melanoma tumour-bearing mice. Porphyrin binding and photocleavage of G-rich mRNA regions were investigated by electrophoresis and RT-PCR. Porphyrin effect on ERK pathway was explored by Western blots. RESULTS: Thanks to its higher lipophylicity C14 was taken up by murine melanoma B78-H1 cells up to 30-fold more efficiently than P4. When photoactivated (7.2 J/cm2) in B78-H1 melanoma cells, P4 and C14, but not control P2, caused a strong inhibition of metabolic activity, clonogenic growth and cell migration. Biodistribution studies on melanoma tumour-bearing mice showed that P4 and C14 localize in the tumour. Upon irradiation (660 nm, 193 J/cm2), P4 and C14 retarded tumour growth and increased the median survival time of the treated mice by ~50% (P <0.01 by ANOVA), whereas porphyrin P2 did not. The light-dependent mechanism mediated by P4 and C14 is likely due to the binding to and photocleavage of G-rich quadruplex-forming sequences within the 5'-untranslated regions of the mitogenic ras genes. This causes a decrease of RAS protein and inhibition of downstream ERK pathway, which stimulates proliferation. Annexin V/propidium iodide and PARP-1 cleavage assays showed that the porphyrins arrested tumour growth by apoptosis and necrosis. C14 also showed an intrinsic light-independent anticancer activity, as recently reported for G4-RNA binders. CONCLUSIONS: Porphyrins P4 and C14 impair the clonogenic growth and migration of B78-H1 melanoma cells and inhibit melanoma tumour growth in vivo. Evidence is provided that C14 acts through light-dependent (mRNA photocleavage) and light-independent (translation inhibition) mechanisms.


Subject(s)
Photochemotherapy , Photosensitizing Agents/administration & dosage , Porphyrins/administration & dosage , Skin Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , G-Quadruplexes/drug effects , Humans , MAP Kinase Signaling System/drug effects , Mice , Porphyrins/chemistry
14.
Biochim Biophys Acta Mol Cell Res ; 1871(1): 119606, 2024 01.
Article in English | MEDLINE | ID: mdl-37852325

ABSTRACT

Nitric oxide is a pleiotropic free radical produced by three nitric oxide synthases (NOS1-3), of which inducible NOS2 is involved in tumor initiation and progression. In this study, RNA-seq, ChIP-seq and qRT-PCR experiments combined with bioinformatic analyses showed that NRF2 is a repressor of NOS2 gene by maintaining a distal enhancer located 22 kb downstream of TSS in an inactive state. Deletion of NRF2 leads to activation of the enhancer, which exerts a pioneering function before it is fully activated. Specifically, NRF2 controls the expression of NOS2 in response to intracellular oxidative stress and extracellular oxygen pressure. We found that abrogation of NOS2 expression by siRNAs partially reduced the ability of WT Panc-1 cells to form 3D spheroids, but strongly reduced the formation of 3D spheroids by NRF2-depleted Panc-1 cells. Mechanistically, this effect correlates with the finding that NOS2 and nitric oxide stimulate epithelial-to-mesenchymal transition in NRF2-depleted Panc-1 and MIA PaCa-2 cells. We also found that knockdown of NOS2 leads to blockade of 3D matrigel invasion of NRF2-depleted PDAC cells, demonstrating that a short-circuit in the reciprocal regulation of NOS2 and NRF2 attenuates the malignancy of PDAC cells. In summary, we show for the first time that: (i) NRF2 is a suppressor of NOS2 in pancreatic cancer cells; (ii) NRF2 binds to and inactivates an enhancer located 22 kb downstream of TSS of the NOS2 gene; (iii) activation of NOS2 requires suppression of NRF2; (iv) NOS2 is required for NRF2-depleted Panc-1 cells to maintain their malignancy and invasiveness.


Subject(s)
NF-E2-Related Factor 2 , Pancreatic Neoplasms , Humans , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
15.
Invest New Drugs ; 31(1): 192-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22688292

ABSTRACT

Pheophorbide a (Pba) is a chlorophyll catabolite that has been proposed as photosensitizer in photodynamic therapy. In a previous study we conjugated Pba to monomethoxy-polyethylene glycol (mPEG-Pba), to increase its solubility and pharmacokinetics. Here, we compare the photodynamic therapy efficacy of free Pba and mPEG-Pba to cure a subcutaneous amelanotic melanoma transplanted in C57/BL6 mice. The photosensitizers, i.p. injected (30 mg/kg), showed no toxicity when the animals were kept in the dark. But, after photoactivation with a 660 nm laser (fluence of 193 J/cm(2)), both photosensitizers, in particular mPEG-Pba, showed a strong efficacy to cure the tumor, both in terms of tumor growth delay and increase of Kaplan-Meier median survival time. Together, our in vivo data demonstrate that mPEG-conjugated Pba is a promising photosensitizer for the photodynamic therapy of cancer.


Subject(s)
Chlorophyll/analogs & derivatives , Melanoma, Amelanotic/drug therapy , Polyethylene Glycols/chemistry , Radiation-Sensitizing Agents/administration & dosage , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Chlorophyll/administration & dosage , Chlorophyll/chemistry , Female , Light , Mice , Mice, Inbred C57BL , Photochemotherapy , Radiation-Sensitizing Agents/chemistry
16.
iScience ; 26(12): 108566, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38144458

ABSTRACT

In pancreatic ductal adenocarcinomas (PDAC), the KRASG12D-NRF2 axis controls cellular functions such as redox homeostasis and metabolism. Disruption of this axis through suppression of NRF2 leads to profound reprogramming of metabolism. Unbiased transcriptome and metabolome analyses showed that PDAC cells with disrupted KRASG12D-NRF2 signaling (NRF2-/- cells) shift from aerobic glycolysis to metabolic pathways fed by amino acids. Metabolome, RNA-seq and qRT-PCR analyses revealed a blockade of the urea cycle, making NRF2-/- cells dependent on exogenous arginine for survival. Arginine is channeled into anabolic pathways, including the synthesis of phosphocreatine, which generates an energy buffer essential for cell growth. A similar switch was observed in tumor clones that had survived FOLFIRINOX therapy or blockade of KRAS signaling. Inhibition of the creatine pathway with cyclocreatine reduced both ATP and invasion rate in 3D spheroids from NRF2-deficient PDAC cells. Our study provides basis for the rational development of combination therapies for pancreatic cancer.

17.
Front Immunol ; 13: 917998, 2022.
Article in English | MEDLINE | ID: mdl-35757716

ABSTRACT

Bi-directional transcription of Human Endogenous Retroviruses (hERVs) is a common feature of autoimmunity, neurodegeneration and cancer. Higher rates of cancer incidence, neurodegeneration and autoimmunity but a lower prevalence of autoimmune diseases characterize elderly people. Although the re-expression of hERVs is commonly observed in different cellular models of senescence as a result of the loss of their epigenetic transcriptional silencing, the hERVs modulation during aging is more complex, with a peak of activation in the sixties and a decline in the nineties. What is clearly accepted, instead, is the impact of the re-activation of dormant hERV on the maintenance of stemness and tissue self-renewing properties. An innate cellular immunity system, based on the RLR-MAVS circuit, controls the degradation of dsRNAs arising from the transcription of hERV elements, similarly to what happens for the accumulation of cytoplasmic DNA leading to the activation of cGAS/STING pathway. While agonists and inhibitors of the cGAS-STING pathway are considered promising immunomodulatory molecules, the effect of the RLR-MAVS pathway on innate immunity is still largely based on correlations and not on causality. Here we review the most recent evidence regarding the activation of MDA5-RIG1-MAVS pathway as a result of hERV de-repression during aging, immunosenescence, cancer and autoimmunity. We will also deal with the epigenetic mechanisms controlling hERV repression and with the strategies that can be adopted to modulate hERV expression in a therapeutic perspective. Finally, we will discuss if the RLR-MAVS signalling pathway actively modulates physiological and pathological conditions or if it is passively activated by them.


Subject(s)
Endogenous Retroviruses , Neoplasms , Aged , Aging , Endogenous Retroviruses/genetics , Humans , Immunity, Innate , Neoplasms/genetics , Signal Transduction/physiology
18.
J Photochem Photobiol B ; 231: 112449, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35504235

ABSTRACT

Cationic porphyrins bearing an alkyl side chain of 14 (2b) or 18 (2d) carbons dramatically inhibit proliferation of pancreatic cancer cells following treatment with light. We have compared two different ways of delivering porphyrin 2d: either in free form or engrafted into palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine liposomes (L-2d). Cell cytometry shows that while free 2d is taken up by pancreatic cancer cells by active (endocytosis) and passive (membrane fusion) transports, L-2d is internalized solely by endocytosis. Confocal microscopy showed that free 2d co-localizes with the cell membrane and lysosomes, whereas L-2d partly co-localizes with lysosomes and ER. It is found that free 2d inhibits the KRAS-Nrf2-GPX4 axis and strongly triggers lipid peroxidation, resulting in cell death by ferroptosis. By contrast, L-2d does not affect the KRAS-Nrf2-GPX4 axis and activates cell death mainly through apoptosis. Overall, our study demonstrates for the first time that cationic alkyl porphyrins, which have a IC50 ~ 23 nM, activate a dual mechanism of cell death, ferroptosis and apoptosis, where the predominant form depends on the delivery mode.


Subject(s)
Pancreatic Neoplasms , Porphyrins , Apoptosis , Cations , Humans , Liposomes/chemistry , NF-E2-Related Factor 2 , Pancreatic Neoplasms/drug therapy , Porphyrins/pharmacology , Proto-Oncogene Proteins p21(ras) , Pancreatic Neoplasms
19.
J Biol Chem ; 285(29): 22003-16, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20457603

ABSTRACT

The murine KRAS promoter contains a G-rich nuclease hypersensitive element (GA-element) upstream of the transcription start site that is essential for transcription. Pulldown and chromatin immunoprecipitation assays demonstrate that this GA-element is bound by the Myc-associated zinc finger (MAZ) and poly(ADP-ribose) polymerase 1 (PARP-1) proteins. These proteins are crucial for transcription, because when they are knocked down by short hairpin RNA, transcription is down-regulated. This is also the case when the poly(ADP-ribosyl)ation activity of PARP-1 is inhibited by 3,4-dihydro-5-[4-(1-piperidinyl) butoxyl]-1(2H) isoquinolinone. We found that MAZ specifically binds to the duplex and quadruplex conformations of the GA-element, whereas PARP-1 shows specificity only for the G-quadruplex. On the basis of fluorescence resonance energy transfer melting and polymerase stop assays we saw that MAZ stabilizes the KRAS quadruplex. When the capacity of folding in the GA-element is abrogated by specific G --> T or G --> A point mutations, KRAS transcription is down-regulated. Conversely, guanidine-modified phthalocyanines, which specifically interact with and stabilize the KRAS G-quadruplex, push the promoter activity up to more than double. Collectively, our data support a transcription mechanism for murine KRAS that involves MAZ, PARP-1 and duplex-quadruplex conformational changes in the promoter GA-element.


Subject(s)
DNA-Binding Proteins/metabolism , G-Quadruplexes , Guanine/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins p21(ras)/genetics , Transcription Factors/metabolism , Animals , Base Sequence , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Fluorescence Resonance Energy Transfer , Indoles/chemistry , Isoindoles , Ligands , Mice , Molecular Sequence Data , Mutant Proteins/metabolism , NIH 3T3 Cells , Nucleic Acid Conformation , Protein Binding , Protein Stability , Transcription, Genetic
20.
Lasers Surg Med ; 43(7): 575-85, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22057485

ABSTRACT

BACKGROUND AND OBJECTIVE: Photodynamic therapy (PDT) is a therapeutic modality whose efficacy depends on several factors including type of photosensitizer, light fluence and cellular response. Cell recurrence is one of the problems still unsolved in PDT. In this work we found that in B78-H1 murine amelanotic melanoma cells there is a correlation between cell recurrence and the NF-κB/Snail/RKIP loop. MATERIALS AND METHODS: Proliferation and migration of surviving cells were analyzed by MTT and wound-scratch assays. The levels of ROS/NO in B78-H1 melanoma cells treated with pheophorbide a (Pba) and light (Pba/PDT) were measured by FACS, while expression of NF-κB, Snail and RKIP were determined by Western blots. The mechanism of cell death was investigated by caspase and microscopy assays. RESULTS: Our data show that after a low-dose Pba/PDT treatment, B78-H1 cells are able to recover. This correlates with a low level of NO production, which blocks apoptosis via NF-κB pathway. Western blot analyses showed that a low-dose Pba/PDT increases the expression of NF-κB and anti-apoptotic Snail, but reduces the expression of pro-apoptotic RKIP. The role played by NF-κB in the modulation of Snail and RKIP was investigated using DHMEQ: a NF-κB inhibitor which behaves as NO donor. DHMEQ caused a decrease of Snail and an increase of RKIP expression. When B78-H1 cells were treated with a low dose Pba/PDT and DHMEQ, the NO level strongly increased, with the result that Snail was down-regulated and RKIP was upregulated, as observed with a high-dose Pba/PDT. CONCLUSION: One major problem in PDT is the cellular rescue occurring in tissue regions receiving a low-dose PDT. To minimize this problem and sensitize cancer cells to PDT we propose a combined treatment in which the photosensitizer is delivered with a donor of NO acting on the NF-κB/Snail/RKIP loop.


Subject(s)
Apoptosis/drug effects , Chlorophyll/analogs & derivatives , NF-kappa B/metabolism , Phosphatidylethanolamine Binding Protein/metabolism , Photochemotherapy , Photosensitizing Agents/pharmacology , Transcription Factors/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Cell Survival , Chlorophyll/pharmacology , Chlorophyll/therapeutic use , Dose-Response Relationship, Drug , Melanoma, Amelanotic/drug therapy , Melanoma, Amelanotic/pathology , Mice , Nitric Oxide/metabolism , Photosensitizing Agents/therapeutic use , Reactive Oxygen Species/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Snail Family Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL