Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
1.
Proc Natl Acad Sci U S A ; 120(18): e2218033120, 2023 05 02.
Article in English | MEDLINE | ID: mdl-37094129

ABSTRACT

As a decoy receptor, soluble ST2 (sST2) interferes with the function of the inflammatory cytokine interleukin (IL)-33. Decreased sST2 expression in colorectal cancer (CRC) cells promotes tumor growth via IL-33-mediated bioprocesses in the tumor microenvironment. In this study, we discovered that hypoxia reduced sST2 expression in CRC cells and explored the associated molecular mechanisms, including the expression of key regulators of ST2 gene transcription in hypoxic CRC cells. In addition, the effect of the recovery of sST2 expression in hypoxic tumor regions on malignant progression was investigated using mouse CRC cells engineered to express sST2 in response to hypoxia. Our results indicated that hypoxia-dependent increases in nuclear IL-33 interfered with the transactivation activity of GATA3 for ST2 gene transcription. Most importantly, hypoxia-responsive sST2 restoration in hypoxic tumor regions corrected the inflammatory microenvironment and suppressed tumor growth and lung metastasis. These results indicate that strategies targeting sST2 in hypoxic tumor regions could be effective for treating malignant CRC.


Subject(s)
Colorectal Neoplasms , Interleukin-33 , Animals , Mice , Interleukin-33/metabolism , Down-Regulation , Interleukin-1 Receptor-Like 1 Protein/metabolism , Cell Nucleus/metabolism , Colorectal Neoplasms/genetics , Tumor Microenvironment , GATA3 Transcription Factor/metabolism
2.
Int J Cancer ; 152(5): 962-976, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36214789

ABSTRACT

Cervical cancer remains a major threat to women's health, especially in countries with limited medical resources, and new drugs are needed to improve patient survival and minimize adverse effects. Here, we examine the effects of a triphenylphosphonium (TPP)-conjugated pyrrole-imidazole polyamide (CCC-h1005) targeting the common homoplasmic mitochondrial DNA (mtDNA) cancer risk variant (ATP6 8860A>G) on the survival of cervical cancer cell lines, cisplatin-resistant HeLa cells and patient-derived cervical clear cell carcinoma cells as models of cervical cancer treatment. We found that CCC-h1005 induced death in these cells and suppressed the growth of xenografted HeLa tumors with no severe adverse effects. These results suggest that PIP-TPP designed to target mtDNA cancer risk variants can be used to treat many cervical cancers harboring high copies of the target variant, providing a foundation for clinical trials of this class of molecules for treating cervical cancer and other types of cancers.


Subject(s)
Uterine Cervical Neoplasms , Humans , Female , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Nylons/pharmacology , DNA, Mitochondrial/genetics , HeLa Cells , Pyrroles/pharmacology , Imidazoles/pharmacology
3.
Cancer Med ; 12(5): 5821-5832, 2023 03.
Article in English | MEDLINE | ID: mdl-36262061

ABSTRACT

BACKGROUND: Activating mutations of the KRAS occurs in >90% of pancreatic ductal adenocarcinoma (PDAC) cases. However, direct pharmacological targeting of the activated KRAS protein has been challenging. We previously reported that KR12, a DNA-alkylating pyrrole-imidazole polyamide designed to recognize the KRAS G12D/V mutation, showed an anti-tumor effect in colorectal cancer. In this study, we evaluated the anti-tumor effect of KR12 in PDAC. METHODS: KR12 was synthesized by an automated peptide synthesizer PSSM-8 and tested for anti-tumor effect in PDAC mouse models. RESULT: KR12 inhibited tumor growth in a spontaneous PDAC mouse model, although the anti-tumor activity appeared to be limited in a human PDAC xenograft model. We developed a pyrrole-imidazole polyamide screening process based on the hypothesis that genetic elements otherwise unaffected by KR12 could exert attenuating effects on KRAS-suppression-resistant PDAC. We identified RAD51 as a potential therapeutic target in human PDAC cells. A RAD51 inhibitor showed an inhibitory effect on cell growth and affected the cytotoxic activity of KR12 in PDAC cells. CONCLUSION: These data suggested that the simultaneous inhibition of RAD51 and mutant KRAS blockage would be an important therapeutic strategy for PDAC.


Subject(s)
Antineoplastic Agents , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Mice , Humans , Nylons/pharmacology , Nylons/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , DNA/therapeutic use , Imidazoles/pharmacology , Imidazoles/therapeutic use , Pancreatic Neoplasms
4.
Cancer Sci ; 113(4): 1321-1337, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35112436

ABSTRACT

Certain somatic mutations in mtDNA were associated with tumor progression and frequently found in a homoplasmic state. We recently reported that pyrrole-imidazole polyamide conjugated with the mitochondria-delivering moiety triphenylphosphonium (PIP-TPP) targeting an mtDNA mutation efficiently induced apoptosis in cancer cells with the mutation but not normal cells. Here, we synthesized the novel PIP-TPP, CCC-021-TPP, targeting ND6 14582A > G homoplasmic missense mutation that is suggested to enhance metastasis of non-small-cell lung cancer A549 cells. CCC-021-TPP did not induce apoptosis but caused cellular senescence in the cells, accompanied by a significant induction of antiapoptotic BCL-XL. Simultaneous treatment of A549 cells with CCC-021-TPP and the BCL-XL selective inhibitor A-1155463 resulted in apoptosis induction. Importantly, the combination induced apoptosis and suppressed tumor growth in an A549 xenografted model. These results highlight the potential of anticancer therapy with PIP-TPPs targeting mtDNA mutations to induce cell death even in apoptosis-resistant cancer cells when combined with senolytics.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Apoptosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , DNA, Mitochondrial/genetics , Humans , Imidazoles , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Nylons/pharmacology , Pyrroles/pharmacology , Pyrroles/therapeutic use , Senotherapeutics
5.
BMC Mol Cell Biol ; 22(1): 52, 2021 Oct 07.
Article in English | MEDLINE | ID: mdl-34615464

ABSTRACT

BACKGROUND: Mitochondrial DNA (mtDNA) carrying certain pathogenic mutations or single nucleotide variants (SNVs) enhances the invasion and metastasis of tumor cells, and some of these mutations are homoplasmic in tumor cells and even in tumor tissues. On the other hand, intercellular transfer of mitochondria and cellular components via extracellular vesicles (EVs) and tunneling nanotubes (TNTs) has recently attracted intense attention in terms of cell-to-cell communication in the tumor microenvironment. It remains unclear whether metastasis-enhancing pathogenic mutant mtDNA in tumor cells is intercellularly transferred between tumor cells and stromal cells. In this study, we investigated whether mtDNA with the NADH dehydrogenase subunit 6 (ND6) G13997A pathogenic mutation in highly metastatic cells can be horizontally transferred to low-metastatic cells and stromal cells in the tumor microenvironment. RESULTS: When MitoTracker Deep Red-labeled high-metastatic Lewis lung carcinoma A11 cells carrying the ND6 G13997A mtDNA mutation were cocultured with CellLight mitochondria-GFP-labeled low-metastatic P29 cells harboring wild-type mtDNA, bidirectional transfer of red- and green-colored vesicles, probably mitochondria-related EVs, was observed in a time-dependent manner. Similarly, intercellular transfer of mitochondria-related EVs occurred between A11 cells and α-smooth muscle actin (α-SMA)-positive cancer-associated fibroblasts (CAFs, WA-mFib), macrophages (RAW264.7) and cytotoxic T cells (CTLL-2). Intercellular transfer was suppressed by inhibitors of EV release. The large and small EV fractions (L-EV and S-EV, respectively) prepared from the conditioned medium by differential ultracentrifugation both were found to contain mtDNA, although only S-EVs were efficiently incorporated into the cells. Several subpopulations had evidence of LC3-II and contained degenerated mitochondrial components in the S-EV fraction, signaling to the existence of autophagy-related S-EVs. Interestingly, the S-EV fraction contained a MitoTracker-positive subpopulation, which was inhibited by the respiration inhibitor antimycin A, indicating the presence of mitochondria with membrane potential. It was also demonstrated that mtDNA was transferred into mtDNA-less ρ0 cells after coculture with the S-EV fraction. In syngeneic mouse subcutaneous tumors formed by a mixture of A11 and P29 cells, the mitochondria-related EVs released from A11 cells reached distantly positioned P29 cells and CAFs. CONCLUSIONS: These results suggest that metastasis-enhancing pathogenic mtDNA derived from metastatic tumor cells is transferred to low-metastatic tumor cells and stromal cells via S-EVs in vitro and in the tumor microenvironment, inferring a novel mechanism of enhancement of metastatic potential during tumor progression.


Subject(s)
DNA, Mitochondrial , Extracellular Vesicles , Animals , Cell Membrane Structures , DNA, Mitochondrial/genetics , Extracellular Vesicles/metabolism , Mice , Mitochondria/genetics , Mutation , Nanotubes , Stromal Cells
6.
Biochem Biophys Res Commun ; 576: 93-99, 2021 10 22.
Article in English | MEDLINE | ID: mdl-34482029

ABSTRACT

Somatic mutations in mitochondrial DNA may provide a new avenue for cancer therapy due to their associations to a number of cancers and a tendency of homoplasmicity. In consideration of mitochondrial features and its relatively small genome size, a nucleotide-based targeting approach is a considerably more promising option. To explore the efficacy of short linear N-methylpyrrole-N-methylimidazole polyamide (PI polyamide), we synthesized a five-ring short PI polyamide that provided sequence-specific homing for the A3243G mitochondrial mutation upon conjugation with triphenylphosphonium cation (TPP). This PI polyamide-TPP was able to induce cytotoxicity in HeLamtA3243G cybrid cells, while preserving preferential binding for oligonucleotides containing the A3243G motif from melting temperature assays. The PI polyamide-TPP also localized in the mitochondria in HeLamtA3243G cells and induced mitochondrial reactive oxygen species production, mitophagy and apoptosis in a mutation-specific fashion compared to the wild-type HeLamtHeLa cybrids; normal human dermal fibroblasts were also relatively unaffected to suggest discriminating selectivity for the mutant mitochondria, offering a novel outlook for cancer therapy via mitochondrial homing of short linear PIP-TPPs.


Subject(s)
Antineoplastic Agents/pharmacology , DNA, Mitochondrial/drug effects , Imidazoles/chemistry , Mutation , Nylons/chemistry , Organoselenium Compounds/chemistry , Pyrroles/chemistry , Uterine Cervical Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Apoptosis/physiology , DNA, Mitochondrial/genetics , Female , HeLa Cells , Humans , Mitophagy/physiology , Reactive Oxygen Species/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
7.
Cancer Sci ; 112(12): 4834-4843, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34533888

ABSTRACT

As the energy factory for the cell, the mitochondrion, through its role of adenosine triphosphate production by oxidative phosphorylation, can be regarded as the guardian of well regulated cellular metabolism; the integrity of mitochondrial functions, however, is particularly vulnerable in cancer due to the lack of superstructures such as histone and lamina folds to protect the mitochondrial genome from unintended exposure, which consequently elevates risks of mutation. In cancer, mechanisms responsible for enforcing quality control surveillance for identifying and eliminating defective mitochondria are often poorly regulated, and certain uneliminated mitochondrial DNA (mtDNA) mutations and polymorphisms can be advantageous for the proliferation, progression, and metastasis of tumor cells. Such pathogenic mtDNA aberrations are likely to increase and occasionally be homoplasmic in cancer cells and, intriguingly, in normal cells in the proximity of tumor microenvironments as well. Distinct characteristics of these abnormalities in mtDNA may provide a new path for cancer therapy. Here we discuss a promising novel therapeutic strategy, using the sequence-specific properties of pyrrole-imidazole polyamide-triphenylphosphonium conjugates, against cancer for clearing abnormal mtDNA by reactivating mitochondrial quality control surveillance.


Subject(s)
Mitochondria/genetics , Neoplasms/genetics , Organophosphorus Compounds/pharmacology , Genome, Mitochondrial/drug effects , Humans , Mitochondria/drug effects , Molecular Targeted Therapy , Mutation , Neoplasms/drug therapy , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/therapeutic use
8.
Sci Rep ; 11(1): 13302, 2021 06 25.
Article in English | MEDLINE | ID: mdl-34172808

ABSTRACT

Pathogenic mitochondrial NADH dehydrogenase (ND) gene mutations enhance the invasion and metastasis of various cancer cells, and they are associated with metastasis in human non-small cell lung cancer (NSCLC). Moreover, monocarboxylate transporter 4 (MCT4) is overexpressed in solid cancers and plays a role in cancer cell proliferation and survival. Here, we report that MCT4 is exclusively expressed in mouse transmitochondrial cybrids with metastasis-enhancing pathogenic ND6 mutations. A high level of MCT4 is also detected in human NSCLC cell lines and tissues predicted to carry pathogenic ND mutations and is associated with poor prognosis in NSCLC patients. MCT4 expression in the cell lines is suppressed by N-acetyl-L-cysteine. Phosphatidylinositol-3 kinase (PI3K), AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin (mTOR) are involved in the regulation of MCT4 expression in the transmitochondrial cybrid cells. An MCT1/4 inhibitor effectively kills NSCLC cells with predicted pathogenic ND mutations, but an MCT1/2 inhibitor does not have the same effect. Thus, MCT4 expression is augmented by pathogenic ND mutations and could be a biomarker and a therapeutic target in pathogenic ND mutation-harbouring metastatic tumours.


Subject(s)
Monocarboxylic Acid Transporters/genetics , Muscle Proteins/genetics , Mutation/genetics , NADH Dehydrogenase/genetics , A549 Cells , Animals , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Humans , Lung Neoplasms/genetics , Male , Mice , Mice, Inbred C57BL
9.
Cancer Sci ; 112(6): 2504-2512, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33811417

ABSTRACT

Mitochondrial DNA (mtDNA) mutations occur frequently in cancer cells, and some of them are often homoplasmic. Targeting such mtDNA mutations could be a new method for killing cancer cells with minimal impact on normal cells. Pyrrole-imidazole polyamides (PIPs) are cell-permeable minor groove binders that show sequence-specific binding to double-stranded DNA and inhibit the transcription of target genes. PIP conjugated with the lipophilic triphenylphosphonium (TPP) cation can be delivered to mitochondria without uptake into the nucleus. Here, we investigated the feasibility of the use of PIP-TPP to target a mtDNA mutation in order to kill cancer cells that harbor the mutation. We synthesized hairpin-type PIP-TPP targeting the A3243G mutation and examined its effects on the survival of HeLa cybrid cells with or without the mutation (HeLamtA3243G cells or HeLamtHeLa cells, respectively). A surface plasmon resonance assay demonstrated that PIP-TPP showed approximately 60-fold higher binding affinity for the mutant G-containing synthetic double-stranded DNA than for the wild-type A-containing DNA. When added to cells, it localized in mitochondria and induced mitochondrial reactive oxygen species production, extensive mitophagy, and apoptosis in HeLamtA3243G cells, while only slightly exerting these effects in HeLamtHeLa cells. These results suggest that PIP-TPPs targeting mtDNA mutations could be potential chemotherapeutic drugs to treat cancers without severe adverse effects.


Subject(s)
DNA, Mitochondrial/drug effects , Imidazoles/pharmacology , Mitochondria/genetics , Neoplasms/genetics , Pyrroles/chemistry , Sulfonium Compounds/chemistry , Cell Survival/drug effects , DNA, Mitochondrial/genetics , HeLa Cells , Humans , Imidazoles/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , Mitophagy , Mutation , Neoplasms/drug therapy , Neoplasms/metabolism , Nylons/chemistry , Reactive Oxygen Species/metabolism , Surface Plasmon Resonance
10.
Sci Rep ; 11(1): 2923, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33536560

ABSTRACT

The antidiabetic adiponectin receptor agonist AdipoRon has been shown to suppress the tumour growth of human pancreatic cancer cells. Because obesity and diabetes affect pancreatic cancer progression and chemoresistance, we investigated the effect of AdipoRon on orthotopic tumour growth of Panc02 pancreatic cancer cells in DIO (diet-induced obese) prediabetic mice. Administration of AdipoRon into DIO mice fed high-fat diets, in which prediabetic conditions were alleviated to some extent, did not reduce either body weight or tumour growth. However, when the DIO mice were fed low-fat diets, body weight and the blood leptin level gradually decreased, and importantly, AdipoRon became effective in suppressing tumour growth, which was accompanied by increases in necrotic areas and decreases in Ki67-positive cells and tumour microvessels. AdipoRon inhibited cell growth and induced necrotic cell death of Panc02 cells and suppressed angiogenesis of endothelial MSS31 cells. Insulin and IGF-1 only slightly reversed the AdipoRon-induced suppression of Panc02 cell survival but had no effect on the AdipoRon-induced suppression of MSS31 cell angiogenesis. Leptin significantly ameliorated AdipoRon-induced suppression of angiogenesis through inhibition of ERK1/2 activation. These results suggest that obesity-associated factors weaken the anticancer effect of AdipoRon, which indicates the importance of weight loss in combating pancreatic cancer.


Subject(s)
Obesity/complications , Pancreatic Neoplasms/drug therapy , Piperidines/pharmacology , Receptors, Adiponectin/antagonists & inhibitors , Animals , Cell Line, Tumor/transplantation , Diet, High-Fat/adverse effects , Disease Models, Animal , Drug Resistance, Neoplasm , Humans , Male , Mice , Obesity/metabolism , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/pathology , Piperidines/therapeutic use , Receptors, Adiponectin/metabolism
11.
Cancer Sci ; 112(3): 1141-1149, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33377228

ABSTRACT

PIK3CA is the most frequently mutated oncogene in cervical cancer, and somatic mutations in the PIK3CA gene result in increased activity of PI3K. In cervical cancer, the E545K mutation in PIK3CA leads to elevated cell proliferation and reduced apoptosis. In the present study, we designed and synthesized a novel pyrrole-imidazole polyamide-seco-CBI conjugate, P3AE5K, to target the PIK3CA gene bearing the E545K mutation, rendered possible by nuclear access and the unique sequence specificity of pyrrole-imidazole polyamides. P3AE5K interacted with double-stranded DNA of the coding region containing the E545K mutation. When compared with conventional PI3K inhibitors, P3AE5K demonstrated strong cytotoxicity in E545K-positive cervical cancer cells at lower concentrations. PIK3CA mutant cells exposed to P3AE5K exhibited reduced expression levels of PIK3CA mRNA and protein, and subsequent apoptotic cell death. Moreover, P3AE5K significantly decreased the tumor growth in mouse xenograft models derived from PIK3CA mutant cells. Overall, the present data strongly suggest that the alkylating pyrrole-imidazole polyamide P3AE5K should be a promising new drug candidate targeting a constitutively activating mutation of PIK3CA in cervical cancer.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Uterine Cervical Neoplasms/drug therapy , Animals , Antineoplastic Agents, Alkylating/chemical synthesis , Antineoplastic Agents, Alkylating/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Class I Phosphatidylinositol 3-Kinases/genetics , Female , Gain of Function Mutation , Humans , Imidazoles/chemical synthesis , Imidazoles/pharmacology , Imidazoles/therapeutic use , Mice , Nylons/chemical synthesis , Nylons/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/therapeutic use , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Pyrroles/therapeutic use , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Xenograft Model Antitumor Assays
12.
PLoS One ; 15(4): e0232230, 2020.
Article in English | MEDLINE | ID: mdl-32340025

ABSTRACT

BACKGROUND: Proinflammatory interleukin-33 (IL-33) binds to its receptor ST2L and is involved in inflammation and the malignant behavior of cancer cells. However, the role of IL-33-ST2L and the IL-33 decoy receptor sST2 in the tumor microenvironment of pancreatic cancer is unclear. Because we previously reported that sST2 derived from colon cancer cells profoundly influences malignant tumor growth, we hypothesized that sST2 released from pancreatic cancer cells also modulates IL-33-ST2L signaling in the tumor microenvironment, thereby influencing tumor growth. METHODS: ST2 (ST2L and sST2) expression in mouse pancreatic cancer Panc02 cells was downregulated by shRNAs. mRNA expression levels of IL-33, ST2, cytokines and chemokines in the cells and tumor tissues were examined using real-time PCR. sST2 secretion and the amount of CXCL3 in tumor tissues were measured using ELISA. Tumor growth was investigated after injection of the cells into the pancreas of C57BL/6 mice. MPO+, F4/80+ and CD20+ cells in tumor tissues were detected using immunohistochemistry. RESULTS: Some but not all human and mouse pancreatic cancer cell lines preferentially expressed sST2. Then, we investigated the role of sST2 in orthotopic tumor growth of sST2-expressing mouse pancreatic cancer Panc02 cells in immunocompetent mice. shRNA-mediated knockdown of sST2 expression in the cells suppressed orthotopic tumor growth, which was partially recovered by overexpression of shRNA-resistant sST2 mRNA but was not evident in IL-33 knockout mice. This was associated with decreases in Cxcl3 expression, vessel density and accumulation of cancer-associated neutrophils but not cancer-associated macrophages. Administration of SB225002, an inhibitor of the CXCL3 receptor CXCR2, induced similar effects. CONCLUSIONS: Cancer cell-derived sST2 enhances tumor growth through upregulation of CXCL3 via inhibition of IL-33-ST2L signaling in the tumor microenvironment of pancreatic cancer. These results suggest that the sST2 and the CXCL3-CXCR2 axis could be therapeutic targets.


Subject(s)
Cell Proliferation/physiology , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PC-3 Cells , Pancreas/metabolism , Pancreas/pathology , Receptors, Interleukin-1/metabolism , Signal Transduction/physiology , Tumor Microenvironment/physiology , Up-Regulation/physiology
13.
PLoS One ; 14(4): e0215247, 2019.
Article in English | MEDLINE | ID: mdl-30964912

ABSTRACT

In the search for new pharmaceutical leads, especially with DNA-binding molecules or genome editing methods, the issue of side and off-target effects have always been thorny in nature. A particular case is the investigation into the off-target effects of N-methylpyrrole-N-methylimidazole polyamides, a naturally inspired class of DNA binders with strong affinity to the minor-groove and sequence specificity, but at < 20 bases, their relatively short motifs also insinuate the possibility of non-unique genomic binding. Binding at non-intended loci potentially lead to the rise of off-target effects, issues that very few approaches are able to address to-date. We here report an analytical method to infer off-target binding, via expression profiling, based on probing the relative impact to various biochemical pathways; we also proposed an accompanying side effect prediction engine for the systematic screening of candidate polyamides. This method marks the first attempt in PI polyamide research to identify elements in biochemical pathways that are sensitive to the treatment of a candidate polyamide as an approach to infer possible off-target effects. Expression changes were then considered to assess possible outward phenotypic changes, manifested as side effects, should the same PI polyamide candidate be administered clinically. We validated some of these effects with a series of animal experiments, and found agreeable corroboration in certain side effects, such as changes in aspartate transaminase levels in ICR and nude mice post-administration.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Metabolic Networks and Pathways/drug effects , Nylons/metabolism , Nylons/pharmacology , Algorithms , Animals , Binding Sites/genetics , Biochemical Phenomena , Cell Line , DNA/genetics , DNA/metabolism , Drug Discovery , Female , Gene Editing/methods , Gene Expression Profiling , Humans , Imidazoles/metabolism , Imidazoles/pharmacology , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred ICR , Mice, Nude , Oligonucleotide Array Sequence Analysis , Pyrroles/metabolism , Pyrroles/pharmacology
14.
Cancer Res ; 79(4): 830-840, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30584073

ABSTRACT

Amplification of MYCN plays a pivotal role in multiple types of tumors and correlates with poor prognosis in high-risk neuroblastoma. Despite recent advances in the treatment of neuroblastoma, no approaches directly target the master oncogene MYCN. Difficulties in targeting the MYCN protein inspired us to develop a new gene-level-inhibitory strategy using a sequence-specific gene regulator. Here, we generated a MYCN-targeting pyrrole-imidazole (PI) polyamide, MYCN-A3, which directly binds to and alkylates DNA at homing motifs within the MYCN transcript. Pharmacologic suppression of MYCN inhibited the proliferation of cancer cells harboring MYCN amplification compared with MYCN nonamplified cancer cells. In neuroblastoma xenograft mouse models, MYCN-A3 specifically downregulated MYCN expression and suppressed tumor progression with no detectable adverse effects and resulted in prolonged overall survival. Moreover, treatment with MYCN-A3, but not MYCN nontargeting PI polyamide, precipitated a copy number reduction of MYCN in neuroblastoma cells with MYCN amplification. These findings suggest that directly targeting MYCN with MYCN-A3 is a novel therapeutic approach to reduce copy number of the MYCN gene for MYCN-amplified neuroblastoma. SIGNIFICANCE: This study presents a novel approach to drugging an amplified oncogene by showing that targeting gene amplification of MYCN suppresses MYCN expression and neuroblastoma growth.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Gene Amplification/drug effects , Gene Expression Regulation, Neoplastic/drug effects , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Neuroblastoma/prevention & control , Nylons/pharmacology , Alkylation , Animals , Antineoplastic Agents, Alkylating/chemistry , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Female , Humans , Imidazoles/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Neuroblastoma/pathology , Nylons/chemistry , Pyrroles/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Bioorg Med Chem ; 26(9): 2337-2344, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29622411

ABSTRACT

To examine the hydrophobic structure of PI polyamides on tumor accumulation in vivo, PI polyamide-fluorescein conjugates 1-5 with the distinct number of N-methylimidazole (Im) units were synthesized. There existed an inverse relationship between the Im unit number of the compounds and their hydrophobicity. Compound 1 with one Im unit and 3 with three Im units accumulated and retained preferentially in tumor tissues compared to 5 with five Im units. These results suggest the importance of a PI polyamide's primary structure, which partly contributes to its hydrophobic property, on its accumulation and/or retention in tumor tissues in vivo.


Subject(s)
Imidazoles/metabolism , Neoplasms/metabolism , Nylons/metabolism , Pyrroles/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Female , Fluoresceins/chemical synthesis , Fluoresceins/chemistry , Fluoresceins/metabolism , Fluorescence , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Imidazoles/chemical synthesis , Imidazoles/chemistry , Mice, Inbred BALB C , Molecular Structure , Nylons/chemical synthesis , Nylons/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Tissue Distribution
16.
Sci Rep ; 7(1): 15535, 2017 Nov 14.
Article in English | MEDLINE | ID: mdl-29138417

ABSTRACT

Cancer cells have more mutations in their mitochondrial DNA (mtDNA) than do normal cells, and pathogenic mutations in the genes encoding mitochondrial NADH dehydrogenase (ND) subunits have been found to enhance the invasive and metastatic ability of various tumour cells in animal experiments. However, it is unknown whether single-nucleotide variants (SNVs) of the ND genes that decrease complex I activity are involved in distant metastasis in human clinical samples. Here, we demonstrated the enhancement of the distant metastasis of Lewis lung carcinoma cells by the ND6 13885insC mutation, which is accompanied by the overexpression of metastasis-related genes, metabolic reprogramming, the enhancement of tumour angiogenesis and the acquisition of resistance to stress-induced cell death. We then sequenced ND genes in primary tumour lesions with or without distant metastases as well as metastatic tumour lesions from 115 patients with non-small cell lung cancer (NSCLC) and colon cancer, and we subsequently selected 14 SNVs with the potential to decrease complex I activity. Intriguingly, a significant correlation was observed (P < 0.05 by Chi-square test) between the incidence of the selected mutations and distant metastasis. Thus, these results strongly suggest that pathogenic ND gene mutations participate in enhancing distant metastasis in human cancers.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/secondary , Colonic Neoplasms/genetics , Colonic Neoplasms/secondary , Lung Neoplasms/genetics , Lung Neoplasms/pathology , NADH Dehydrogenase/genetics , Adult , Aged , Aged, 80 and over , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , DNA, Mitochondrial/genetics , Female , Genes, Mitochondrial/genetics , Humans , Male , Mice, Inbred C57BL , Middle Aged , Mitochondria/genetics , Mutation , Neoplasm Metastasis , Polymorphism, Single Nucleotide
17.
PLoS One ; 11(10): e0165581, 2016.
Article in English | MEDLINE | ID: mdl-27798693

ABSTRACT

Pyrrole-imidazole polyamides are versatile DNA minor groove binders and attractive therapeutic options against oncological targets, especially upon functionalization with an alkylating agent such as seco-CBI. These molecules also provide an alternative for oncogenes deemed "undruggable" at the protein level, where the absence of solvent-accessible pockets or structural crevices prevent the formation of protein-inhibitor ligands; nevertheless, the genome-wide effect of pyrrole-imidazole polyamide binding remain largely unclear to-date. Here we propose a next-generation sequencing-based workflow combined with whole genome expression arrays to address such issue using a candidate anti-cancer alkylating agent, KR12, against codon 12 mutant KRAS. Biotinylating KR12 enables the means to identify its genome-wide effects in living cells and possible biological implications via a coupled workflow of enrichment-based sequencing and expression microarrays. The subsequent computational pathway and expression analyses allow the identification of its genomic binding sites, as well as a route to explore a polyamide's possible genome-wide effects. Among the 3,343 KR12 binding sites identified in the human LS180 colorectal cancer genome, the reduction of KR12-bound gene expressions was also observed. Additionally, the coupled microarray-sequencing analysis also revealed some insights about the effect of local chromatin structure on pyrrole-imidazole polyamide, which had not been fully understood to-date. A comparative analysis with KR12 in a different human colorectal cancer genome SW480 also showed agreeable agreements of KR12 binding affecting gene expressions. Combination of these analyses thus suggested the possibility of applying this approach to other pyrrole-imidazole polyamides to reveal further biological details about the effect of polyamide binding in a genome.


Subject(s)
Antineoplastic Agents, Alkylating/metabolism , Binding Sites , Colorectal Neoplasms/genetics , Genome, Human , Imidazoles/metabolism , Nylons/metabolism , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Chromatin Immunoprecipitation , Colorectal Neoplasms/metabolism , DNA/chemistry , DNA/genetics , DNA/metabolism , Gene Expression Regulation, Neoplastic/drug effects , High-Throughput Nucleotide Sequencing , Histones/metabolism , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Mutation , Nucleosomes/metabolism , Nucleotide Motifs , Nylons/chemistry , Nylons/pharmacology , Protein Binding , Proto-Oncogene Proteins p21(ras)/genetics , Sequence Analysis, DNA
18.
Nat Commun ; 6: 6706, 2015 Apr 27.
Article in English | MEDLINE | ID: mdl-25913614

ABSTRACT

Despite extensive efforts to target mutated RAS proteins, anticancer agents capable of selectively killing tumour cells harbouring KRAS mutations have remained unavailable. Here we demonstrate the direct targeting of KRAS mutant DNA using a synthetic alkylating agent (pyrrole-imidazole polyamide indole-seco-CBI conjugate; KR12) that selectively recognizes oncogenic codon 12 KRAS mutations. KR12 alkylates adenine N3 at the target sequence, causing strand cleavage and growth suppression in human colon cancer cells with G12D or G12V mutations, thus inducing senescence and apoptosis. In xenograft models, KR12 infusions induce significant tumour growth suppression, with low host toxicity in KRAS-mutated but not wild-type tumours. This newly developed approach may be applicable to the targeting of other mutant driver oncogenes in human tumours.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Imidazoles/chemical synthesis , Imidazoles/therapeutic use , Neoplasms, Experimental/drug therapy , Nylons/chemical synthesis , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Caco-2 Cells , Cellular Senescence/drug effects , DNA Damage , Drug Evaluation, Preclinical , Female , HT29 Cells , Humans , Imidazoles/pharmacology , Mice, Nude , Mutation , Nylons/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics
19.
Cancer Sci ; 106(4): 421-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25611295

ABSTRACT

The MYC transcription factor plays a crucial role in the regulation of cell cycle progression, apoptosis, angiogenesis, and cellular transformation. Due to its oncogenic activities and overexpression in a majority of human cancers, it is an interesting target for novel drug therapies. MYC binding to the E-box (5'-CACGTGT-3') sequence at gene promoters contributes to more than 4000 MYC-dependent transcripts. Owing to its importance in MYC regulation, we designed a novel sequence-specific DNA-binding pyrrole-imidazole (PI) polyamide, Myc-5, that recognizes the E-box consensus sequence. Bioinformatics analysis revealed that the Myc-5 binding sequence appeared in 5'- MYC binding E-box sequences at the eIF4G1, CCND1, and CDK4 gene promoters. Furthermore, ChIP coupled with detection by quantitative PCR indicated that Myc-5 has the ability to inhibit MYC binding at the target gene promoters and thus cause downregulation at the mRNA level and protein expression of its target genes in human Burkitt's lymphoma model cell line, P493.6, carrying an inducible MYC repression system and the K562 (human chronic myelogenous leukemia) cell line. Single i.v. injection of Myc-5 at 7.5 mg/kg dose caused significant tumor growth inhibition in a MYC-dependent tumor xenograft model without evidence of toxicity. We report here a compelling rationale for the identification of a PI polyamide that inhibits a part of E-box-mediated MYC downstream gene expression and is a model for showing that phenotype-associated MYC downstream gene targets consequently inhibit MYC-dependent tumor growth.


Subject(s)
Burkitt Lymphoma/genetics , E-Box Elements/drug effects , Imidazoles/chemistry , Nylons/chemistry , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Pyrroles/chemistry , Animals , Apoptosis/drug effects , Binding Sites/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/genetics , Cyclin-Dependent Kinase 4/genetics , DNA-Binding Proteins , E-Box Elements/genetics , Eukaryotic Initiation Factor-4G/genetics , Humans , Mice , Mice, SCID , Nylons/chemical synthesis , Promoter Regions, Genetic , Protein Binding/drug effects , Proto-Oncogene Proteins c-myc/genetics , Xenograft Model Antitumor Assays
20.
FEBS Open Bio ; 4: 328-34, 2014.
Article in English | MEDLINE | ID: mdl-24918046

ABSTRACT

Gene amplification and/or overexpression of the transcription factor c-MYC, which binds to the E-box sequence (5'-CACGTG-3'), has been observed in many human tumors. In this study, we have designed 5 pyrrole-imidazole (PI) polyamides recognizing E-box, and found that, among them, Myc-6 significantly suppresses malignant phenotypes of human osteosarcoma MG63 cells both in vitro and in vivo. Intriguingly, knockdown of the putative Myc-6 target MALAT1 encoding long noncoding RNA remarkably impaired cell growth of MG63 cells. Collectively, our present findings strongly suggest that Myc-6 exerts its tumor-suppressive ability at least in part through the specific down-regulation of MALAT1.

SELECTION OF CITATIONS
SEARCH DETAIL