Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Med Chem ; 67(7): 5473-5501, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38554135

ABSTRACT

Proteolysis-Targeting Chimeras (PROTACs) are bifunctional molecules that bring a target protein and an E3 ubiquitin ligase into proximity to append ubiquitin, thus directing target degradation. Although numerous PROTACs have entered clinical trials, their development remains challenging, and their large size can produce poor drug-like properties. To overcome these limitations, we have modified our Coferon platform to generate Combinatorial Ubiquitination REal-time PROteolysis (CURE-PROs). CURE-PROs are small molecule degraders designed to self-assemble through reversible bio-orthogonal linkers to form covalent heterodimers. By modifying known ligands for Cereblon, MDM2, VHL, and BRD with complementary phenylboronic acid and diol/catechol linkers, we have successfully created CURE-PROs that direct degradation of BRD4 both in vitro and in vivo. The combinatorial nature of our platform significantly reduces synthesis time and effort to identify the optimal linker length and E3 ligase partner to each target and is readily amenable to screening for new targets.


Subject(s)
Nuclear Proteins , Transcription Factors , Proteolysis , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Ubiquitination , Ubiquitin-Protein Ligases/metabolism , Ligands
2.
ACS Chem Biol ; 18(7): 1582-1593, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37422908

ABSTRACT

PROteolysis TArgeting Chimeras (PROTACs) are of significant current interest for the development of probe molecules and drug leads. However, they suffer from certain limitations. PROTACs are rule-breaking molecules with sub-optimal cellular permeability, solubility, and other drug-like properties. In particular, they exhibit an unusual dose-response curve where high concentrations of the bivalent molecule inhibit degradation activity, a phenomenon known as the hook effect. This will likely complicate their use in vivo. In this study, we explore a novel approach to create PROTACs that do not exhibit a hook effect. This is achieved by equipping the target protein and E3 ubiquitin ligase ligands with functionalities that undergo rapid and reversible covalent assembly in cellulo. We report the development of Self-Assembled Proteolysis Targeting Chimeras that mediate the degradation of the Von Hippel-Lindau E3 ubiquitin ligase and do not evince a hook effect.


Subject(s)
Proteolysis Targeting Chimera , Ubiquitin-Protein Ligases , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Proteins/metabolism , Ligands
3.
Curr Cancer Drug Targets ; 21(4): 306-325, 2021.
Article in English | MEDLINE | ID: mdl-33535953

ABSTRACT

Epigenetic modulation of gene expression is essential for tissue-specific development and maintenance in mammalian cells. Disruption of epigenetic processes, and the subsequent alteration of gene functions, can result in inappropriate activation or inhibition of various cellular signaling pathways, leading to cancer. Recent advancements in the understanding of the role of epigenetics in cancer initiation and progression have uncovered functions for DNA methylation, histone modifications, nucleosome positioning, and non-coding RNAs. Epigenetic therapies have shown some promise for hematological malignancies, and a wide range of epigenetic-based drugs are undergoing clinical trials. However, in a dynamic survival strategy, cancer cells exploit their heterogeneous population which frequently results in the rapid acquisition of therapy resistance. Here, we describe novel approaches in drug discovery targeting the epigenome, highlighting recent advances the selective degradation of target proteins using Proteolysis Targeting Chimera (PROTAC) to address drug resistance.


Subject(s)
Drug Resistance, Neoplasm , Molecular Targeted Therapy , Neoplasms , Proteolysis , Drug Discovery/methods , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Epigenome/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Neoplasms/drug therapy , Neoplasms/genetics
4.
J Mol Diagn ; 22(7): 885-900, 2020 07.
Article in English | MEDLINE | ID: mdl-32407802

ABSTRACT

The analysis of CpG methylation in circulating tumor DNA fragments has emerged as a promising approach for the noninvasive early detection of solid tumors, including colorectal cancer (CRC). The most commonly employed assay involves bisulfite conversion of circulating tumor DNA, followed by targeted PCR, then real-time quantitative PCR (alias methylation-specific PCR). This report demonstrates the ability of a multiplex bisulfite PCR-ligase detection reaction-real-time quantitative PCR assay to detect seven methylated CpG markers (CRC or colon specific), in both simulated (approximately 30 copies of fragmented CRC cell line DNA mixed with approximately 3000 copies of fragmented peripheral blood DNA) and CRC patient-derived cell-free DNAs. This scalable assay is designed for multiplexing and incorporates steps for improved sensitivity and specificity, including the enrichment of methylated CpG fragments, ligase detection reaction, the incorporation of ribose bases in primers, and use of uracil DNA glycosylase. Six of the seven CpG markers (located in promoter regions of PPP1R16B, KCNA3, CLIP4, GDF6, SEPT9, and GSG1L) were identified through integrated analyses of genome-wide methylation data sets for 31 different types of cancer. These markers were mapped to CpG sites at the promoter region of VIM; VIM and SEPT9 are established epigenetic markers of CRC. Additional bioinformatics analyses show that the methylation at these CpG sites negatively correlates with the transcription of their corresponding genes.


Subject(s)
Colorectal Neoplasms/blood , Colorectal Neoplasms/genetics , DNA Methylation/genetics , Multiplex Polymerase Chain Reaction/methods , Real-Time Polymerase Chain Reaction/methods , Base Sequence/genetics , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Circulating Tumor DNA/blood , Circulating Tumor DNA/genetics , Cohort Studies , Colorectal Neoplasms/diagnosis , Computational Biology/methods , CpG Islands/genetics , HT29 Cells , Humans , Ligases/genetics , Promoter Regions, Genetic/genetics , Septins/blood , Septins/genetics , Vimentin/blood , Vimentin/genetics
5.
J Med Chem ; 63(6): 3004-3027, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32057241

ABSTRACT

ß-Tryptase, a homotetrameric serine protease, has four identical active sites facing a central pore, presenting an optimized setting for the rational design of bivalent inhibitors that bridge two adjacent sites. Using diol, hydroxymethyl phenols or benzoyl methyl hydroxamates, and boronic acid chemistries to reversibly join two [3-(1-acylpiperidin-4-yl)phenyl]methanamine core ligands, we have successfully produced a series of self-assembling heterodimeric inhibitors. These heterodimeric tryptase inhibitors demonstrate superior activity compared to monomeric modes of inhibition. X-ray crystallography validated the dimeric mechanism of inhibition, and compounds demonstrated high selectivity against related proteases, good target engagement, and tryptase inhibition in HMC1 xenograft models. Screening 3872 possible combinations from 44 boronic acid and 88 diol derivatives revealed several combinations that produced nanomolar inhibition, and seven unique pairs produced greater than 100-fold improvement in potency over monomeric inhibition. These heterodimeric tryptase inhibitors demonstrate the power of target-driven combinatorial chemistry to deliver bivalent drugs in a small molecule form.


Subject(s)
Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Tryptases/antagonists & inhibitors , Animals , Boronic Acids/chemistry , Boronic Acids/pharmacology , Crystallography, X-Ray , Female , Humans , Mice , Molecular Docking Simulation , Protein Conformation/drug effects , Protein Multimerization/drug effects , Tryptases/chemistry , Tryptases/metabolism
6.
BMC Cancer ; 20(1): 85, 2020 Jan 31.
Article in English | MEDLINE | ID: mdl-32005108

ABSTRACT

BACKGROUND: Interrogation of site-specific CpG methylation in circulating tumor DNAs (ctDNAs) has been employed in a number of studies for early detection of breast cancer (BrCa). In many of these studies, the markers were identified based on known biology of BrCa progression, and interrogated using methyl-specific PCR (MSP), a technique involving bisulfite conversion, PCR, and qPCR. METHODS: In this report, we are demonstrating the development of a novel assay (Multiplex Bisulfite PCR-LDR-qPCR) which can potentially offer improvements to MSP, by integrating additional steps such as ligase detection reaction (LDR), methylated CpG target enrichment, carryover protection (use of uracil DNA glycosylase), and minimization of primer-dimer formation (use of ribose primers and RNAseH2). The assay is designed to for breast cancer-specific CpG markers identified through integrated analyses of publicly available genome-wide methylation datasets for 31 types of primary tumors (including BrCa), as well as matching normal tissues, and peripheral blood. RESULTS: Our results indicate that the PCR-LDR-qPCR assay is capable of detecting ~ 30 methylated copies of each of 3 BrCa-specific CpG markers, when mixed with excess amount unmethylated CpG markers (~ 3000 copies each), which is a reasonable approximation of BrCa ctDNA overwhelmed with peripheral blood cell-free DNA (cfDNA) when isolated from patient plasma. The bioinformatically-identified CpG markers are located in promoter regions of NR5A2 and PRKCB, and a non-coding region of chromosome 1 (upstream of EFNA3). Additional bioinformatic analyses would reveal that these methylation markers are independent of patient race and age, and positively associated with signaling pathways associated with BrCa progression (such as those related to retinoid nuclear receptor, PTEN, p53, pRB, and p27). CONCLUSION: This report demonstrates the potential utilization of bisulfite PCR-LDR-qPCR assay, along with bioinformatically-driven biomarker discovery, in blood-based BrCa detection.


Subject(s)
Biomarkers, Tumor/blood , Breast Neoplasms/diagnosis , Cell-Free Nucleic Acids/blood , DNA Methylation , Breast Neoplasms/blood , Breast Neoplasms/genetics , Cell Line, Tumor , CpG Islands , Female , Humans , MCF-7 Cells , Multiplex Polymerase Chain Reaction , Protein Kinase C beta/genetics , Receptors, Cytoplasmic and Nuclear/genetics
7.
Hum Mutat ; 41(5): 1051-1068, 2020 05.
Article in English | MEDLINE | ID: mdl-31950578

ABSTRACT

Detection of low-abundance mutations in cell-free DNA is being used to identify early cancer and early cancer recurrence. Here, we report a new PCR-LDR-qPCR assay capable of detecting point mutations at a single-molecule resolution in the presence of an excess of wild-type DNA. Major features of the assay include selective amplification and detection of mutant DNA employing multiple nested primer-binding regions as well as wild-type sequence blocking oligonucleotides, prevention of carryover contamination, spatial sample dilution, and detection of multiple mutations in the same position. Our method was tested to interrogate the following common cancer somatic mutations: BRAF:c.1799T>A (p.Val600Glu), TP53:c.743G>A (p.Arg248Gln), KRAS:c.35G>C (p.Gly12Ala), KRAS:c.35G>T (p.Gly12Val), KRAS:c.35G>A (p.Gly12Asp), KRAS:c.34G>T (p.Gly12Cys), and KRAS:c.34G>A (p.Gly12Ser). The single-well version of the assay detected 2-5 copies of these mutations, when diluted with 10,000 genome equivalents (GE) of wild-type human genomic DNA (hgDNA) from buffy coat. A 12-well (pixel) version of the assay was capable of single-molecule detection of the aforementioned mutations at TP53, BRAF, and KRAS (specifically p.Gly12Val and p.Gly12Cys), mixed with 1,000-2,250 GE of wild-type hgDNA from plasma or buffy coat. The assay described herein is highly sensitive, specific, and robust, and potentially useful in liquid biopsies.


Subject(s)
Biomarkers, Tumor/genetics , Neoplasms/genetics , Point Mutation , Real-Time Polymerase Chain Reaction , Single Molecule Imaging/methods , Alleles , Amino Acid Substitution , Cell Line, Tumor , Circulating Tumor DNA , DNA Mutational Analysis/methods , Genotype , Humans , Real-Time Polymerase Chain Reaction/methods
8.
ACS Med Chem Lett ; 9(8): 827-831, 2018 Aug 09.
Article in English | MEDLINE | ID: mdl-30128075

ABSTRACT

Tryptase, a serine protease released from mast cells, is implicated in many allergic and inflammatory disorders. Human tryptase is a donut-shaped tetramer with the active sites facing inward forming a central pore. Bivalent ligands spanning two active sites potently inhibit this configuration, but these large compounds have poor drug-like properties. To overcome some of these challenges, we developed self-assembling molecules, called coferons, which deliver a larger compound in two parts. Using a pharmacophoric core and reversibly binding linkers to span two active sites, we have successfully produced three novel homodimeric tryptase inhibitors. Upon binding to tryptase, compounds reassembled into flexible homodimers, with significant improvements in IC50 (0.19 ± 0.08 µM) over controls (5.50 ± 0.09 µM), and demonstrate good activity in mast cell lines. These studies provide validation for this innovative technology that is especially well-suited for the delivery of dimeric drugs to modulate intracellular macromolecular targets.

9.
Pharmacology ; 102(5-6): 233-243, 2018.
Article in English | MEDLINE | ID: mdl-30134249

ABSTRACT

ß-Tryptase is released from mast cells upon degranulation in response to allergic and inflammatory stimuli. Human tryptase is a homotetrameric serine protease with 4 identical active sites directed toward a central pore. These active sites present an optimized scenario for the rational design of bivalent inhibitors, which bridge 2 adjacent active sites. Using (3-[1-acylpiperidin-4-yl]phenyl)methanamine as the pharmacophoric core and a disiloxane linker to span 2 active sites we have successfully produced a novel bivalent tryptase inhibitor, compound 1a, with a comparable profile to previously described inhibitors. Pharmacological properties of compound 1a were studied in a range of in vitro enzymic and cellular screening assays, and in vivo xenograft models. This non-peptide inhibitor of tryptase demonstrated superior activity (IC50 at 100 pmol/L tryptase = 1.82 nmol/L) compared to monomeric modes of inhibition. X-ray crystallography validated the dimeric mechanism of inhibition, and 1a demonstrated good oral bioavailability and efficacy in HMC-1 xenograft models. Furthermore, compound 1a demonstrated extremely slow off rates and high selectivity against-related proteases. This highly potent, orally bioavailable and selective inhibitor of human tryptase will be an invaluable tool in future studies to explore the therapeutic potential of attenuating the activity of this elusive target.


Subject(s)
Mast Cells/drug effects , Silanes/chemistry , Silanes/pharmacology , Tryptases/antagonists & inhibitors , Animals , Cell Line , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Design , Humans , Immunohistochemistry , Male , Mast Cells/enzymology , Mast Cells/pathology , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Pharmacokinetics , Silanes/analysis , Silanes/pharmacokinetics
10.
Proc Natl Acad Sci U S A ; 106(17): 7131-6, 2009 Apr 28.
Article in English | MEDLINE | ID: mdl-19359472

ABSTRACT

During disease progression the cells that comprise solid malignancies undergo significant changes in gene copy number and chromosome structure. Colorectal cancer provides an excellent model to study this process. To indentify and characterize chromosomal abnormalities in colorectal cancer, we performed a statistical analysis of 299 expression and 130 SNP arrays profiled at different stages of the disease, including normal tissue, adenoma, stages 1-4 adenocarcinoma, and metastasis. We identified broad (> 1/2 chromosomal arm) and focal (< 1/2 chromosomal arm) events. Broad amplifications were noted on chromosomes 7, 8q, 13q, 20, and X and broad deletions on chromosomes 4, 8p, 14q, 15q, 17p, 18, 20p, and 22q. Focal events (gains or losses) were identified in regions containing known cancer pathway genes, such as VEGFA, MYC, MET, FGF6, FGF23, LYN, MMP9, MYBL2, AURKA, UBE2C, and PTEN. Other focal events encompassed potential new candidate tumor suppressors (losses) and oncogenes (gains), including CCDC68, CSMD1, POLR1D, and PMEPA1. From the expression data, we identified genes whose expression levels reflected their copy number changes and used this relationship to impute copy number changes to samples without accompanying SNP data. This analysis provided the statistical power to show that deletions of 8p, 4p, and 15q are associated with survival and disease progression, and that samples with simultaneous deletions in 18q, 8p, 4p, and 15q have a particularly poor prognosis. Annotation analysis reveals that the oxidative phosphorylation pathway shows a strong tendency for decreased expression in the samples characterized by poor prognosis.


Subject(s)
Chromosomal Instability/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Genome, Human/genetics , Chromosome Deletion , Disease Progression , Fibroblast Growth Factor-23 , Gene Dosage/genetics , Gene Expression Regulation, Neoplastic , Humans , Oligonucleotide Array Sequence Analysis , Oxidation-Reduction , Phosphorylation , Polymorphism, Single Nucleotide/genetics , RNA, Untranslated/genetics , Survival Rate
11.
Cancer Res ; 69(3): 723-7, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19155292

ABSTRACT

High-density single nucleotide polymorphism (SNP) mapping arrays have identified chromosomal features whose importance to cancer predisposition and progression is not yet clearly defined. Of interest is that the genomes of normal somatic cells (reflecting the combined parental germ-line contributions) often contain long homozygous stretches. These chromosomal segments may be explained by the common ancestry of the individual's parents and thus may also be called autozygous. Several studies link consanguinity to higher rates of cancer, suggesting that autozygosity (a genomic consequence of consanguinity) may be a factor in cancer predisposition. SNP array analysis has also identified chromosomal regions of somatic uniparental disomy (UPD) in cancer genomes. These are chromosomal segments characterized by loss of heterozygosity (LOH) and a normal copy number (two) but which are not autozygous in the germ-line or normal somatic cell genome. In this review, we will also discuss a model [cancer gene activity model (CGAM)] that may explain how autozygosity influences cancer predisposition. CGAM can also explain how the occurrence of certain chromosomal aberrations (copy number gain, LOH, and somatic UPDs) during carcinogenesis may be dependent on the germ-line genotypes of important cancer-related genes (oncogenes and tumor suppressors) found in those chromosomal regions.


Subject(s)
Neoplasms/genetics , Polymorphism, Single Nucleotide , Genetic Predisposition to Disease , Humans
12.
Clin Cancer Res ; 14(19): 6005-13, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18829479

ABSTRACT

PURPOSE: Aberrant promoter methylation and genomic instability occur frequently during colorectal cancer development. CpG island methylator phenotype (CIMP) has been shown to associate with microsatellite instability, and BRAF mutation and is often found in the right-side colon. Nevertheless, the relative importance of CIMP and chromosomal instability (CIN) for tumorigenesis has yet to be thoroughly investigated in sporadic colorectal cancers. EXPERIMENTAL DESIGN: We determined CIMP in 161 primary colorectal cancers and 66 matched normal mucosae using a quantitative bisulfite/PCR/ligase detection reaction (LDR)/Universal Array assay. The validity of CIMP was confirmed in a subset of 60 primary tumors using MethyLight assay and five independent markers. In parallel, CIN was analyzed in the same study cohort using Affymetrix 50K Human Mapping arrays. RESULTS: The identified CIMP-positive cancers correlate with microsatellite instability (P = 0.075) and the BRAF mutation V600E (P = 0.00005). The array-based high-resolution analysis of chromosomal aberrations indicated that the degree of aneuploidy is spread over a wide spectrum among analyzed colorectal cancers. Whether CIN was defined by copy number variations in selected microsatellite loci (criterion 1) or considered as a continuous variable (criterion 2), CIMP-positive samples showed a strong correlation with low-degree chromosomal aberrations (P = 0.075 and P = 0.012, respectively). Similar correlations were observed when CIMP was determined by MethyLight assay (P = 0.001 and P = 0.013, respectively). CONCLUSION: CIMP-positive tumors generally possess lower chromosomal aberrations, which may only be revealed using a genome-wide approach. The significant difference in the degree of chromosomal aberrations between CIMP-positive and the remainder of samples suggests that epigenetic (CIMP) and genetic (CIN) abnormalities may arise from independent molecular mechanisms of tumor progression.


Subject(s)
Chromosome Aberrations , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , CpG Islands , DNA Methylation , Gene Expression Regulation, Neoplastic , Mutation , Chromosomal Instability , Epigenesis, Genetic , Female , Genome, Human , Humans , Male , Microsatellite Repeats , Phenotype , Proto-Oncogene Proteins B-raf/genetics
13.
Cancer Res ; 68(8): 2610-21, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18375840

ABSTRACT

Previous studies have shown that among populations with a high rate of consanguinity, there is a significant increase in the prevalence of cancer. Single nucleotide polymorphism (SNP) array data (Affymetrix, 50K XbaI) analysis revealed long regions of homozygosity in genomic DNAs taken from tumor and matched normal tissues of colorectal cancer (CRC) patients. The presence of these regions in the genome may indicate levels of consanguinity in the individual's family lineage. We refer to these autozygous regions as identity-by-descent (IBD) segments. In this study, we compared IBD segments in 74 mostly Caucasian CRC patients (mean age of 66 years) to two control data sets: (a) 146 Caucasian individuals (mean age of 80 years) who participated in an age-related macular degeneration (AMD) study and (b) 118 cancer-free Caucasian individuals from the Framingham Heart Study (mean age of 67 years). Our results show that the percentage of CRC patients with IBD segments (>or=4 Mb length and 50 SNPs probed) in the genome is at least twice as high as the AMD or Framingham control groups. Also, the average length of these IBD regions in the CRC patients is more than twice the length of the two control data sets. Compared with control groups, IBD segments are found to be more common among individuals of Jewish background. We believe that these IBD segments within CRC patients are likely to harbor important CRC-related genes with low-penetrance SNPs and/or mutations, and, indeed, two recently identified CRC predisposition SNPs in the 8q24 region were confirmed to be homozygous in one particular patient carrying an IBD segment covering the region.


Subject(s)
Colorectal Neoplasms/genetics , Homozygote , Loss of Heterozygosity , Polymorphism, Single Nucleotide , Aged , Aged, 80 and over , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/mortality , Consanguinity , Female , Heterozygote , Humans , Incidence , Irritable Bowel Syndrome/genetics , Macular Degeneration , Male , United States
14.
Biochem Biophys Res Commun ; 362(3): 747-52, 2007 Oct 26.
Article in English | MEDLINE | ID: mdl-17761141

ABSTRACT

The current study aimed to understand the anti-apoptotic effect of overexpressed gap junction forming protein connexin (Cx) 43 in C6 glioma cells. C6 cells exposed to hydrogen peroxide (H2O2) or staurosporine demonstrated morphological and biochemical changes consistent with apoptosis, whereas C6 cells expressing Cx43 demonstrated relative resistance to H2O2, but not to staurosporine. This selective protection against H2O2 was due to inhibition of caspase-3 activation in Cx43 expressing cells. siRNA knockdown experiments in rat primary astrocytes confirmed the presence of endogenous Cx43-mediated anti-apoptotic effect. Cx43 interacts with the upstream apoptosis signal-regulating kinase 1 known to mediate H2O2-induced apoptosis providing a possible mechanism for protection. These findings provided new evidence for regulation of the mitogen activated protein kinase pathway and apoptosis by Cx43 implicating this protein in intracellular signaling beyond its role as a gap junction forming protein on the plasma membrane.


Subject(s)
Apoptosis , Brain Neoplasms/pathology , Brain/pathology , Connexin 43/metabolism , Connexin 43/physiology , Glioma/pathology , Hydrogen Peroxide/pharmacology , Animals , Astrocytes/metabolism , Brain/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Gap Junctions/metabolism , MAP Kinase Kinase Kinase 5/biosynthesis , Rats , Signal Transduction
15.
Biochem Biophys Res Commun ; 352(1): 97-103, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17107662

ABSTRACT

Connexin 43 (Cx43) is thought to be present largely in the plasma membrane and its function solely to provide low resistance electrical connection between myocytes. A recent report suggested the presence of Cx43 in the mitochondria as well. We confirmed the presence of Cx43 in the mitochondria isolated from adult rat ventricles with the Cx43 immunoreactivity fractionating to the outer mitochondrial membrane. Mitochondrial Cx43 is mostly phosphorylated only detected by a phospho-specific antibody. Using a Ca2+ -sensitive electrode and Western blot, we showed that the gap junction inhibitors 18-beta-glycyrrhetinic acid (beta-GA), oleamide, and heptanol all induced concomitant release of Ca2+ and cytochrome C in isolated mitochondria whereas the inactive analog 18-beta-glycyrrhizic acid failed to do so. In low density neonatal myocyte culture with no appreciable cell-cell contacts, beta-GA induced apoptosis as assessed by TUNEL staining. Our results suggest a novel role of Cx43 as a regulator of mitochondrial physiology and myocyte apoptosis.


Subject(s)
Apoptosis , Connexin 43/metabolism , Mitochondria, Heart/metabolism , Myocardium/cytology , Myocardium/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Connexin 43/genetics , Connexin 43/immunology , Cytochromes c/metabolism , Gap Junctions/metabolism , Glycyrrhetinic Acid/pharmacology , Monocytes/cytology , Monocytes/drug effects , Rats
16.
Clin Cancer Res ; 12(20 Pt 1): 5951-9, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17062666

ABSTRACT

PURPOSE: The growth-related oncogene alpha (GROalpha) is a secreted interleukin-like molecule that interacts with the CXCR2 G-protein-coupled receptor. We found that the mRNA and protein products of GROalpha are more highly expressed in neoplastic than normal colon epithelium, and we studied potential mechanisms by which GROalpha may contribute to tumor initiation or growth. EXPERIMENTAL DESIGN: Cell lines that constitutively overexpress GROalpha were tested for growth rate, focus formation, and tumor formation in a xenograft model. GROalpha expression was determined by Affymetrix GeneChip (241 microdissected colon samples), real-time PCR (n = 32), and immunohistochemistry. Primary colon cancer samples were also employed to determine copy number changes and loss of heterozygosity related to the GROalpha and fibulin-1 genes. RESULTS: In cell cultures, GROalpha transfection transformed NIH 3T3 cells, whereas inhibition of GROalpha by inhibitory RNA was associated with apoptosis, decreased growth rate, and marked up-regulation of the matrix protein fibulin-1. Forced expression of GROalpha was associated with decreased expression of fibulin-1. Expression of GROalpha mRNA was higher in primary adenocarcinomas (n = 132), adenomas (n = 32), and metastases (n = 52) than in normal colon epithelium (P < 0.001). These results were confirmed by real-time PCR and by immunohistochemistry. Samples of primary and metastatic colon cancer showed underexpression of fibulin-1 when compared with normal samples. There were no consistent changes in gene copy number of GROalpha or fibulin-1, implying a transcriptional basis for these findings. CONCLUSION: Elevated expression of GROalpha is frequent in adenocarcinoma of the colon and is associated with down-regulation of the matrix protein fibulin-1 in experimental models and in clinical samples. GROalpha overexpression abrogates contact inhibition in cell culture models, whereas inhibition of GROalpha expression is associated with apoptosis. Importantly, coexpression of fibulin-1 with GROalpha abrogates key aspects of the transformed phenotype, including tumor formation in a murine xenograft model. Targeting GRO proteins may provide new opportunities for treatment of colon cancer.


Subject(s)
Adenocarcinoma/genetics , Calcium-Binding Proteins/genetics , Chemokines, CXC/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/genetics , 3T3 Cells , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Cell Survival , Chemokine CXCL1 , Colonic Neoplasms/pathology , DNA, Neoplasm/genetics , Gene Expression Profiling , Humans , Mice , Neoplasm Transplantation , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Transplantation, Heterologous
17.
Cardiovasc Res ; 68(2): 259-67, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16054122

ABSTRACT

OBJECTIVE: Long-term cardiac memory (LTCM), expressed as a specific pattern of T-wave change on ECG, is associated with 1) reduced transient outward potassium current (I(to)), 2) reduced mRNA for the pore-forming protein of I(to), Kv4.3, 3) reduced cAMP response element binding protein (CREB), and 4) diminished binding to its docking site on the DNA, the cAMP response element (CRE). We hypothesized a causal link between the decrease of the transcription factor CREB and down-regulation of I(to) and one of its channel subunits, KChIP2, in LTCM. METHODS: After three weeks of left ventricular pacing to induce LTCM (8 paced, 7 sham control dogs), epicardial KChIP2 mRNA and protein levels were assessed by real-time PCR and Western blotting. Mimicking the CREB down-regulation in LTCM, CREB was knocked down in situ in other dogs using adenoviral anti-sense. Effects on the action potential notch, reflecting I(to), were investigated in situ using monophasic action potential (MAP) recordings and at the cellular level by the whole-cell patch clamp technique. CREB binding in the KChIP2 promoter region was ascertained by electrophoretic mobility-shift assays. RESULTS: In LTCM, epicardial KChIP2 mRNA and protein were reduced by 62% and 76%, respectively, compared to shams (p < 0.05). CREB binding by the canine KChIP2 promoter region was demonstrated. CREB knockdown led to disappearance of the phase1 notch in MAP and ablation of I(to). CONCLUSIONS: These results strengthen the hypothesis that down-regulation of CREB-mediated transcription underlies the attenuation of epicardial I(to) in LTCM. They also emphasize that ventricular pacing exerts effects at a subcellular level contributing to memory and conceivably to other forms of cardiac remodeling.


Subject(s)
Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Cyclic AMP Response Element-Binding Protein/metabolism , Myocardium/metabolism , Potassium Channels, Voltage-Gated/metabolism , Action Potentials , Animals , Blotting, Western/methods , Calcium-Binding Proteins/metabolism , Cardiac Pacing, Artificial , Cyclic AMP Response Element-Binding Protein/immunology , Dogs , Down-Regulation , Electrocardiography , Electrophoretic Mobility Shift Assay , Models, Animal , Oligonucleotides, Antisense/pharmacology , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/analysis , Potassium Channels, Voltage-Gated/genetics , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Ventricular Remodeling
18.
Biochem Biophys Res Commun ; 329(2): 624-31, 2005 Apr 08.
Article in English | MEDLINE | ID: mdl-15737631

ABSTRACT

T-cadherin is a 95kDa glycoprotein member of the cadherin family of adhesion molecules attached to the extracellular surface of the cell membrane through a glycosyl-phosphatidylinositol (GPI)-anchor. Whether a T-cadherin ectodomain apical targeting signal or the GPI-anchor itself targets this protein to the apical membrane is not known. Chimeras of the reporter EGFP and T-cadherin have demonstrated that a minimal construct consisting of the C-terminal 25 amino acids including the N690 (omega-site) of T-cadherin was sufficient to GPI-anchor the EGFP protein. However, efficient GPI-anchor with minimal secretion of the protein required an additional 5 residues (omega-1 to omega-5). The GPI-anchored chimeras fractionated to the Triton X-100 detergent insoluble fraction and were released to the cell culture supernatant by phosphoinositide-specific phospho-lipase C digestion. When expressed in MDCK cells, all GPI-anchored chimeras targeted to the basolateral membrane, while the T/N-chimera and the wild-type T-cadherin targeted to the apical membrane. Therefore, T-cadherin is an example of another rare GPI-anchored protein where the anchor itself is not sufficient for apical targeting in MDCK cells.


Subject(s)
Cadherins/metabolism , Glycosylphosphatidylinositols/metabolism , Kidney/cytology , Kidney/metabolism , Protein Transport/physiology , Animals , Cell Line , Dogs
19.
Br J Pharmacol ; 135(7): 1733-42, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11934814

ABSTRACT

1. Previous studies have suggested that neuronal apoptosis is the result of an abortive attempt to re-enter the cell cycle, and more recently the cyclin-dependent (CDKs) and the mitogen-activated protein (MAP) kinases, two superfamilies of kinases that influence and control cell cycle progression, have been implicated in neuronal apoptosis. 2. Here, to examine whether CDK/MAPK related pathways are involved in excitotoxicity, we studied the actions of various kinase inhibitors on apoptosis induced by the ionotropic glutamate (Glu) receptor agonist, kainate (KA), in primary cultures of murine cerebellar granule cells (CGCs). 3. KA-mediated neurotoxicity was concentration-dependent, as determined by a cell viability assay monitoring the reduction of 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyltetrazolium bromide (MTT), and largely apoptotic in nature, as shown by morphological examination and labelling of DNA fragmentation in situ using terminal deoxynucleotidyl transferase (TdT)-mediated dUTP digoxigenin nick-end labelling (TUNEL). 4. KA-mediated neurotoxicity and apoptosis was completely attenuated by the mixed CDK and MAP kinase inhibitor, olomoucine, in a concentration-dependent manner (50 - 600 microM), and partially by roscovitine (1 - 100 microM), a more selective CDK inihibitor. 5. The p38 MAP kinase inhibitor, SB203580 (1 - 100 microM), partially attenuated KA receptor-mediated apoptosis, as did the MAP kinase kinase inhibitors PD98509 (1 - 100 microM) and U0126 (1 - 100 microM). 6. These findings provide new evidence for a complex network of interacting pathways involving CDK/MAPK that control apoptosis downstream of KA receptor activation in excitotoxic neuronal cell death.


Subject(s)
Apoptosis/physiology , Cerebellar Cortex/cytology , Cyclin-Dependent Kinases/antagonists & inhibitors , Receptors, Kainic Acid/physiology , Animals , Cells, Cultured , Cerebellar Cortex/drug effects , Enzyme Inhibitors/pharmacology , Kainic Acid/pharmacology , Mice , Neurotoxicity Syndromes/etiology , Protein Kinase Inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...