Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Gene Ther ; 31(5-6): 234-241, 2024 05.
Article in English | MEDLINE | ID: mdl-38135787

ABSTRACT

EPM1 is the most common form of Progressive Myoclonus Epilepsy characterized by late-childhood onset, ever-worsening and disabling myoclonus, seizures, ataxia, psychiatric disease, and shortened lifespan. EPM1 is caused by expansions of a dodecamer repeat sequence in the promoter of CSTB (cystatin B), which dramatically reduces, but does not eliminate, gene expression. The relatively late onset and consistent presence of a minimal amount of protein product makes EPM1 a favorable target for gene replacement therapy. If treated early, these children's normally developed brains could be rescued from the neurodegeneration that otherwise follows, and their cross-reactive immunological material (CRIM) positive status greatly reduces transgene related toxicity. We performed a proof-of-concept CSTB gene replacement study in Cstb knockout mice by introducing full-length human CSTB driven by the CBh promoter packaged in AAV9 and administered at postnatal days 21 and 60. Mice were sacrificed at 2 or 9 months of age, respectively. We observed significant improvements in expression levels of neuroinflammatory pathway genes and cerebellar granule cell layer apoptosis, as well as amelioration of motor impairment. The data suggest that gene replacement is a promising therapeutic modality for EPM1 and could spare affected children and families the ravages of this otherwise severe neurodegenerative disease.


Subject(s)
Cystatin B , Genetic Therapy , Mice, Knockout , Neuroinflammatory Diseases , Animals , Mice , Genetic Therapy/methods , Cystatin B/genetics , Neuroinflammatory Diseases/therapy , Neuroinflammatory Diseases/genetics , Humans , Ataxia/genetics , Ataxia/therapy , Myoclonic Epilepsies, Progressive/genetics , Myoclonic Epilepsies, Progressive/therapy , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors/genetics , Genetic Vectors/administration & dosage
2.
Neurotherapeutics ; 19(3): 982-993, 2022 04.
Article in English | MEDLINE | ID: mdl-35347645

ABSTRACT

Adult polyglucosan body disease (APBD) and Lafora disease (LD) are autosomal recessive glycogen storage neurological disorders. APBD is caused by mutations in the glycogen branching enzyme (GBE1) gene and is characterized by progressive upper and lower motor neuron dysfunction and premature death. LD is a fatal progressive myoclonus epilepsy caused by loss of function mutations in the EPM2A or EPM2B gene. These clinically distinct neurogenetic diseases share a common pathology. This consists of time-dependent formation, precipitation, and accumulation of an abnormal form of glycogen (polyglucosan) into gradually enlarging inclusions, polyglucosan bodies (PBs) in ever-increasing numbers of neurons and astrocytes. The growth and spread of PBs are followed by astrogliosis, microgliosis, and neurodegeneration. The key defect in polyglucosans is that their glucan branches are longer than those of normal glycogen, which prevents them from remaining in solution. Since the lengths of glycogen branches are determined by the enzyme glycogen synthase, we hypothesized that downregulating this enzyme could prevent or hinder the generation of the pathogenic PBs. Here, we pursued an adeno-associated virus vector (AAV) mediated RNA-interference (RNAi) strategy. This approach resulted in approximately 15% reduction of glycogen synthase mRNA and an approximately 40% reduction of PBs across the brain in the APBD and both LD mouse models. This was accompanied by improvements in early neuroinflammatory markers of disease. This work represents proof of principle toward developing a single lifetime dose therapy for two fatal neurological diseases: APBD and LD. The approach is likely applicable to other severe and common diseases of glycogen storage.


Subject(s)
Lafora Disease , MicroRNAs , Animals , Disease Models, Animal , Glucans , Glycogen , Glycogen Storage Disease , Glycogen Synthase/genetics , Lafora Disease/genetics , Lafora Disease/pathology , Lafora Disease/therapy , Mice , Nervous System Diseases , Neuroinflammatory Diseases
3.
Brain ; 144(10): 2985-2993, 2021 11 29.
Article in English | MEDLINE | ID: mdl-33993268

ABSTRACT

Lafora disease is a fatal progressive myoclonus epilepsy. At root, it is due to constant acquisition of branches that are too long in a subgroup of glycogen molecules, leading them to precipitate and accumulate into Lafora bodies, which drive a neuroinflammatory response and neurodegeneration. As a potential therapy, we aimed to downregulate glycogen synthase, the enzyme responsible for glycogen branch elongation, in mouse models of the disease. We synthesized an antisense oligonucleotide (Gys1-ASO) that targets the mRNA of the brain-expressed glycogen synthase 1 gene (Gys1). We administered Gys1-ASO by intracerebroventricular injection and analysed the pathological hallmarks of Lafora disease, namely glycogen accumulation, Lafora body formation, and neuroinflammation. Gys1-ASO prevented Lafora body formation in young mice that had not yet formed them. In older mice that already exhibited Lafora bodies, Gys1-ASO inhibited further accumulation, markedly preventing large Lafora bodies characteristic of advanced disease. Inhibition of Lafora body formation was associated with prevention of astrogliosis and strong trends towards correction of dysregulated expression of disease immune and neuroinflammatory markers. Lafora disease manifests gradually in previously healthy teenagers. Our work provides proof of principle that an antisense oligonucleotide targeting the GYS1 mRNA could prevent, and halt progression of, this catastrophic epilepsy.


Subject(s)
Glycogen Synthase/administration & dosage , Lafora Disease/drug therapy , Lafora Disease/pathology , Oligoribonucleotides, Antisense/administration & dosage , Animals , Female , Injections, Intraventricular , Lafora Disease/genetics , Male , Mice , Mice, Knockout , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics
4.
Neurotherapeutics ; 18(2): 1414-1425, 2021 04.
Article in English | MEDLINE | ID: mdl-33830476

ABSTRACT

Many adult and most childhood neurological diseases have a genetic basis. CRISPR/Cas9 biotechnology holds great promise in neurological therapy, pending the clearance of major delivery, efficiency, and specificity hurdles. We applied CRISPR/Cas9 genome editing in its simplest modality, namely inducing gene sequence disruption, to one adult and one pediatric disease. Adult polyglucosan body disease is a neurodegenerative disease resembling amyotrophic lateral sclerosis. Lafora disease is a severe late childhood onset progressive myoclonus epilepsy. The pathogenic insult in both is formation in the brain of glycogen with overlong branches, which precipitates and accumulates into polyglucosan bodies that drive neuroinflammation and neurodegeneration. We packaged Staphylococcus aureus Cas9 and a guide RNA targeting the glycogen synthase gene, Gys1, responsible for brain glycogen branch elongation in AAV9 virus, which we delivered by neonatal intracerebroventricular injection to one mouse model of adult polyglucosan body disease and two mouse models of Lafora disease. This resulted, in all three models, in editing of approximately 17% of Gys1 alleles and a similar extent of reduction of Gys1 mRNA across the brain. The latter led to approximately 50% reductions of GYS1 protein, abnormal glycogen accumulation, and polyglucosan bodies, as well as ameliorations of neuroinflammatory markers in all three models. Our work represents proof of principle for virally delivered CRISPR/Cas9 neurotherapeutics in an adult-onset (adult polyglucosan body) and a childhood-onset (Lafora) neurological diseases.


Subject(s)
Brain/metabolism , Glucans/metabolism , Glycogen Storage Disease/genetics , Glycogen Synthase/genetics , Glycogen/metabolism , Lafora Disease/genetics , Nervous System Diseases/genetics , Neuroinflammatory Diseases/genetics , RNA, Messenger/metabolism , Animals , CRISPR-Cas Systems , Disease Models, Animal , Gene Editing , Glycogen Storage Disease/metabolism , Glycogen Storage Disease/therapy , Lafora Disease/metabolism , Lafora Disease/therapy , Mice , Nervous System Diseases/metabolism , Nervous System Diseases/therapy , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/therapy , Proof of Concept Study
5.
Nat Metab ; 2(12): 1401-1412, 2020 12.
Article in English | MEDLINE | ID: mdl-33257855

ABSTRACT

In non-small-cell lung cancer (NSCLC), concurrent mutations in the oncogene KRAS and the tumour suppressor STK11 (also known as LKB1) encoding the kinase LKB1 result in aggressive tumours prone to metastasis but with liabilities arising from reprogrammed metabolism. We previously demonstrated perturbed nitrogen metabolism and addiction to an unconventional pathway of pyrimidine synthesis in KRAS/LKB1 co-mutant cancer cells. To gain broader insight into metabolic reprogramming in NSCLC, we analysed tumour metabolomes in a series of genetically engineered mouse models with oncogenic KRAS combined with mutations in LKB1 or p53. Metabolomics and gene expression profiling pointed towards activation of the hexosamine biosynthesis pathway (HBP), another nitrogen-related metabolic pathway, in both mouse and human KRAS/LKB1 co-mutant tumours. KRAS/LKB1 co-mutant cells contain high levels of HBP metabolites, higher flux through the HBP pathway and elevated dependence on the HBP enzyme glutamine-fructose-6-phosphate transaminase [isomerizing] 2 (GFPT2). GFPT2 inhibition selectively reduced KRAS/LKB1 co-mutant tumour cell growth in culture, xenografts and genetically modified mice. Our results define a new metabolic vulnerability in KRAS/LKB1 co-mutant tumours and provide a rationale for targeting GFPT2 in this aggressive NSCLC subtype.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Hexosamines/biosynthesis , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Metabolic Networks and Pathways , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , AMP-Activated Protein Kinase Kinases , Animals , Azaserine/therapeutic use , Carcinoma, Non-Small-Cell Lung/mortality , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/antagonists & inhibitors , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/genetics , Humans , Lung Neoplasms/mortality , Metabolomics , Mice , Mutation , Survival Analysis , Tumor Stem Cell Assay
6.
Clin Cancer Res ; 26(22): 6017-6027, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32847935

ABSTRACT

PURPOSE: Itraconazole has been repurposed as an anticancer therapeutic agent for multiple malignancies. In preclinical models, itraconazole has antiangiogenic properties and inhibits Hedgehog pathway activity. We performed a window-of-opportunity trial to determine the biologic effects of itraconazole in human patients. EXPERIMENTAL DESIGN: Patients with non-small cell lung cancer (NSCLC) who had planned for surgical resection were administered with itraconazole 300 mg orally twice daily for 10-14 days. Patients underwent dynamic contrast-enhanced MRI and plasma collection for pharmacokinetic and pharmacodynamic analyses. Tissues from pretreatment biopsy, surgical resection, and skin biopsies were analyzed for itraconazole and hydroxyitraconazole concentration, and vascular and Hedgehog pathway biomarkers. RESULTS: Thirteen patients were enrolled in this study. Itraconazole was well-tolerated. Steady-state plasma concentrations of itraconazole and hydroxyitraconazole demonstrated a 6-fold difference across patients. Tumor itraconazole concentrations trended with and exceeded those of plasma. Greater itraconazole levels were significantly and meaningfully associated with reduction in tumor volume (Spearman correlation, -0.71; P = 0.05) and tumor perfusion (Ktrans; Spearman correlation, -0.71; P = 0.01), decrease in the proangiogenic cytokines IL1b (Spearman correlation, -0.73; P = 0.01) and GM-CSF (Spearman correlation, -1.00; P < 0.001), and reduction in tumor microvessel density (Spearman correlation, -0.69; P = 0.03). Itraconazole-treated tumors also demonstrated distinct metabolic profiles. Itraconazole treatment did not alter transcription of GLI1 and PTCH1 mRNA. Patient size, renal function, and hepatic function did not predict itraconazole concentrations. CONCLUSIONS: Itraconazole demonstrates concentration-dependent early antivascular, metabolic, and antitumor effects in patients with NSCLC. As the number of fixed dose cancer therapies increases, attention to interpatient pharmacokinetics and pharmacodynamics differences may be warranted.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Itraconazole/administration & dosage , Neovascularization, Pathologic/drug therapy , Adult , Angiogenesis Inhibitors/adverse effects , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Biopsy , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/surgery , Female , Hedgehog Proteins/genetics , Humans , Itraconazole/analogs & derivatives , Itraconazole/blood , Itraconazole/pharmacokinetics , Magnetic Resonance Imaging , Male , Middle Aged , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/surgery , Patched-1 Receptor/genetics , Zinc Finger Protein GLI1/genetics
7.
Oncogene ; 39(16): 3258-3275, 2020 04.
Article in English | MEDLINE | ID: mdl-32108165

ABSTRACT

Activation of the Hedgehog (Hh) signaling pathway by mutations within its components drives the growth of several cancers. However, the role of Hh pathway activation in lung cancers has been controversial. Here, we demonstrate that the canonical Hh signaling pathway is activated in lung stroma by Hh ligands secreted from transformed lung epithelia. Genetic deletion of Shh, the primary Hh ligand expressed in the lung, in KrasG12D/+;Trp53fl/fl autochthonous murine lung adenocarcinoma had no effect on survival. Early abrogation of the pathway by an anti-SHH/IHH antibody 5E1 led to significantly worse survival with increased tumor and metastatic burden. Loss of IHH, another Hh ligand, by in vivo CRISPR led to more aggressive tumor growth suggesting that IHH, rather than SHH, activates the pathway in stroma to drive its tumor suppressive effects-a novel role for IHH in the lung. Tumors from mice treated with 5E1 had decreased blood vessel density and increased DNA damage suggestive of reactive oxygen species (ROS) activity. Treatment of KrasG12D/+;Trp53fl/fl mice with 5E1 and N-acetylcysteine, as a ROS scavenger, decreased tumor DNA damage, inhibited tumor growth and prolonged mouse survival. Thus, IHH induces stromal activation of the canonical Hh signaling pathway to suppress tumor growth and metastases, in part, by limiting ROS activity.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Hedgehog Proteins/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/genetics , Acetylcysteine/pharmacology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Animals , Antibodies, Anti-Idiotypic/pharmacology , Blood Vessels/drug effects , Cell Proliferation/drug effects , DNA Damage/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , Lung/metabolism , Lung/pathology , Mice , Mutation/genetics , Neoplasm Metastasis , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
8.
Cancer Res ; 77(16): 4448-4459, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28652248

ABSTRACT

Lung squamous cell carcinoma (SCC), strongly associated with smoking, is treated primarily with traditional cytotoxic chemotherapy due to a lack of FDA-approved targeted agents available. Here, we identify the Hedgehog pathway transcription factor GLI1 as a critical driver of lung SCC. Analysis of human lung cancer datasets showed that GLI1 mRNA was highly expressed in human lung SCC and portended a poor prognosis. Inhibition of GLI1 in human lung SCC cell lines suppressed tumor cell clonogenicity and proliferation in culture and in vivo Addition of SHH ligand, SMO antagonists, or other Hedgehog pathway agonists did not affect GLI1 expression in lung SCC cells. However, GLI1 expression was modulated by either inhibition or activation of the PI3K and MAPK pathways. Furthermore, in vivo growth of SCC harboring amplifications of the PI3K gene PIK3CA was attenuated by antagonizing GLI1 and PI3K. Thus, a combinatorial therapeutic strategy that targets the PI3K-mTOR pathway and GLI1 may lead to effective outcomes for PI3K pathway-dependent cancers, in contrast to recent results of human trials with single-agent PI3K antagonists. Cancer Res; 77(16); 4448-59. ©2017 AACR.


Subject(s)
Carcinoma, Squamous Cell/therapy , Lung Neoplasms/therapy , Phosphoinositide-3 Kinase Inhibitors , Zinc Finger Protein GLI1/antagonists & inhibitors , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Drug Synergism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Transfection , Xenograft Model Antitumor Assays , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism
9.
RSC Adv ; 3(10): 3260-3269, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23495364

ABSTRACT

HOXC13 is a homeobox containing gene that plays crucial roles in hair development and origin of replication. Herein, we investigated the biochemical functions of HOXC13 and explored its potential roles in tumor cell viability. We have designed a phosphorothioate based antisense-oligonucleotide that specifically knockdown HOXC13 in cultured cells. Cell viability and cytotoxicity assays demonstrated that HOXC13 is essential for cell growth and viability. Antisense-mediated knockdown of HOXC13 affected the cell viability and induced apoptosis in cultured tumor cells. HOXC13 regulates the expression of cyclins and antisense-mediated knockdown of HOXC13 resulted in cell cycle arrest and apoptosis in colon cancer cells. Finally over expression of HOXC13 resulted in 3D-colony formation in soft-agar assay indicating its potential roles in cell proliferation and tumorigenesis.

10.
J Mol Biol ; 425(19): 3707-22, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-23375982

ABSTRACT

HOTAIR (HOX antisense intergenic RNA) is a long noncoding RNA (lncRNA) that is transcribed from the antisense strand of homeobox C gene locus in chromosome 12. HOTAIR coordinates with chromatin-modifying enzymes and regulates gene silencing. It is overexpressed in various carcinomas including breast cancer. Herein, we demonstrated that HOTAIR is crucial for cell growth and viability and its knockdown induced apoptosis in breast cancer cells. We also demonstrated that HOTAIR is transcriptionally induced by estradiol (E2). Its promoter contains multiple functional estrogen response elements (EREs). Estrogen receptors (ERs) along with various ER coregulators such as histone methylases MLL1 (mixed lineage leukemia 1) and MLL3 and CREB-binding protein/p300 bind to the promoter of HOTAIR in an E2-dependent manner. Level of histone H3 lysine-4 trimethylation, histone acetylation, and RNA polymerase II recruitment is enriched at the HOTAIR promoter in the presence of E2. Knockdown of ERs and MLLs downregulated the E2-induced HOTAIR expression. Thus, similar to protein-coding gene transcription, E2-induced transcription of antisense transcript HOTAIR is coordinated via ERs and ER coregulators, and this mechanism of HOTAIR overexpression potentially contributes towards breast cancer progression.


Subject(s)
Antisense Elements (Genetics)/genetics , Estradiol/pharmacology , RNA, Long Noncoding/genetics , Transcription, Genetic , Antisense Elements (Genetics)/metabolism , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Cell Survival/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Knockdown Techniques , Gene Silencing , Histone-Lysine N-Methyltransferase , Histones/genetics , Histones/metabolism , Humans , MCF-7 Cells , Microchip Analytical Procedures , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , RNA, Long Noncoding/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Response Elements/drug effects
11.
Mol Endocrinol ; 27(1): 92-105, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23192982

ABSTRACT

High-density lipoprotein receptors scavenger receptor class B type I [HDLR-SR-B1 (SR-B1)] is a key player in reverse cholesterol transport and maintaining blood cholesterol. We demonstrated that human SR-B1 is transcriptionally activated by 17ß-estradiol (E2) in HEPG2 and JAR cells. SR-B1 promoter contains multiple estrogen response elements (ERE half-sites) along with some Sp1 binding sites. Knockdown of estrogen receptor (ER)α and ERß down-regulated E2-induced SR-B1 expression. ERs were bound to SR-B1 promoter EREs in an E2-dependent manner. Along with ERs, mixed-lineage leukemia (MLL) histone methylases, especially MLL1 and MLL2, play key roles in E2-mediated SR-B1 activation. MLL1 and MLL2 bind to SR-B1 promoter in an E2-dependent manner and control the assembly of transcription pre-initiation complex and RNA polymerase II (RNAPII) recruitment. ERs and MLLs play critical roles in determining the cholesterol uptake by steroidogenic tissues/cells, and their knockdown suppressed the E2-induced cholesterol uptake efficiencies of the cells. Intriguingly, MLL2 knockdown in mice resulted in a 33% increase in plasma cholesterol level and also reduced SR-B1 expression in mice liver, demonstrating its crucial functions in controlling plasma cholesterol in vivo.


Subject(s)
Cholesterol/blood , DNA-Binding Proteins/physiology , Estrogens/physiology , Gene Expression Regulation , Myeloid-Lymphoid Leukemia Protein/physiology , Neoplasm Proteins/physiology , Scavenger Receptors, Class B/genetics , Animals , Estradiol/physiology , Gene Knockdown Techniques , Genes, Reporter , Hep G2 Cells , Histone-Lysine N-Methyltransferase , Humans , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Male , Mice , Mice, Nude , Oligonucleotides, Antisense/genetics , Protein Binding , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Response Elements , Scavenger Receptors, Class B/metabolism , Transcription Initiation, Genetic
12.
FEBS J ; 279(19): 3715-3726, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22863320

ABSTRACT

HOXB9 is a homeobox-containing gene and is critical for the development of mammary gland and sternum. HOXB9 is also regulated by estrogen and is critical for angiogenesis. We investigated the biochemical roles of HOXB9 and its homeodomain in cell-cycle progression and tumorigenesis. Our studies demonstrated that HOXB9 is overexpressed in breast cancer tissue. HOXB9 overexpression stimulated 3D formation in soft agar assay. HOXB9 binds to the promoters of various tumor growth and angiogenic factors and regulates their expression. The homeodomain of HOXB9 plays crucial roles in transcriptional regulation of tumor growth factors and also in 3D colony formation, indicating crucial roles of the HOXB9 homeodomain in tumorigenesis. Overall, we demonstrated that HOXB9 is a critical regulator of tumor growth factors and is associated with tumorigenesis.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Blotting, Western , Breast Neoplasms/genetics , Cells, Cultured , Chromatin Immunoprecipitation , Colony-Forming Units Assay , Female , Fluorescent Antibody Technique , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Immunoenzyme Techniques , In Vitro Techniques , Intercellular Signaling Peptides and Proteins/genetics , Oligonucleotides, Antisense/pharmacology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis
13.
J Inorg Biochem ; 111: 33-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22484498

ABSTRACT

A series of new water soluble Ru(III) pyrazole complexes mer-[RuCl(3)(DMSO-S)(pyz)(2)] 1, mer-[RuCl(3)(DMSO-S)(DMSO-O)(pyz)] 2, mer-[RuCl(3)(bpy)(dmpyz)] 3, and mer-[RuCl(3)(DMSO-S)(dmpyz)(2)] 4 (pyz=pyrazole; dmpyz=3,5-dimethylpyrazole, bpy=2,2'-bipyridine) have been synthesized and characterized by use of a combination of spectroscopy (IR and UV-visible), X-ray diffraction, and cyclic voltammetry. The molecular X-ray structure of all reported compounds (1-4) revealed distorted octahedral coordination around ruthenium. The cytotoxicity assay on human breast cancer cells (MCF7) demonstrated that compounds 1 and 4 affect cell viability, whereas compounds 2 and 3 do not show appreciable activity. The IC(50) values for 1 and 4 lie within the range of 71-32µM in MCF7 cells.


Subject(s)
Coordination Complexes/chemical synthesis , Coordination Complexes/pharmacology , Pyrazoles/chemistry , Ruthenium/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/chemistry , Dose-Response Relationship, Drug , Electrochemical Techniques , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Spectrophotometry , X-Ray Diffraction
14.
Inorg Chem ; 51(6): 3343-5, 2012 Mar 19.
Article in English | MEDLINE | ID: mdl-22372979

ABSTRACT

The copper complex [Cu((t)BuPhimp)(Cl)] (1) derived from tridentate ligand (t)BuPhimpH having N(2)O donors was synthesized, and its molecular structure was determined. A phenoxyl radical complex was generated in solution at room temperature using Ce(IV). The nuclease and anticancer activities of 1 were investigated. The roles of the tert-butyl group and singlet oxygen in the DNA cleavage activity were also discussed.


Subject(s)
Antineoplastic Agents/pharmacology , Copper/pharmacology , Endonucleases/metabolism , Cell Line, Tumor , Humans , Inhibitory Concentration 50 , Ligands
15.
J Mol Endocrinol ; 48(1): 61-75, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22143955

ABSTRACT

HOXC10 is a critical player in the development of spinal cord, formation of neurons, and associated with human leukemia. We found that HOXC10 is overexpressed in breast cancer and transcriptionally regulated by estrogen (17ß-estradiol, E(2)). The HOXC10 promoter contains several estrogen response elements (ERE1-7, half-sites). A luciferase-based reporter assay showed that ERE1 and ERE6 of HOXC10 promoter are E(2) responsive. ERα and ERß play critical roles in E(2)-mediated activation of HOXC10. Knockdown of ERα and ERß downregulated E(2)-induced HOXC10 expression. ERα and ERß bind to ERE1 and ERE6 regions in an E(2)-dependent manner. Additionally, knockdown of histone methylases MLL3 and MLL4 (but not MLL1 and MLL2) diminished E(2)-induced expression of HOXC10. MLL3 and MLL4 were bound to the ERE1 and ERE6 regions of HOXC10 promoter in an E(2)-dependent manner. Overall, we demonstrated that HOXC10 is overexpressed in breast cancer, and it is an E(2)-responsive gene. Histone methylases MLL3 and MLL4, along with ERs, regulate HOXC10 gene expression in the presence of E(2).


Subject(s)
Breast Neoplasms/genetics , DNA-Binding Proteins/metabolism , Estrogens/pharmacology , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/metabolism , Homeodomain Proteins/genetics , Base Sequence , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Cell Line, Tumor , Choriocarcinoma/genetics , Choriocarcinoma/metabolism , Estrogen Antagonists/pharmacology , Female , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , Histone Methyltransferases , Humans , Nucleotide Motifs , Pregnancy , Promoter Regions, Genetic , Protein Binding , Receptors, Estrogen/metabolism , Response Elements/drug effects , Tamoxifen/pharmacology , Transcription, Genetic/drug effects , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism
16.
J Mol Biol ; 411(2): 334-49, 2011 Aug 12.
Article in English | MEDLINE | ID: mdl-21683083

ABSTRACT

Homeobox (HOX)-containing gene HOXC6 is a critical player in mammary gland development and milk production, and is overexpressed in breast and prostate cancers. We demonstrated that HOXC6 is transcriptionally regulated by estrogen (E2). HOXC6 promoter contains two putative estrogen response elements (EREs), termed as ERE1(1/2) and ERE2(1/2). Promoter analysis using luciferase-based reporter assay demonstrated that both EREs are responsive to E2, with ERE1(1/2) being more responsive than ERE2(1/2). Estrogen receptors (ERs) ERα and ERß bind to these EREs in an E2-dependent manner, and antisense-mediated knockdown of ERs suppressed the E2-dependent activation of HOXC6 expression. Similarly, knockdown of histone methylases MLL2 and MLL3 decreased the E2-mediated activation of HOXC6. However, depletion of MLL1 or MLL4 showed no significant effect. MLL2 and MLL3 were bound to the HOXC6 EREs in an E2-dependent manner. In contrast, MLL1 and MLL4 that were bound to the HOXC6 promoter in the absence of E2 decreased upon exposure to E2. MLL2 and MLL3 play key roles in histone H3 lysine-4 trimethylation and in the recruitment of general transcription factors and RNA polymerase II in the HOXC6 promoter during E2-dependent transactivation. Nuclear receptor corepressors N-CoR and SAFB1 were bound in the HOXC6 promoter in the absence of E2, and that binding was decreased upon E2 treatment, indicating their critical roles in suppressing HOXC6 gene expression under nonactivated conditions. Knockdown of either ERα or ERß abolished E2-dependent recruitment of MLL2 and MLL3 into the HOXC6 promoter, demonstrating key roles of ERs in the recruitment of these mixed lineage leukemias into the HOXC6 promoter. Overall, our studies demonstrated that HOXC6 is an E2-responsive gene, and that histone methylases MLL2 and MLL3, in coordination with ERα and ERß, transcriptionally regulate HOXC6 in an E2-dependent manner.


Subject(s)
DNA-Binding Proteins/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Estrogens/metabolism , Gene Expression Regulation , Homeodomain Proteins/metabolism , Neoplasm Proteins/metabolism , Cell Line, Tumor , DNA/metabolism , Homeodomain Proteins/genetics , Humans , Models, Biological , Promoter Regions, Genetic , Protein Binding , Transcription, Genetic
17.
Biochemistry ; 50(17): 3517-27, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21428455

ABSTRACT

Homeobox gene HOXB9 is a critical player in development of mammary gland and sternum and in regulation of renin which is closely linked with blood pressure control. Our studies demonstrated that HOXB9 gene is transcriptionally regulated by estrogen (E2). HOXB9 promoter contains several estrogen-response elements (ERE). Reporter assay based experiments demonstrated that HOXB9 promoter EREs are estrogen responsive. Estrogen receptors ERα and ERß are essential for E2-mediated transcriptional activation of HOXB9. Chromatin immunoprecipitation assay demonstrated that ERs bind to HOXB9 EREs as a function of E2. Similarly, histone methylases MLL1 and MLL3 also bind to HOXB9 EREs and play a critical role in E2-mediated transcriptional activation of HOXB9. Overall, our studies demonstrated that HOXB9 is an E2-responsive gene and ERs coordinate with MLL1 and MLL3 in E2-mediated transcriptional regulation of HOXB9.


Subject(s)
Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Estrogens/pharmacology , Histone-Lysine N-Methyltransferase/metabolism , Homeodomain Proteins/biosynthesis , Cell Line, Tumor , Gene Knockdown Techniques , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Homeodomain Proteins/genetics , Humans , Promoter Regions, Genetic , Protein Binding , Protein Transport , Transcriptional Activation
18.
J Biomol Screen ; 16(1): 26-35, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21045212

ABSTRACT

To explore the apoptotic and antitumor activities of metallo-salens, the authors have synthesized several Fe(III)-salen and salphen complexes and analyzed their effects on human cancer and noncancer cells. Their results demonstrated that Fe(III)-salen and salphen complexes affect cell viability and induce nuclear fragmentation and apoptosis in breast cancer (MCF7) cells. The IC(50) values for the active metallo-salen complexes ranged between 0.3 and 22 µM in MCF7 cells. Biochemically active Fe(III)-salen and salphen complexes induced caspase-3/7 activation and release of cytochrome c from the mitochondria to cytosol, suggesting the involvement of the mitochondrial pathway of apoptosis. Comparison of IC(50) values toward 3 different cell lines demonstrated that selected Fe(III)-salen complexes induce tumor cell-selective apoptosis in cultured cells. Overall, the studies demonstrated that Fe(III)-salen and salphen complexes induced efficient apoptosis in cultured human cells. The nature of the substituents and the bridging spacer between diamino groups play critical roles in determining the apoptotic activities of Fe(III)-salen and salphen complexes.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Ethylenediamines/pharmacology , Ferric Compounds/pharmacology , Phenylenediamines/pharmacology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cytochromes c/metabolism , Ethylenediamines/chemical synthesis , Ferric Compounds/chemical synthesis , Humans , Inhibitory Concentration 50 , Mitochondria/drug effects , Mitochondria/metabolism , Phenylenediamines/chemical synthesis , Structure-Activity Relationship
19.
FEBS J ; 276(24): 7400-11, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19922474

ABSTRACT

HOXC13, a homeobox-containing gene, is involved in hair development and human leukemia. The regulatory mechanism that drives HOXC13 expression is mostly unknown. Our studies have demonstrated that HOXC13 is transcriptionally activated by the steroid hormone estrogen (17beta-estradiol; E2). The HOXC13 promoter contains several estrogen-response elements (EREs), including ERE1 and ERE2, which are close to the transcription start site, and are associated with E2-mediated activation of HOXC13. Knockdown of the estrogen receptors (ERs) ERalpha and ERbeta suppressed E2-mediated activation of HOXC13. Similarly, knockdown of mixed lineage leukemia histone methylase (MLL)3 suppressed E2-induced activation of HOXC13. MLLs (MLL1-MLL4) were bound to the HOXC13 promoter in an E2-dependent manner. Knockdown of either ERalpha or ERbeta affected the E2-dependent binding of MLLs (MLL1-MLL4) into HOXC13 EREs, suggesting critical roles of ERs in recruiting MLLs in the HOXC13 promoter. Overall, our studies have demonstrated that HOXC13 is transcriptionally regulated by E2 and MLLs, which, in coordination with ERalpha and ERbeta, play critical roles in this process. Although MLLs are known to regulate HOX genes, the roles of MLLs in hormone-mediated regulation of HOX genes are unknown. Herein, we have demonstrated that MLLs are critical players in E2-dependent regulation of the HOX gene.


Subject(s)
Estradiol/physiology , Histone-Lysine N-Methyltransferase/physiology , Homeodomain Proteins/genetics , Myeloid-Lymphoid Leukemia Protein/physiology , Estrogen Receptor alpha/physiology , Estrogen Receptor beta/physiology , Gene Knockdown Techniques , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Humans , Tumor Cells, Cultured
20.
Dalton Trans ; (40): 8525-31, 2009 Oct 28.
Article in English | MEDLINE | ID: mdl-19809727

ABSTRACT

We analyzed the apoptosis and anti-tumour activities of several Mn(III)-salen and -salphen complexes (1-14) towards three different cultured human cancer and non-cancer cells. We demonstrated that most of the Mn(III)-salen and -salphen complexes affect cell viability and induce apoptosis in MCF7 cells. Biochemically active Mn(III)-salen and -salphen complexes induced nuclear fragmentation and release of cytochrome c from the mitochondria to cytosol indicating involvement of mitochondrial pathway of apoptosis. The nature and position of the substituents and the bridging group on the salen ligands play crucial roles in determining the apoptotic activities of Mn(III)-salen and -salphen complexes. The IC50 values for the active Mn(III)-salen complexes ranged between 12 and 55 microM. For Mn(III)-salen complexes with ethylenediamine bridges, methoxy substituted complexes were more active than the corresponding hydroxy derivatives. However, this correlation does not hold when the bridging group was changed from ethylenediamine to o-phenylenediamine. Importantly, several Mn(III)-salen and -salphen complexes showed about 2-3 fold selectivity toward cancer cells such as MCF7 (breast cancer), and CCL228 (colon cancer) over a normal non-malignant cell MCF10 (breast epithelial cells) indicating their potential application towards novel anti-tumour therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Ethylenediamines/chemistry , Manganese/chemistry , Organometallic Compounds/pharmacology , Phenylenediamines/chemistry , Antineoplastic Agents/chemistry , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Organometallic Compounds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...