Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Am J Pathol ; 192(9): 1321-1335, 2022 09.
Article in English | MEDLINE | ID: mdl-35750257

ABSTRACT

Toll-like receptor 3 (TLR3) is an endosomal receptor expressed in several immune and epithelial cells. Recent studies have highlighted its expression also in solid tumors, including prostate cancer (PCa), and have described its role primarily in the proinflammatory response and induction of apoptosis. It is up-regulated in some castration-resistant prostate cancers. However, the role of TLR3 in prostate cancer progression remains largely unknown. The current study experimentally demonstrated that exogenous TLR3 activation in PCa cell lines leads to a significant induction of secretion of the cytokines IL-6, IL-8, and interferon-ß, depending on the model and chemoresistance status. Transcriptomic analysis of TLR3-overexpressing cells revealed a functional program that is enriched for genes involved in the regulation of cell motility, migration, and tumor invasiveness. Increased motility, migration, and invasion in TLR3-overexpressing cell line were confirmed by several in vitro assays and using an orthotopic prostate xenograft model in vivo. Furthermore, TLR3-ligand induced apoptosis via cleavage of caspase-3/7 and poly (ADP-ribose) polymerase, predominantly in TLR3-overexpressing cells. These results indicate that TLR3 may be involved in prostate cancer progression and metastasis; however, it might also represent an Achilles heel of PCa, which can be exploited for targeted therapy.


Subject(s)
Prostatic Neoplasms , Toll-Like Receptor 3 , Animals , Apoptosis , Cell Line, Tumor , Humans , Male , Poly I-C/pharmacology , Prostate/pathology , Prostatic Neoplasms/pathology , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism
2.
Clin Exp Metastasis ; 39(2): 375-390, 2022 04.
Article in English | MEDLINE | ID: mdl-34994868

ABSTRACT

The transcription factor c-Myb is an oncoprotein promoting cell proliferation and survival when aberrantly activated/expressed, thus contributing to malignant transformation. Overexpression of c-Myb has been found in leukemias, breast, colon and adenoid cystic carcinoma. Recent studies revealed its expression also in osteosarcoma cell lines and suggested its functional importance during bone development. However, the relevance of c-Myb in control of osteosarcoma progression remains unknown. A retrospective clinical study was carried out to assess a relationship between c-Myb expression in archival osteosarcoma tissues and prognosis in a cohort of high-grade osteosarcoma patients. In addition, MYB was depleted in metastatic osteosarcoma cell lines SAOS-2 LM5 and 143B and their growth, chemosensitivity, migration and metastatic activity were determined. Immunohistochemical analysis revealed that high c-Myb expression was significantly associated with poor overall survival in the cohort and metastatic progression in young patients. Increased level of c-Myb was detected in metastatic osteosarcoma cell lines and its depletion suppressed their growth, colony-forming capacity, migration and chemoresistance in vitro in a cell line-dependent manner. MYB knock-out resulted in reduced metastatic activity of both SAOS-2 LM5 and 143B cell lines in immunodeficient mice. Transcriptomic analysis revealed the c-Myb-driven functional programs enriched for genes involved in the regulation of cell growth, stress response, cell adhesion and cell differentiation/morphogenesis. Wnt signaling pathway was identified as c-Myb target in osteosarcoma cells. Taken together, we identified c-Myb as a negative prognostic factor in osteosarcoma and showed its involvement in the regulation of osteosarcoma cell growth, chemosensitivity, migration and metastatic activity.


Subject(s)
Bone Neoplasms , Osteosarcoma , Animals , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Mice , Osteosarcoma/pathology , Prognosis , Retrospective Studies , Wnt Signaling Pathway
3.
Mol Oncol ; 14(10): 2487-2503, 2020 10.
Article in English | MEDLINE | ID: mdl-32579780

ABSTRACT

As treatment options for patients with incurable metastatic castration-resistant prostate cancer (mCRPC) are considerably limited, novel effective therapeutic options are needed. Checkpoint kinase 1 (CHK1) is a highly conserved protein kinase implicated in the DNA damage response (DDR) pathway that prevents the accumulation of DNA damage and controls regular genome duplication. CHK1 has been associated with prostate cancer (PCa) induction, progression, and lethality; hence, CHK1 inhibitors SCH900776 (also known as MK-8776) and the more effective SCH900776 analog MU380 may have clinical applications in the therapy of PCa. Synergistic induction of DNA damage with CHK1 inhibition represents a promising therapeutic approach that has been tested in many types of malignancies, but not in chemoresistant mCRPC. Here, we report that such therapeutic approach may be exploited using the synergistic action of the antimetabolite gemcitabine (GEM) and CHK1 inhibitors SCH900776 and MU380 in docetaxel-resistant (DR) mCRPC. Given the results, both CHK1 inhibitors significantly potentiated the sensitivity to GEM in a panel of chemo-naïve and matched DR PCa cell lines under 2D conditions. MU380 exhibited a stronger synergistic effect with GEM than clinical candidate SCH900776. MU380 alone or in combination with GEM significantly reduced spheroid size and increased apoptosis in all patient-derived xenograft 3D cultures, with a higher impact in DR models. Combined treatment induced premature mitosis from G1 phase resulting in the mitotic catastrophe as a prestage of apoptosis. Finally, treatment by MU380 alone, or in combination with GEM, significantly inhibited tumor growth of both PC339-DOC and PC346C-DOC xenograft models in mice. Taken together, our data suggest that metabolically robust and selective CHK1 inhibitor MU380 can bypass docetaxel resistance and improve the effectiveness of GEM in DR mCRPC models. This approach might allow for dose reduction of GEM and thereby minimize undesired toxicity and may represent a therapeutic option for patients with incurable DR mCRPC.


Subject(s)
Checkpoint Kinase 1/antagonists & inhibitors , Deoxycytidine/analogs & derivatives , Docetaxel/pharmacology , Drug Resistance, Neoplasm/drug effects , Mitosis , Piperidines/pharmacology , Prostatic Neoplasms/pathology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Checkpoint Kinase 1/metabolism , Deoxycytidine/pharmacology , Humans , Male , Mice, SCID , Mitosis/drug effects , Piperidines/chemistry , Pyrazoles/chemistry , Pyrimidines/chemistry , S Phase/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
4.
Sci Rep ; 9(1): 5695, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30952903

ABSTRACT

Skp2 is a crucial component of SCFSkp2 E3 ubiquitin ligase and is often overexpressed in various types of cancer, including prostate cancer (PCa). The epithelial-to-mesenchymal transition (EMT) is involved in PCa progression. The acquisition of a mesenchymal phenotype that results in a cancer stem cell (CSC) phenotype in PCa was described. Therefore, we aimed to investigate the expression and localization of Skp2 in clinical samples from patients with PCa, the association of Skp2 with EMT status, and the role of Skp2 in prostate CSC. We found that nuclear expression of Skp2 was increased in patients with PCa compared to those with benign hyperplasia, and correlated with high Gleason score in PCa patients. Increased Skp2 expression was observed in PCa cell lines with mesenchymal and CSC-like phenotype compared to their epithelial counterparts. Conversely, the CSC-like phenotype was diminished in cells in which SKP2 expression was silenced. Furthermore, we observed that Skp2 downregulation led to the decrease in subpopulation of CD44+CD24- cancer stem-like cells. Finally, we showed that high expression levels of both CD24 and CD44 were associated with favorable recurrence-free survival for PCa patients. This study uncovered the Skp2-mediated CSC-like phenotype with oncogenic functions in PCa.


Subject(s)
Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/physiology , Prostatic Neoplasms/genetics , S-Phase Kinase-Associated Proteins/genetics , Animals , CD24 Antigen/genetics , Cell Line, Tumor , Humans , Hyaluronan Receptors/genetics , Male , Mice , Mice, Nude , Neoplasm Grading , Neoplastic Stem Cells/metabolism , PC-3 Cells , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/physiopathology , Xenograft Model Antitumor Assays
5.
J Assist Reprod Genet ; 35(8): 1407-1417, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29948426

ABSTRACT

PURPOSE: The purpose of the study was to determine whether the GDF-15 is present in follicular fluid; to evaluate if there is a relation between follicular and serum levels of GDF-15 and fertility status of study subjects; and to test whether granulosa cells, oocytes, or both produce GDF-15. METHODS: This study used follicular fluid (FF, serum, and oocytes obtained under informed consent from women undergoing oocyte retrieval for in vitro fertilization. It also used ovaries from deceased preterm newborns. Collection of FF and blood at the time of oocyte retrieval, ELISA and western blot were performed to determine levels and forms of GDF-15. Concentrations of GDF-15 in FF and serum, its expression in ovarian tissue, and secretion from granulosa cells were analyzed. RESULTS: GDF-15 concentration in FF ranged from 35 to 572 ng/ml, as determined by ELISA. Western blot analysis revealed the GDF-15 pro-dimer only in FF. Both normal healthy and cancerous granulosa cells secreted GDF-15 into culture media. Primary oocytes displayed cytoplasmic GDF-15 positivity in immunostained newborn ovaries, and its expression was also observed in fully grown human oocytes. CONCLUSIONS: To the best of our knowledge, this is the first documentation of cytokine GDF-15 presence in follicular fluid. Its concentration was not associated with donor/patient fertility status. Our data also show that GDF-15 is expressed and inducible in both normal healthy and cancerous granulosa cells, as well as in oocytes.


Subject(s)
Cell Differentiation/genetics , Follicular Fluid/metabolism , Granulosa Cells/metabolism , Growth Differentiation Factor 15/genetics , Adult , Female , Fertilization in Vitro , Gene Expression Regulation, Developmental , Growth Differentiation Factor 15/isolation & purification , Humans , Oocyte Retrieval , Oocytes/metabolism
6.
Mol Cancer Ther ; 16(9): 1831-1842, 2017 09.
Article in English | MEDLINE | ID: mdl-28619751

ABSTRACT

Checkpoint-mediated dependency of tumor cells can be deployed to selectively kill them without substantial toxicity to normal cells. Specifically, loss of CHK1, a serine threonine kinase involved in the surveillance of the G2-M checkpoint in the presence of replication stress inflicted by DNA-damaging drugs, has been reported to dramatically influence the viability of tumor cells. CHK1's pivotal role in maintaining genomic stability offers attractive opportunity for increasing the selectivity, effectivity, and reduced toxicity of chemotherapy. Some recently identified CHK1 inhibitors entered clinical trials in combination with DNA antimetabolites. Herein, we report synthesis and profiling of MU380, a nontrivial analogue of clinically profiled compound SCH900776 possessing the highly unusual N-trifluoromethylpyrazole motif, which was envisioned not to undergo metabolic oxidative dealkylation and thereby provide greater robustness to the compound. MU380 is a selective and potent inhibitor of CHK1 which sensitizes a variety of tumor cell lines to hydroxyurea or gemcitabine up to 10 times. MU380 shows extended inhibitory effects in cells, and unlike SCH900776, does not undergo in vivo N-dealkylation to the significantly less selective metabolite. Compared with SCH900776, MU380 in combination with GEM causes higher accumulation of DNA damage in tumor cells and subsequent enhanced cell death, and is more efficacious in the A2780 xenograft mouse model. Overall, MU380 represents a novel state-of-the-art CHK1 inhibitor with high potency, selectivity, and improved metabolic robustness to oxidative N-dealkylation. Mol Cancer Ther; 16(9); 1831-42. ©2017 AACR.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Checkpoint Kinase 1/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Biomarkers , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Dealkylation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Methylation , Mice , Molecular Structure , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Xenograft Model Antitumor Assays
7.
Nat Commun ; 7: 13701, 2016 12 06.
Article in English | MEDLINE | ID: mdl-27922010

ABSTRACT

Lung cancer is the leading cause of cancer deaths, and effective treatments are urgently needed. Loss-of-function mutations in the DNA damage response kinase ATM are common in lung adenocarcinoma but directly targeting these with drugs remains challenging. Here we report that ATM loss-of-function is synthetic lethal with drugs inhibiting the central growth factor kinases MEK1/2, including the FDA-approved drug trametinib. Lung cancer cells resistant to MEK inhibition become highly sensitive upon loss of ATM both in vitro and in vivo. Mechanistically, ATM mediates crosstalk between the prosurvival MEK/ERK and AKT/mTOR pathways. ATM loss also enhances the sensitivity of KRAS- or BRAF-mutant lung cancer cells to MEK inhibition. Thus, ATM mutational status in lung cancer is a mechanistic biomarker for MEK inhibitor response, which may improve patient stratification and extend the applicability of these drugs beyond RAS and BRAF mutant tumours.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Cell Proliferation/drug effects , Lung Neoplasms/prevention & control , Mutation , Protein Kinase Inhibitors/pharmacology , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Benzamides/pharmacology , Cell Line, Tumor , Cell Proliferation/genetics , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice, Nude , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Pyridones/pharmacology , Pyrimidinones/pharmacology , RNA Interference , Thiophenes/pharmacology , Urea/analogs & derivatives , Urea/pharmacology , Xenograft Model Antitumor Assays , ras Proteins/genetics , ras Proteins/metabolism
8.
Oncotarget ; 6(34): 36156-71, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26416355

ABSTRACT

Plasticity of cancer cells, manifested by transitions between epithelial and mesenchymal phenotypes, represents a challenging issue in the treatment of neoplasias. Both epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) are implicated in the processes of metastasis formation and acquisition of stem cell-like properties. Mouse double minute (MDM) 2 and MDMX are important players in cancer progression, as they act as regulators of p53, but their function in EMT and metastasis may be contradictory. Here, we show that the EMT phenotype in multiple cellular models and in clinical prostate and breast cancer samples is associated with a decrease in MDM2 and increase in MDMX expression. Modulation of EMT-accompanying changes in MDM2 expression in benign and transformed prostate epithelial cells influences their migration capacity and sensitivity to docetaxel. Analysis of putative mechanisms of MDM2 expression control demonstrates that in the context of defective p53 function, MDM2 expression is regulated by EMT-inducing transcription factors Slug and Twist. These results provide an alternative context-specific role of MDM2 in EMT, cell migration, metastasis, and therapy resistance.


Subject(s)
Breast Neoplasms/metabolism , Epithelial-Mesenchymal Transition/physiology , Nuclear Proteins/biosynthesis , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins , Cell Line, Tumor , Female , Heterografts , Humans , Male , Mice , Mice, Nude , Phenotype , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transfection
9.
Mol Cancer ; 13: 113, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24884804

ABSTRACT

BACKGROUND: Tumor heterogeneity and the plasticity of cancer cells present challenges for effective clinical diagnosis and therapy. Such challenges are epitomized by neuroendocrine transdifferentiation (NED) and the emergence of neuroendocrine-like cancer cells in prostate tumors. This phenomenon frequently arises from androgen-depleted prostate adenocarcinoma and is associated with the development of castration-resistant prostate cancer and poor prognosis. RESULTS: In this study, we showed that NED was evoked in both androgen receptor (AR)-positive and AR-negative prostate epithelial cell lines by growing the cells to a high density. Androgen depletion and high-density cultivation were both associated with cell cycle arrest and deregulated expression of several cell cycle regulators, such as p27Kip1, members of the cyclin D protein family, and Cdk2. Dual inhibition of Cdk1 and Cdk2 using pharmacological inhibitor or RNAi led to modulation of the cell cycle and promotion of NED. We further demonstrated that the cyclic adenosine 3', 5'-monophosphate (cAMP)-mediated pathway is activated in the high-density conditions. Importantly, inhibition of cAMP signaling using a specific inhibitor of adenylate cyclase, MDL-12330A, abolished the promotion of NED by high cell density. CONCLUSIONS: Taken together, our results imply a new relationship between cell cycle attenuation and promotion of NED and suggest high cell density as a trigger for cAMP signaling that can mediate reversible NED in prostate cancer cells.


Subject(s)
Cell Transdifferentiation , Neuroendocrine Cells/pathology , Prostatic Neoplasms/pathology , Androgens/pharmacology , CDC2 Protein Kinase , Cell Count , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Transdifferentiation/drug effects , Cyclic AMP/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinases/metabolism , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/pathology , Humans , Immunohistochemistry , Male , Neuroendocrine Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Receptors, Androgen/metabolism , Signal Transduction/drug effects
10.
Eur J Med Chem ; 78: 54-64, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24675180

ABSTRACT

A substitution of the ammine ligands of cisplatin, cis-[Pt(NH3)2Cl2], for cyclin dependent kinase (CDK) inhibitor bohemine (boh), [2-(3-hydroxypropylamino)-6-benzylamino-9-isopropylpurine], results in a compound, cis-[Pt(boh)2Cl2] (C1), with the unique anticancer profile which may be associated with some features of the damaged DNA and/or its cellular processing (Travnicek Z et al. (2003) J Inorg Biochem94, 307-316; Liskova B (2012) Chem Res Toxicol25, 500-509). A combination of biochemical and molecular biology techniques was used to establish mechanistic differences between cisplatin and C1 with respect to the DNA damage they produce and their interactions with critical DNA-binding proteins, DNA-processing enzymes and glutathione. The results show that replacement of the NH3 groups in cisplatin by bohemine modulates some aspects of the mechanism of action of C1. More specifically, the results of the present work are consistent with the thesis that, in comparison with cisplatin, effects of other factors, such as: (i) slower rate of initial binding of C1 to DNA; (ii) the lower efficiency of C1 to form bifunctional adducts; (iii) the reduced bend of longitudinal DNA axis induced by the major 1,2-GG intrastrand cross-link of C1; (iv) the reduced affinity of HMG domain proteins to the major adduct of C1; (v) the enhanced efficiency of the DNA adducts of C1 to block DNA polymerization and to inhibit transcription activity of human RNA pol II and RNA transcription; (vi) slower rate of the reaction of C1 with glutathione, may partially contribute to the unique activity of C1.


Subject(s)
Antineoplastic Agents/pharmacology , Organoplatinum Compounds/pharmacology , Purines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , CDC2 Protein Kinase/antagonists & inhibitors , DNA/drug effects , DNA/metabolism , DNA Damage , Dose-Response Relationship, Drug , HeLa Cells , Humans , Molecular Conformation , Molecular Structure , Organoplatinum Compounds/chemical synthesis , Organoplatinum Compounds/chemistry , Polymerization/drug effects , Purines/chemistry , RNA Polymerase II/antagonists & inhibitors , RNA Polymerase II/genetics , Structure-Activity Relationship , Transcription, Genetic/drug effects
11.
J Inorg Biochem ; 114: 15-23, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22687561

ABSTRACT

Design of new antitumor Pt drugs is currently also focused on those new Pt complexes which form on DNA major adducts that can hardly be removed by DNA repair systems. An attempt of this kind has already been done by designing and synthesizing new antitumor azolato-bridged dinuclear Pt(II) complexes, such as [{cis-Pt(NH(3))(2)}(2)(µ-OH)(µ-pyrazolate)](2+) (AMPZ). This new Pt(II) complex exhibits markedly higher toxic effects in some tumor cell lines than conventional mononuclear cisplatin. The primary objective in the present study was to further delineate differences in the interactions of AMPZ and cisplatin with natural, high-molecular-mass DNA using a combination of biochemical and molecular biophysics techniques. The results demonstrate for the first time that little conformational distortions induced by AMPZ in highly polymeric DNA with a random nucleotide sequence represent a structural motif recognizable by DNA repair systems less efficiently than distortions induced by cisplatin. Thus, DNA adducts of azolato-bridged dinuclear Pt(II) complexes can escape repair mechanisms more easily than those of cisplatin, which may potentiate antitumor effects of these new metallodrugs in cancer cells.


Subject(s)
Antineoplastic Agents/chemical synthesis , DNA Adducts/chemistry , DNA Repair/drug effects , Organoplatinum Compounds/chemical synthesis , Platinum/chemistry , Pyrazoles/chemical synthesis , Antineoplastic Agents/pharmacology , Base Sequence , Cell Survival/drug effects , Cisplatin/pharmacology , DNA Damage , HeLa Cells , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Nucleic Acid Denaturation , Organoplatinum Compounds/pharmacology , Plasmids , Pyrazoles/pharmacology , Spectrometry, Fluorescence , Thiourea/chemistry
12.
J Inorg Biochem ; 108: 69-79, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22019433

ABSTRACT

Platinum diamine complexes are able to crosslink the guanines of d(GC)(2) dinucleotides within double-stranded DNA. The interstrand crosslink thus formed causes a bend of the double helix toward the minor groove and the helical sense changes locally to left-handed, resulting in a considerable unwinding. The bend and unwinding angles have been shown to depend on the platinum ligands. Here, we have used molecular dynamics simulations to investigate the DNA 20-mer d(C(1)T(2)C(3)T(4)C(5)C(6)T(7)T(8)G*(9)C(10)T(11)C(12)T(13)C(14)C(15)T(16)T(17)C(18)T(19)C(20))-d(G(21)A(22)G(23)A(24)A(25)G(26)G(27)A(28)G(29)A(30)G*(31)C(32)A(33)A(34)G(35)G(36)A(37)G(38)A(39)G(40)) with the G* guanines crosslinked by cis-Pt(NH(3))(2)(2+), Pt(R,R-DACH)(2+), or Pt(S,S-DACH)(2+). Previous investigations on cisplatin interstrand adducts indicated that the structure is similar in solid state and in solution; thus, we used the reported X-ray structure of a cisplatin adduct as a starting model. Replacing in the MD-relaxed model for the DNA duplex crosslinked with cis-Pt(NH(3))(2)(2+) the two NH(3) platinum ligands by R,R-DACH or S,S-DACH led to clashes between the DACH residue and the deoxyribose of C(12). Confrontation of MD-derived models with gel shift measurements suggested that these clashes are avoided differently in the adducts of Pt(R,R-DACH)(2+)versus Pt(S,S-DACH)(2+). The R,R-isomer avoids the clash by untwisting the T(11)/A(30)-C(12)/G(29) step, thus increasing the global unwinding. In contrast, the S,S-isomer modifies the shift and slide parameters of this step, which dislocates the helical axis and enhances the bend angle. The clash that leads to the differentiation of the structures as a function of the diamine ligand is related to a hydrogen bond between the platinum complex and the T(11) base and could be characteristic of interstrand crosslinks at d(pyG*Cpy)-d(puG*Cpu) sequences.


Subject(s)
DNA/chemistry , Organoplatinum Compounds/chemistry , Molecular Dynamics Simulation , Molecular Structure , Nucleic Acid Conformation , Platinum/chemistry
13.
Mol Pharm ; 8(6): 2368-78, 2011 Dec 05.
Article in English | MEDLINE | ID: mdl-22050363

ABSTRACT

The new trinuclear tridentate Pt(II) complex [Pt(3)Cl(3)(hptab)](3+) (1; hptab = N,N,N',N',N'',N''-hexakis(2-pyridylmethyl)-1,3,5-tris(aminomethyl)benzene) exhibits promising cytotoxic effects in human and mouse tumor cells including those resistant to conventional cisplatin (Dalton Trans. 2006, 2617; Chem. Eur. J. 2009, 15, 5245). The present study is focused on the molecular pharmacology of 1, in particular on its interactions with DNA (which is the major pharmacological target of platinum antitumor drugs), to elucidate more deeply the mechanism underlying its antitumor effects. Results obtained with the aid of methods of molecular biophysics and pharmacology reveal new details of DNA modifications by 1. Complex 1 binds to DNA forming in the absence of proteins and molecular crowding agents mainly trifunctional intrastrand cross-links. In these DNA adducts all three Pt(II) centers of 1 are coordinated to DNA base residues, which leads to extensive conformational alterations in DNA. An intriguing aspect of the DNA-binding mode of this trinuclear Pt(II) complex 1 is that it can cross-link proteins to DNA. Even more interestingly, 1 can cross-link in the presence of molecular crowding agent, which mimics environmental conditions in cell nucleus, two DNA duplexes in a high yield--a feature observed for the first time for antitumor trinuclear platinum complexes. Thus, the concept for the design of agents capable of forming intramolecular tridentate DNA adducts, DNA-protein and interduplex DNA-DNA cross-links based on trinuclear tridentate Pt(II) complexes with semirigid aromatic linkers may result in new compounds which exhibit a variety of biological effects and can be also useful in nucleic acids research.


Subject(s)
Antineoplastic Agents , DNA/chemistry , Organoplatinum Compounds/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/metabolism , Humans , Mice , Molecular Structure , Organoplatinum Compounds/metabolism
14.
Chem Res Toxicol ; 23(11): 1833-42, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20939593

ABSTRACT

cis-Amminedichlorido(cyclohexylamine)platinum(II) (JM118) is an antitumor Pt(II) analogue of cisplatin exhibiting considerably higher activity than cisplatin in human tumor cells. JM118 is also the major metabolite of the first orally administered Pt(IV) drug satraplatin. In an effort to design improved platinum antitumor agents, it is important to elucidate the biochemical factors that affect the cytotoxic properties of existing platinum drugs. Since DNA is considered the major pharmacological target of platinum drugs, the objective in the present work was to understand more fully the DNA binding mode of antitumor JM118. We examined the rate of aquation of the first chloride of bifunctional JM118 and found that it was considerably lower than that of cisplatin; consequently, the rate of the reaction of JM118 with DNA was lower compared to cisplatin. The influence of global modification by JM118 and its major site-specific adducts on DNA conformation by biochemical methods was investigated as well. While examination of the global modification revealed in several cases no substantial differences in the lesions induced by JM118 and cisplatin, DNA bending due to the 1,2-GG intrastrand adduct of JM118 was lower than that of cisplatin. The bending angles afforded by the adducts of JM118 were only slightly affected by the orientation of the cyclohexylamine ligand toward the 3' or 5' direction of the duplex. We also used in vitro assays that make it possible to monitor DNA repair synthesis by cell-free extracts and DNA-protein cross-linking to probe properties of DNA adducts of JM118. These results showed a higher DNA-protein cross-linking efficiency of JM118 and a less efficient removal from DNA of the adducts of JM118 in comparison with cisplatin. Thus, the results of the present work provide additional evidence that DNA binding of JM118 is in several aspects different from that of conventional cisplatin.


Subject(s)
Antineoplastic Agents/chemistry , Cisplatin/chemistry , DNA/chemistry , Organoplatinum Compounds/chemistry , Antineoplastic Agents/toxicity , Base Sequence , Cell Line, Tumor , Circular Dichroism , Cisplatin/toxicity , DNA Adducts/chemistry , DNA Repair , Humans , Nucleic Acid Denaturation , Organoplatinum Compounds/toxicity , Transition Temperature
15.
Biochem Pharmacol ; 80(3): 344-51, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20399754

ABSTRACT

The primary objective was to understand more deeply the molecular mechanism underlying different antitumor effects of dinuclear Pt(II) complexes containing aromatic linkers of different length, {[cis-Pt(NH(3))(2)Cl](2)(4,4'-methylenedianiline)}(2+) (1) and {[cis-Pt(NH(3))(2)Cl](2)(alpha,alpha'-diamino-p-xylene)}(2+) (2). These complexes belong to a new generation of promising polynuclear platinum drugs resistant to decomposition by sulfur nucleophiles which hampers clinical use of bifunctional polynuclear trans Pt(II) complexes hitherto tested. Results obtained with the aid of methods of molecular biophysics and pharmacology reveal differences and new details of DNA modifications by 1 and 2 and recognition of these modifications by cellular components. The results indicate that the unique properties of DNA interstrand cross-links of this class of polynuclear platinum complexes and recognition of these cross-links may play a prevalent role in antitumor effects of these metallodrugs. Moreover, the results show for the first time a strong specific recognition and binding of high-mobility-group-domain proteins, which are known to modulate antitumor effects of clinically used platinum drugs, to DNA modified by a polynuclear platinum complex.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cisplatin/chemistry , Cisplatin/pharmacology , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/pharmacology , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cisplatin/metabolism , Cross-Linking Reagents/metabolism , DNA Adducts/chemistry , DNA Adducts/metabolism , DNA Damage/drug effects , DNA Damage/physiology , HeLa Cells , Humans
16.
Chemistry ; 16(19): 5744-54, 2010 May 17.
Article in English | MEDLINE | ID: mdl-20376825

ABSTRACT

We studied the thermodynamic properties, conformation, and recognition of DNA duplexes site-specifically modified by monofunctional adducts of Ru(II) complexes of the type [Ru(II)(eta(6)-arene)(Cl)(en)](+), in which arene=para-, meta-, or ortho-terphenyl (complexes 1, 2, and 3, respectively) and en=1,2-diaminoethane. It has been shown (J. Med. Chem. 2008, 51, 5310) that 1 exhibits promising cytotoxic effects in human tumor cells, whereas 2 and 3 are much less cytotoxic; concomitantly with the high cytotoxicity of 1, its DNA binding mode involves combined intercalative and monofunctional (coordination) binding modes, whereas less cytotoxic compounds 2 and 3 bind to DNA only through a monofunctional coordination to DNA bases. An analysis of conformational distortions induced in DNA by adducts of 1 and 2 revealed more extensive and stronger distortion and concomitantly greater thermodynamic destabilization of DNA by the adducts of nonintercalating 2. Moreover, affinity of replication protein A to the DNA duplex containing adduct of 1 was pronouncedly lower than to the adduct of 2. On the other hand, another damaged-DNA-binding protein, xeroderma pigmentosum protein A, did not recognize the DNA adduct of 1 or 2. Importantly, the adducts of 1 induced a considerably lower level of repair synthesis than the adducts of 2, which suggests enhanced persistence of the adducts of the more potent and intercalating 1 in comparison with the adducts of the less potent and nonintercalating 2. Also interestingly, the adducts of 1 inhibited DNA polymerization more efficiently than the adducts of 2, and they could also be bypassed by DNA polymerases with greater difficulty. Results of the present work along with those previously published support the view that monodentate Ru(II) arene complexes belong to a class of anticancer agents for which structure-pharmacological relationships might be correlated with their DNA-binding modes.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , DNA Adducts/chemistry , DNA Repair/drug effects , DNA/chemistry , DNA/drug effects , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Ruthenium/chemistry , Terphenyl Compounds/chemistry , Terphenyl Compounds/pharmacology , Base Sequence , Calorimetry , Hydrogen Bonding , Molecular Structure , Nucleic Acid Conformation , Protein Binding
17.
Biochem Pharmacol ; 79(4): 552-64, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19782655

ABSTRACT

Earlier studies have described promising antitumor activity of a large-ring chelate complex [PtCl(2)(cis-1,4-DACH)] (DACH=diaminocyclohexane). Encouraging antitumor activity of this analogue of cisplatin prompted us to perform studies focused on the mechanistic basis of pharmacological effects of this complex. Four early steps in the mechanism of biological activity of cisplatin have been delineated: cell entry, reactions with sulfur-containing compounds, platinum-DNA binding along with processing platinated DNA by proteins (enzymes) and DNA repair. Here, we describe comparative experiments (involving also cisplatin) revealing: (i) improved cytotoxicity (3.4-5.4-fold) of [PtCl(2)(cis-1,4-DACH)] in human tumor ovarian cell lines; (ii) enhanced cellular uptake (approximately 1.5-fold) of [PtCl(2)(cis-1,4-DACH)]; (iii) somewhat enhanced rate of reactions of [PtCl(2)(cis-1,4-DACH)] with glutathione (approximately 1.5-fold), but a similar rate of reactions with metallothionenin-2; (iv) enhanced rate of DNA binding of [PtCl(2)(cis-1,4-DACH)] in cell-free media (approximately 2-fold); (v) similar sequence preference of DNA binding of [PtCl(2)(cis-1,4-DACH)] in cell-free media; (vi) identical DNA interstrand cross-linking efficiency (6%); (vii) similar bending (32 degrees) and enhanced local unwinding (approximately 1.5-fold) induced in DNA by the major 1,2-GG-intrastrand cross-link; (viii) markedly enhanced inhibiting effects of DNA adducts of [PtCl(2)(cis-1,4-DACH)] on processivity of DNA polymerase; and (ix) a slightly lower efficiency of DNA repair systems to remove the adducts of [PtCl(2)(cis-1,4-DACH)] from DNA.


Subject(s)
Antineoplastic Agents/metabolism , Cyclohexylamines/metabolism , DNA Repair/drug effects , DNA, Neoplasm/metabolism , Glutathione/metabolism , Organoplatinum Compounds/metabolism , Organoplatinum Compounds/toxicity , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cattle , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cyclohexylamines/chemistry , Cyclohexylamines/pharmacology , DNA Repair/physiology , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Carriers/toxicity , HeLa Cells , Humans , Ligands , Platinum Compounds/chemistry , Platinum Compounds/metabolism , Platinum Compounds/toxicity , Rabbits
18.
Biochem Pharmacol ; 79(2): 112-21, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19682435

ABSTRACT

Reported herein is a detailed biochemical and molecular biophysics study of the molecular mechanism of action of antitumor dinuclear Pt(II) complex [{PtCl(DACH)}(2)-mu-Y](4+) [DACH=1,2-diaminocyclohexane, Y=H(2)N(CH(2))(6)NH(2)(CH(2))(2)NH(2)(CH(2))(6)NH(2)] (complex 1). This new, long-chain bifunctional dinuclear Pt(II) complex is resistant to metabolic decomposition by sulfur-containing nucleophiles. The results show that DNA adducts of 1 can largely escape repair and yet inhibit very effectively transcription so that they should persist longer than those of conventional cisplatin. Hence, they could trigger a number of downstream cellular effects different from those triggered in cancer cells by DNA adducts of cisplatin. This might lead to the therapeutic effects that could radically improve chemotherapy by platinum complexes. In addition, the findings of the present work make new insights into mechanisms associated with antitumor effects of dinuclear/trinuclear Pt(II) complexes possible.


Subject(s)
Antineoplastic Agents/pharmacology , DNA/chemistry , Nucleic Acid Conformation , Organoplatinum Compounds/pharmacology , Sulfur/chemistry , Antineoplastic Agents/chemistry , Base Sequence , Cell-Free System , DNA Repair , Fluorescence , Glutathione/chemistry , Molecular Sequence Data , Organoplatinum Compounds/chemistry
19.
J Biol Inorg Chem ; 14(1): 75-87, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18777181

ABSTRACT

The global modification of mammalian and plasmid DNAs by the novel platinum compounds cis-[PtCl(2)(isopropylamine)(1-methylimidazole)] and trans-[PtCl(2)(isopropylamine)(1-methylimidazole)] and the reactivity of these compounds with reduced glutathione (GSH) were investigated in cell-free media using various biochemical and biophysical methods. Earlier cytotoxicity studies had revealed that the replacement of the NH(3) groups in cisplatin by the azole and isopropylamine ligands lowers the activity of cisplatin in both sensitive and resistant cell lines. The results of the present work show that this replacement does not considerably affect the DNA modifications by this drug, recognition of these modifications by HMGB1 protein, their repair, and reactivity of the platinum complex with GSH. These results were interpreted to mean that the reduced activity of this analog of cisplatin in tumor cell lines is due to factors that do not operate at the level of the target DNA. In contrast, earlier studies had shown that the replacement of the NH(3) groups in the clinically ineffective trans isomer (transplatin) by the azole and isopropylamine ligands results in a radical enhancement of its activity in tumor cell lines. Importantly, this replacement also markedly alters the DNA binding mode of transplatin, which is distinctly different from that of cisplatin, but does not affect reactivity with GSH. Hence, the results of the present work are consistent with the view and support the hypothesis systematically tested by us and others that platinum drugs that bind to DNA in a fundamentally different manner from that of conventional cisplatin may have altered pharmacological properties.


Subject(s)
Antineoplastic Agents/chemistry , Culture Media/chemistry , DNA/chemistry , Glutathione/chemistry , Organoplatinum Compounds/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Cell-Free System , Circular Dichroism , DNA/drug effects , Glutathione/drug effects , Humans , Organoplatinum Compounds/pharmacology , Spectrophotometry, Ultraviolet , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...