Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Sci Rep ; 8(1): 15973, 2018 10 29.
Article in English | MEDLINE | ID: mdl-30374109

ABSTRACT

Secreted FGF binding proteins (FGFBP) mobilize locally-acting paracrine FGFs from their extracellular storage. Here, we report that FGFBP3 (BP3) modulates fat and glucose metabolism in mouse models of metabolic syndrome. BP3 knockout mice exhibited altered lipid metabolism pathways with reduced hepatic and serum triglycerides. In obese mice the expression of exogenous BP3 reduced hyperglycemia, hepatosteatosis and weight gain, blunted de novo lipogenesis in liver and adipose tissues, increased circulating adiponectin and decreased NEFA. The BP3 protein interacts with endocrine FGFs through its C-terminus and thus enhances their signaling. We propose that BP3 may constitute a new therapeutic to reverse the pathology associated with metabolic syndrome that includes nonalcoholic fatty liver disease and type 2 diabetes mellitus.


Subject(s)
Carbohydrate Metabolism/genetics , Carrier Proteins/genetics , Lipid Metabolism/genetics , Metabolic Syndrome/pathology , Adipose Tissue/metabolism , Animals , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/metabolism , Gluconeogenesis/genetics , Glucose Tolerance Test , Lipogenesis/genetics , Liver/metabolism , Metabolic Syndrome/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Protein Binding , STAT3 Transcription Factor/metabolism , Signal Transduction , Triglycerides/blood
2.
Breast Cancer Res Treat ; 166(3): 681-693, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28808806

ABSTRACT

PURPOSE: We previously identified small molecules that fit into a BRCA1-binding pocket within estrogen receptor-alpha (ERα), mimic the ability of BRCA1 to inhibit ERα activity ("BRCA1-mimetics"), and overcome antiestrogen resistance. One such compound, the hydrochloride salt of NSC35446 ("NSC35446.HCl"), also inhibited the growth of antiestrogen-resistant LCC9 tumor xenografts. The purpose of this study was to investigate the down-stream effects of NSC35446.HCl and its mechanism of action. METHODS: Here, we studied antiestrogen-resistant (LCC9, T47DCO, MCF-7/RR, LY2), ERα-negative (MDA-MB-231, HCC1806, MDA-MB-468), and antiestrogen-sensitive (MCF-7) cell lines. Techniques utilized include RNA-seq, qRT-PCR, cell growth analysis, cell-cycle analysis, Western blotting, luciferase reporter assays, TUNEL assays, in silico analysis of the IKKB gene, and ChIP assays. RESULTS: SC35446.HCl inhibited proliferation and induced apoptosis in antiestrogen-resistant LCC9, T47DCO, MCF-7/RR, and LY2 cells but not in ERα-negative breast cancer cell lines. IKKB (IKKß, IKBKB), an upstream activator of NF-κB, was identified as a BRCA1-mimetic-regulated gene based on an RNA-seq analysis. NSC35446.HCl inhibited IKKB, IKKA, and IKKG/NEMO mRNA and protein expression in LCC9 cells. NSC35446.HCl also inhibited NF-κB activity and expression of NF-κB target genes. In silico analysis of the IKKB promoter identified nine estrogen response element (ERE) half-sites and one ERE-like full-site. ChIP assays revealed that ERα was recruited to the ERE-like full-site and five of the nine half-sites and that ERα recruitment was inhibited by NSC35446.HCl in LCC9 and T47DCO cells. CONCLUSIONS: These studies identify functional EREs in the IKKB promoter and identify IKKB as an ERα and NSC35446.HCl-regulated gene, and they suggest that NF-κB and IKKB, which were previously linked to antiestrogen resistance, are targets for NSC35446.HCl in reversing antiestrogen resistance.


Subject(s)
Breast Neoplasms/drug therapy , Estrogen Antagonists/administration & dosage , Estrogen Receptor alpha/genetics , I-kappa B Kinase/genetics , Apoptosis/genetics , BRCA1 Protein/antagonists & inhibitors , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/genetics , Drug Resistance, Neoplasm/genetics , Estrogens/genetics , Estrogens/metabolism , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , NF-kappa B/genetics , Promoter Regions, Genetic
3.
Oncotarget ; 6(38): 40388-404, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26575173

ABSTRACT

Previous studies indicate that BRCA1 protein binds to estrogen receptor-alpha (ER) and inhibits its activity. Here, we found that BRCA1 over-expression not only inhibits ER activity in anti-estrogen-resistant LCC9 cells but also partially restores their sensitivity to Tamoxifen. To simulate the mechanism of BRCA1 inhibition of ER in the setting of Tamoxifen resistance, we created a three-dimensional model of a BRCA1-binding cavity within the ER/Tamoxifen complex; and we screened a pharmacophore database to identify small molecules that could fit into this cavity. Among the top 40 "hits", six exhibited potent ER inhibitory activity in anti-estrogen-sensitive MCF-7 cells and four of the six exhibited similar activity (IC50 ≤ 1.0 µM) in LCC9 cells. We validated the model by mutation analysis. Two representative compounds (4631-P/1 and 35466-L/1) inhibited ER-dependent cell proliferation in Tamoxifen-resistant cells (LCC9 and LCC2) and partially restored sensitivity to Tamoxifen. The compounds also disrupted the association of BRCA1 with ER. In electrophoretic mobility shift assays, the compounds caused dissociation of ER from a model estrogen response element. Finally, a modified form of compound 35446 (hydrochloride salt) inhibited growth of LCC9 tumor xenografts at non-toxic concentrations. These results identify a novel group of small molecules that can overcome Tamoxifen resistance.


Subject(s)
BRCA1 Protein/antagonists & inhibitors , Benzophenones/pharmacology , Breast Neoplasms/drug therapy , Chalcones/pharmacology , Drug Resistance, Neoplasm/drug effects , Estrogen Antagonists/chemistry , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Piperidines/pharmacology , Tamoxifen/pharmacology , Animals , Apoptosis/drug effects , Benzophenones/chemistry , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Chalcones/chemistry , Electrophoretic Mobility Shift Assay , Estrogens/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoprecipitation , Mice , Mice, Inbred BALB C , Mice, Nude , Piperidines/chemistry , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Mol Endocrinol ; 28(12): 1971-86, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25264941

ABSTRACT

CONTEXT: Resistance to conventional antiestrogens is a major cause of treatment failure and, ultimately, death in breast cancer. OBJECTIVE: The objective of the study was to identify small-molecule estrogen receptor (ER)-α antagonists that work differently from tamoxifen and other selective estrogen receptor modulators. DESIGN: Based on in silico screening of a pharmacophore database using a computed model of the BRCA1-ER-α complex (with ER-α liganded to 17ß-estradiol), we identified a candidate group of small-molecule compounds predicted to bind to a BRCA1-binding interface separate from the ligand-binding pocket and the coactivator binding site of ER-α. Among 40 candidate compounds, six inhibited estradiol-stimulated ER-α activity by at least 50% in breast carcinoma cells, with IC50 values ranging between 3 and 50 µM. These ER-α inhibitory compounds were further studied by molecular and cell biological techniques. RESULTS: The compounds strongly inhibited ER-α activity at concentrations that yielded little or no nonspecific toxicity, but they produced only a modest inhibition of progesterone receptor activity. Importantly, the compounds blocked proliferation and inhibited ER-α activity about equally well in antiestrogen-sensitive and antiestrogen-resistant breast cancer cells. Representative compounds disrupted the interaction of BRCA1 and ER-α in the cultured cells and blocked the interaction of ER-α with the estrogen response element. However, the compounds had no effect on the total cellular ER-α levels. CONCLUSIONS: These findings suggest that we have identified a new class of ER-α antagonists that work differently from conventional antiestrogens (eg, tamoxifen and fulvestrant).


Subject(s)
Estrogen Antagonists/pharmacology , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism , Cell Line, Tumor , Humans , Protein Binding , Selective Estrogen Receptor Modulators/pharmacology , Surface Plasmon Resonance , Tamoxifen/pharmacology
5.
J Med Chem ; 56(21): 8404-21, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24047231

ABSTRACT

Developing novel and selective compounds that desensitize α4ß2 nicotinic acetylcholine receptors (nAChRs) could provide new effective treatments for nicotine addiction, as well as other disorders. Here we report a new class of nAChR ligands that display high selectivity and picomolar binding affinity for α4ß2 nicotinic receptors. The novel compounds have Ki values in the range of 0.031-0.26 nM and properties that should make them good candidates as drugs acting in the CNS. The selected lead compound 1 (VMY-2-95) binds with high affinity and potently desensitizes α4ß2 nAChRs. At a dose of 3 mg/kg, compound 1 significantly reduced rat nicotine self-administration. The overall results support further characterizations of compound 1 and its analogues in preclinical models of nicotine addiction and perhaps other disorders involving nAChRs.


Subject(s)
Azetidines/pharmacology , Drug Discovery , Pyridines/pharmacology , Receptors, Nicotinic/metabolism , Azetidines/chemical synthesis , Azetidines/chemistry , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Ligands , Models, Molecular , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Software , Structure-Activity Relationship
6.
Oncotarget ; 3(10): 1220-35, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23100451

ABSTRACT

Dysregulation of the pathways that preserve mitochondrial integrity hallmarks many human diseases including diabetes, neurodegeration, aging and cancer. The mitochondrial citrate transporter gene, SLC25A1 or CIC, maps on chromosome 22q11.21, a region amplified in some tumors and deleted in developmental disorders known as velo-cardio-facial- and DiGeorge syndromes. We report here that in tumor cells CIC maintains mitochondrial integrity and bioenergetics, protects from mitochondrial damage and circumvents mitochondrial depletion via autophagy, hence promoting proliferation. CIC levels are increased in human cancers and its inhibition has anti-tumor activity, albeit with no toxicity on adult normal tissues. The knock-down of the CIC gene in zebrafish leads to mitochondria depletion and to proliferation defects that recapitulate features of human velo-cardio-facial syndrome, a phenotype rescued by blocking autophagy. Our findings reveal that CIC maintains mitochondrial homeostasis in metabolically active, high proliferating tissues and imply that this protein is a therapeutic target in cancer and likely, in other human diseases.


Subject(s)
Autophagy , Breast Neoplasms/pathology , Cell Proliferation , Homeostasis , Mitochondria/pathology , Repressor Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Blotting, Western , Breast Neoplasms/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/metabolism , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Repressor Proteins/genetics , Zebrafish/genetics
7.
J Med Chem ; 54(3): 809-16, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21241062

ABSTRACT

Isothiocyanates (ITCs) derived from cruciferous vegetables induce apoptosis in cancer cells. We demonstrate that certain naturally occurring ITCs selectively deplete mutant p53 but not the wild-type and do so via a transcription-independent mechanism. Direct p53 binding followed by conformational changes appears to be a mechanism by which mutant p53 is depleted. Structure-activity relationship studies (SARs) using naturally occurring and synthetic ITCs show that depletion is influenced by the ITC side-chain moiety. Furthermore, we show that cells with p53 mutations are more sensitive to cytotoxicity induced by phenethyl isothiocyanate (PEITC) than those with the wild-type protein. 2,2-Diphenylethyl ITC, a synthetic ITC, is one of the most potent depletors of mutant p53 studies and induces apoptosis to the greatest extent in mutant p53 breast cancer cells. Collectively, this study shows that mutant p53 depletion may be an important novel target for cancer chemoprevention and therapy by natural and synthetic ITCs.


Subject(s)
Anticarcinogenic Agents/chemistry , Isothiocyanates/chemistry , Tumor Suppressor Protein p53/metabolism , Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cysteine/chemistry , Drug Screening Assays, Antitumor , Humans , Isothiocyanates/pharmacology , Mutation , Protein Conformation , Structure-Activity Relationship , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
8.
PLoS One ; 5(5): e10599, 2010 May 12.
Article in English | MEDLINE | ID: mdl-20485683

ABSTRACT

Gamma interferon Inducible Lysosomal Thiol reductase (GILT) is a unique lysosomal reductase that reduces disulfide bonds of endocytosed proteins. Lack of GILT clearly decreases CD4 T cell-antigen specific responses against some epitopes of antigens containing disulfide bonds, but not to proteins with few or no disulfide bridges. Hence, global impact of GILT on antigen presentation is currently not well understood. We used Nano-LC-ESI-MS/MS to investigate how GILT affects diversity of self-peptides presented by MHC class II molecules. Surprisingly, the repertoire of self-peptides in the absence of GILT does not appear to be significantly different, as only few peptide species (approximately 2%) were found to be the unique indicators of GILT's presence or absence. In the absence of GILT about thirty peptide species (approximately 5%) were found either uniquely or fourteen to hundred fold more abundantly expressed than in the presence of GILT. Our data indicate that GILT has limited yet unexpected effect on self-peptide species presented by MHC class II antigens.


Subject(s)
Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/immunology , Oxidoreductases/immunology , Peptides/chemistry , Peptides/immunology , Spectrometry, Mass, Electrospray Ionization , Amino Acid Sequence , Animals , Antigen Presentation/immunology , Histocompatibility Antigens Class II/isolation & purification , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Oxidoreductases/deficiency , Oxidoreductases Acting on Sulfur Group Donors , Peptides/isolation & purification , Protein Binding , Reproducibility of Results , Spleen/enzymology , Spleen/immunology
9.
Mol Endocrinol ; 24(1): 76-90, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19887647

ABSTRACT

Inherited mutations of the breast cancer susceptibility gene BRCA1 confer a high risk for breast cancer development. The (300)RXKK and (266)KXK motifs have been identified previously as sites for acetylation of the estrogen receptor-alpha (ER-alpha), and (302)K was also found to be a site for BRCA1-mediated mono-ubiquitination of ER-alpha in vitro. Here we show that ER-alpha proteins with single or double lysine mutations of these motifs (including K303R, a cancer-associated mutant) are resistant to inhibition by BRCA1, even though the mutant ER-alpha proteins retain the ability to bind to BRCA1. We also found that BRCA1 overexpression reduced and knockdown increased the level of acetylated wild-type ER-alpha, without changing the total ER-alpha protein level. Increased acetylation of ER-alpha due to BRCA1 small interfering RNA was dependent upon phosphatidylinositol 3-kinase/Akt signaling and on up-regulation of the coactivator p300. In addition, using an in vitro acetylation assay, we found that in vitro-translated wild-type BRCA1 but not a cancer-associated point mutant (C61G) inhibited p300-mediated acetylation of ER-alpha. Furthermore, BRCA1 overexpression increased the levels of mono-ubiquitinated ER-alpha protein, and a BRCA1 mutant that is defective for ubiquitin ligase activity but retains other BRCA1 functions (I26A) did not ubiquitinate ER-alpha or repress its activity in vivo. Finally, ER-alpha proteins with mutations of the (300)RXKK or (266)KXK motifs showed modest or no BRCA1-induced ubiquitination. We propose a model in which BRCA1 represses ER-alpha activity, in part, by regulating the relative degree of acetylation vs. ubiquitination of ER-alpha.


Subject(s)
BRCA1 Protein/physiology , Estrogen Receptor alpha/metabolism , Genes, BRCA1 , Protein Processing, Post-Translational , Acetylation , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Breast Neoplasms , Cell Line, Tumor , Estradiol/pharmacology , Female , Gene Knockdown Techniques , Genes, Reporter , Humans , Male , Mutation , Phosphoinositide-3 Kinase Inhibitors , Prostatic Neoplasms , Protein Binding , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Interfering , Transfection , Ubiquitination , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
10.
Cancer Res ; 69(1): 102-10, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19117992

ABSTRACT

Prostate cancer (PCa) frequently develops antiapoptotic mechanisms and acquires resistance to anticancer drugs. Therefore, identifying PCa drug resistance determinants should facilitate designing more effective chemotherapeutic regimens. Recently, we described that the PCPH protein becomes highly expressed in human prostatic intraepithelial neoplasia and in PCa, and that the functional interaction between PCPH and protein kinase Cdelta (PKCdelta) increases the invasiveness of human PCa. Here, we report that the functional interaction between PCPH and a different PKC isoform, PKCalpha, confers resistance against cisplatin-induced apoptosis to PCa cells. This interaction elicits a mechanism ultimately resulting in the posttranslational stabilization and subsequent elevated expression of Bcl-2. Stable knockdown of either PCPH, mt-PCPH, or PKCalpha in PCa cells decreased Ser70-phosphorylated Bcl-2 and total Bcl-2 protein, thereby increasing their cisplatin sensitivity. Conversely, forced expression of the PCPH protein or, in particular, of the mt-PCPH oncoprotein increased the levels of phosphorylated PKCalpha concurrently with those of Ser70-phosphorylated and total Bcl-2 protein, thus promoting cisplatin resistance. Consistently, Bcl-2 knockdown sensitized PCa cells to cisplatin treatment and, more importantly, reversed the cisplatin resistance of PCa cells expressing the mt-PCPH oncoprotein. Moreover, reexpression of Bcl-2 in PCPH/mt-PCPH knockdown PCa cells reversed the cisplatin sensitization caused by PCPH or mt-PCPH down-regulation. These findings identify PCPH and mt-PCPH as important participants in the chemotherapy response of PCa cells, establish a role for PCPH-PKCalpha-Bcl-2 functional interactions in the drug response process, and imply that targeting PCPH expression before, or simultaneously with, chemotherapy may improve the treatment outcome for PCa patients.


Subject(s)
Apoptosis/drug effects , Cisplatin/pharmacology , Oncogene Proteins/biosynthesis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Cell Line, Tumor , Down-Regulation/drug effects , Drug Resistance, Neoplasm , Humans , Male , Oncogene Proteins/genetics , Phosphorylation , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/genetics , Pyrophosphatases , RNA, Small Interfering/genetics
11.
Protein Sci ; 17(7): 1232-40, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18467496

ABSTRACT

Alanine scanning has been widely employed as a method of identifying side chains that play important roles in protein-protein and protein-peptide interactions. Here we show how an analogous and complementary technique, hydrophile scanning, can provide additional insight on such interactions. Mutation of a wild-type residue to alanine removes most of the side-chain atoms, and the effect of this removal is typically interpreted to indicate contribution of the deleted side chain to the stability of the complex. Hydrophile scanning involves systematic mutation of wild-type residues to a cationic or anionic residue (lysine or glutamic acid, in this case). We find that the results of these mutations provide insights on interactions between polypeptide surfaces that are complementary to the information obtained via alanine scanning. We have applied this technique to a peptide that corresponds to the BH3 domain of the pro-apoptotic protein Bim. The wild-type Bim BH3 domain binds strongly to the anti-apoptotic proteins Bcl-x(L) and Mcl-1. Combining information from the alanine, lysine, and glutamic acid scans has enabled us to identify Bim BH3 domain mutants containing only two or three sequence changes that bind very selectively either to Bcl-x(L) or Mcl-1. Our findings suggest that hydrophile scanning may prove to be a broadly useful tool for revealing sources of protein-protein recognition and for engineering selectivity into natural sequences.


Subject(s)
Alanine/chemistry , Apoptosis Regulatory Proteins/chemistry , Membrane Proteins/chemistry , Proto-Oncogene Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Circular Dichroism , Fluorescence Polarization , Membrane Proteins/genetics , Mutation , Protein Binding , Proto-Oncogene Proteins/genetics
12.
Brain Res Bull ; 75(5): 598-609, 2008 Mar 28.
Article in English | MEDLINE | ID: mdl-18355636

ABSTRACT

Excitotoxicity involves over activation of brain excitatory glutamate receptors and has been implicated in neurological, neurodegenerative and neuropsychiatric diseases. Metabolism of arachidonic acid (AA) through the phospholipase A(2) (PLA(2))/prostaglandin-endoperoxide synthase (PTGS) pathway is increased after excitotoxic stimulation. However, the individual roles of the PTGS isoforms in this process are not well established. We assessed the role of the PTGS isoforms in the process of excitotoxicity by exposing mice deficient in either PTGS-1 (PTGS-1(-/-)) or PTGS-2 (PTGS-2(-/-)) to the prototypic excitotoxin, kainic acid (KA). Seizure intensity and neuronal damage were significantly elevated in KA-exposed PTGS-2(-/-), but not in PTGS-1(-/-), mice. The increased susceptibility was not associated with an alteration in KA receptor binding activity or mediated through the CB1 endocannabinoid receptor. The frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) was decreased in the CA1 pyramidal neurons of PTGS-2(-/-) mice, suggesting an alteration of GABAergic function. In wild-type mice, six weeks treatment with the PTGS-2 selective inhibitor celecoxib recapitulated the increased susceptibility to KA-induced excitotoxicity observed in PTGS-2(-/-) mice, further supporting the role of PTGS-2 in the excitotoxic process. The increased susceptibility to KA was also associated with decreased brain levels of PGE(2), a biomarker of PTGS-2 activity. Our results suggest that PTGS-2 activity and its specific products may modulate neuronal excitability by affecting GABAergic neurotransmission. Further, inhibition of PTGS-2, but not PTGS-1, may increase the susceptibility to seizures.


Subject(s)
Cyclooxygenase 2/deficiency , Kainic Acid , Seizures/chemically induced , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism , Analysis of Variance , Animals , Autoradiography , Celecoxib , Chromatography, High Pressure Liquid/methods , Cyclooxygenase 1/deficiency , Enzyme-Linked Immunosorbent Assay/methods , Hippocampus/pathology , Male , Membrane Potentials , Membrane Proteins/deficiency , Mice , Mice, Knockout , Neurons/physiology , Patch-Clamp Techniques/methods , Piperidines/administration & dosage , Pyrazoles/administration & dosage , Pyrazoles/blood , Pyrazoles/pharmacology , Seizures/blood , Seizures/pathology , Seizures/prevention & control , Statistics, Nonparametric , Sulfonamides/blood , Sulfonamides/pharmacology
13.
J Comb Chem ; 10(2): 204-15, 2008.
Article in English | MEDLINE | ID: mdl-18275161

ABSTRACT

We describe the use of parallel and split-and-mix library synthesis strategies for exploration of structure-activity relationships among peptidic foldamer ligands for the BH3-recognition cleft of the anti-apoptotic protein Bcl-xL. This effort began with a chimeric (alpha/beta+alpha)-peptide oligomer (composed of an alpha/beta-peptide segment and an alpha-peptide segment) that we previously identified to bind tightly to the target cleft on Bcl-xL. The side chains that interact with Bcl-xL were varied in a 1000-member one-bead-one-compound library. Fluorescence polarization (FP) screening identified four new analogues with binding affinities similar to that of the lead compound but no analogues with enhanced affinity. These results suggested that significant improvements in affinity were unlikely in this series. We then used library synthesis to examine backbone variations in the C-terminal alpha-peptide segment of the lead compound. These studies provided an opportunity for direct comparison of parallel and split-and-mix synthesis formats for foldamer libraries with respect to synthetic variability and assay sensitivity. We found that compounds from both the parallel and one-bead-one-compound libraries could be reliably screened in a competition FP assay without purification of library members. Our findings should facilitate the use of combinatorial library synthesis for exploration of foldamers as inhibitors of protein-protein interactions.


Subject(s)
Proteins/chemistry , Binding Sites , Chromatography, High Pressure Liquid , Combinatorial Chemistry Techniques , Ligands , Proteins/metabolism , Spectrophotometry, Ultraviolet , Structure-Activity Relationship
14.
Biochem Biophys Res Commun ; 365(4): 833-9, 2008 Jan 25.
Article in English | MEDLINE | ID: mdl-18039462

ABSTRACT

Aggregation of alpha-synuclein is known to be a causal factor in the genesis of Parkinson's disease and Dementia with Lewy bodies. Duplication and/or triplication and mutation of the alpha-synuclein gene are associated with sporadic and familial Parkinson's disease. Synucleinopathies appear to primarily affect dopaminergic neurons. The present studies investigate the role of dopamine in alpha-synuclein aggregation through NMR. Dopamine causes aggregation of both wild type and A53T mutant alpha-synuclein in a temperature-dependent manner, but the mutant A53T shows a greater propensity to aggregate in the presence of dopamine only at 37 degrees C. A single point mutation in the alpha-synuclein A53T mutant gene results in a structural change in the protein and drastically increases its propensity to aggregate in the presence of dopamine. The present data indicate that mutation in the alpha-synuclein gene may predispose the protein to dopamine-induced aggregation, thereby contributing to disease pathogenesis.


Subject(s)
Dopamine/administration & dosage , Neurons/chemistry , Neurons/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Cell Line , Dose-Response Relationship, Drug , Humans , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Mutation , Neurons/drug effects , alpha-Synuclein/genetics
15.
J Am Chem Soc ; 129(49): 15279-94, 2007 Dec 12.
Article in English | MEDLINE | ID: mdl-17999504

ABSTRACT

XIAP is a central apoptosis regulator that inhibits apoptosis by binding to and inhibiting the effectors caspase-3/-7 and an initiator caspase-9 through its BIR2 and BIR3 domains, respectively. Smac protein in its dimeric form effectively antagonizes XIAP by concurrently targeting both its BIR2 and BIR3 domains. We report the design, synthesis, and characterization of a nonpeptide, cell-permeable, bivalent small-molecule (SM-164) which mimics Smac protein for targeting XIAP. Our study shows that SM-164 binds to XIAP containing both BIR domains with an IC50 value of 1.39 nM, being 300 and 7000 times more potent than its monovalent counterparts and the natural Smac AVPI peptide, respectively. SM-164 concurrently interacts with both BIR domains in XIAP and functions as an ultrapotent antagonist of XIAP in both cell-free functional and cell-based assays. SM-164 targets cellular XIAP and effectively induces apoptosis at concentrations as low as 1 nM in the HL-60 leukemia cell line. The potency of bivalent SM-164 in binding, functional, and cellular assays is 2-3 orders of magnitude higher than its corresponding monovalent Smac mimetics.


Subject(s)
Antineoplastic Agents/chemistry , Biomimetic Materials/chemistry , Intracellular Signaling Peptides and Proteins/chemistry , Mitochondrial Proteins/chemistry , X-Linked Inhibitor of Apoptosis Protein/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Binding, Competitive , Biomimetic Materials/chemical synthesis , Biomimetic Materials/pharmacology , Caspases/metabolism , Drug Design , HL-60 Cells , Humans , Inhibitory Concentration 50 , Intracellular Signaling Peptides and Proteins/metabolism , Kinetics , Mitochondrial Proteins/metabolism , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Tertiary , X-Linked Inhibitor of Apoptosis Protein/metabolism
16.
Epilepsia ; 48 Suppl 5: 19-26, 2007.
Article in English | MEDLINE | ID: mdl-17910577

ABSTRACT

Seizure incidence during the neonatal period is higher than any other period in the lifespan, yet we know little about this period in terms of the effect of seizures or of the drugs used in their treatment. The fact that several antiepileptic drugs (AEDs) induce pronounced apoptotic neuronal death in specific regions of the immature brain prompts a search for AEDs that may be devoid of this action. Furthermore, there is a clear need to find out if a history of seizures alters the proapoptotic action of the AEDs. Our studies are aimed at both of these issues. Phenytoin, valproate, phenobarbital, and MK801 each induced substantial regionally specific cell death, whereas levetiracetam even in high doses (up to 1,500 mg/kg) did not have this action. In view of our previously findings of neuroprotective actions of repeated seizures in the adult brain, we also examined repeated seizures for a possible antiapoptotic action in the infant rat. Rat pups were preexposed to electroshock seizures (ECS) for 3 days (age 5-7 days) before receiving MK801 on day 7. The effect of ECS, which was consistently a 30% decrease in MK801-induced programmed cell death (PCD), suggests that repeated seizures can exert an antiapoptotic action in the infant brain. In contrast, PCD induced by valproate was not attenuated by ECS preexposure, suggesting that valproate-induced PCD is mechanistically distinct from that induced by MK801 and may not be activity-dependent. Presently, we do not know if this neuroprotective effect of seizures is deleterious or beneficial. If the seizures also enhance the survival of neurons that are destined to undergo naturally occurring PCD, early childhood seizures may have deleterious effects by preventing this necessary component of normal development. While this effect of seizures might be counteracted by AEDs, the fact that several AEDs shift the PCD to the other extreme, and trigger excessive neuronal cell loss, raises concern about whether the drug therapy may be more detrimental than the seizures. In this context, it is encouraging that we have identified at least one AED that is devoid of a proapoptotic action in the infant brain, even in high doses. It is now important to evaluate the long-term consequences of the changes in PCD in infancy by examining behavioral outcomes and seizure susceptibility in the AED- and seizure-exposed neonates when they reach adulthood.


Subject(s)
Anticonvulsants/adverse effects , Brain/growth & development , Seizures/chemically induced , Seizures/physiopathology , Animals , Animals, Newborn , Anticonvulsants/therapeutic use , Apoptosis/drug effects , Apoptosis/physiology , Basal Ganglia/drug effects , Basal Ganglia/physiopathology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Body Weight/drug effects , Brain/drug effects , Dizocilpine Maleate/pharmacology , Electroshock , Levetiracetam , Piracetam/adverse effects , Piracetam/analogs & derivatives , Piracetam/therapeutic use , Rats , Rats, Sprague-Dawley , Seizures/drug therapy , Thalamus/drug effects , Thalamus/physiopathology
17.
Chembiochem ; 8(8): 903-16, 2007 May 25.
Article in English | MEDLINE | ID: mdl-17503422

ABSTRACT

Protein-protein interactions play crucial roles in cell-signaling events and are often implicated in human disease. Molecules that bind tightly to functional protein-surface sites and show high stability to degradative enzymes could be valuable pharmacological tools for dissection of cell-signaling networks and might ultimately lead to therapeutic agents. We recently described oligomers containing both alpha- and beta-amino acid residues that bind tightly to the BH3 recognition site of the anti-apoptotic protein Bcl-x(L). The oligomers with highest affinity had a nine-residue N-terminal segment with a 1:1 alpha:beta residue repeat and a six-residue C-terminal segment containing exclusively proteinogenic alpha-residues. The N-terminal portions of such (alpha/beta+alpha)-peptides are highly resistant to proteolysis, but the C-terminal alpha-segments are susceptible. This study emerged from efforts to modify the alpha-segment in an (alpha/beta+alpha)-peptide in a way that would diminish proteolytic degradation but retain high affinity for Bcl-x(L). Some of the oligomers reported here could prove useful in certain biological applications, particularly those for which extended incubation in a biological milieu is required.


Subject(s)
Amino Acids/chemistry , Peptide Fragments/chemistry , Peptide Hydrolases/chemistry , Proto-Oncogene Proteins/chemistry , bcl-X Protein/chemistry , Binding Sites , Binding, Competitive , Molecular Structure , Peptide Library , Protein Folding , Protein Structure, Secondary , Time Factors
18.
J Am Chem Soc ; 129(1): 139-54, 2007 Jan 10.
Article in English | MEDLINE | ID: mdl-17199293

ABSTRACT

The development of molecules that bind to specific protein surface sites and inhibit protein-protein interactions is a fundamental challenge in molecular recognition. New strategies for approaching this challenge could have important long-term ramifications in biology and medicine. We are exploring the concept that unnatural oligomers with well-defined conformations ("foldamers") can mimic protein secondary structural elements and thereby block specific protein-protein interactions. Here, we describe the identification and analysis of helical peptide-based foldamers that bind to a specific cleft on the anti-apoptotic protein Bcl-xL by mimicking an alpha-helical BH3 domain. Initial studies, employing a fluorescence polarization (FP) competition assay, revealed that among several alpha/beta- and beta-peptide foldamer backbones only alpha/beta-peptides intended to adopt 14/15-helical secondary structure display significant binding to Bcl-xL. The most tightly binding Bcl-xL ligands are chimeric oligomers in which an N-terminal alpha/beta-peptide segment is fused to a C-terminal alpha-peptide segment ((alpha/beta + alpha)-peptides)). Sequence-affinity relationships were probed via standard and nonstandard techniques (alanine scanning and hydrophile scanning, respectively), and the results allowed us to construct a computational model of the ligand/Bcl-xL complex. Analytical ultracentrifugation with a high-affinity (alpha/beta + alpha)-peptide established 1:1 ligand:Bcl-xL stoichiometry under FP assay conditions. Binding selectivity studies with the most potent (alpha/beta + alpha)-peptide, conducted via surface plasmon resonance measurements, revealed that this ligand binds tightly to Bcl-w as well as to Bcl-xL, while binding to Bcl-2 is somewhat weaker. No binding could be detected with Mcl-1. We show that our most potent (alpha/beta + alpha)-peptide can induce cytochrome C release from mitochondria, an early step in apoptosis, in cell lysates, and that this activity is dependent upon inhibition of protein-protein interactions involving Bcl-xL.


Subject(s)
Apoptosis/drug effects , Drug Design , Peptides/chemistry , Peptides/pharmacology , bcl-X Protein/antagonists & inhibitors , Alanine/chemistry , Amino Acid Sequence , Cytochromes c/metabolism , Humans , Ligands , Mitochondria/drug effects , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology , Structure-Activity Relationship
19.
J Med Chem ; 49(21): 6139-42, 2006 Oct 19.
Article in English | MEDLINE | ID: mdl-17034116

ABSTRACT

A structure-based approach was employed to design a new class of small-molecule inhibitors of Bcl-2. The most potent compound 5 (TW-37) binds to Bcl-2 with a K(i) value of 290 nM and also to Bcl-xL and Mcl-1 with high affinities. Compound 5 potently inhibits cell growth in PC-3 prostate cancer cells with an IC(50) value of 200 nM and effectively induces apoptosis in a dose-dependent manner.


Subject(s)
Apoptosis , Benzamides/chemical synthesis , Gossypol/analogs & derivatives , Gossypol/chemical synthesis , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfones/chemical synthesis , Benzamides/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Gossypol/pharmacology , Humans , Models, Molecular , Stereoisomerism , Structure-Activity Relationship , Sulfones/pharmacology
20.
J Biol Chem ; 281(13): 8600-6, 2006 Mar 31.
Article in English | MEDLINE | ID: mdl-16443602

ABSTRACT

The induction of apoptosis by p53 in response to cellular stress is its most conserved function and crucial for p53 tumor suppression. We recently reported that p53 directly induces oligomerization of the BH1,2,3 effector protein Bak, leading to outer mitochondrial membrane permeabilization (OMMP) with release of apoptotic activator proteins. One important mechanism by which p53 achieves OMMP is by forming an inhibitory complex with the anti-apoptotic BclXL protein. In contrast, the p53 complex with the Bcl2 homolog has not been interrogated. Here we have undertaken a detailed characterization of the p53-Bcl2 interaction using structural, biophysical, and mutational analyses. We have identified the p53 DNA binding domain as the binding interface for Bcl2 using solution NMR. The affinity of the p53-Bcl2 complex was determined by surface plasmon resonance analysis (BIAcore) to have a dominant component KD 535 +/- 24 nm. Moreover, in contrast to wild type p53, endogenous missense mutants of p53 are unable to form complexes with endogenous Bcl2 in human cancer cells. Functionally, these mutants are all completely or strongly compromised in mediating OMMP, as measured by cytochrome c release from isolated mitochondria. These data implicate p53-Bcl2 complexes in contributing to the direct mitochondrial p53 pathway of apoptosis and further support the notion that the DNA binding domain of p53 is a dual function domain, mediating both its transactivation function and its direct mitochondrial apoptotic function.


Subject(s)
DNA-Binding Proteins/metabolism , Mitochondria/metabolism , Mutation , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/metabolism , Binding Sites , Blotting, Western , Camptothecin/pharmacology , Cell Line, Tumor , Centrifugation, Density Gradient , Cytochromes c/metabolism , DNA-Binding Proteins/chemistry , Enzyme Inhibitors/pharmacology , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Permeability , Precipitin Tests , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/genetics , Structure-Activity Relationship , Surface Plasmon Resonance , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...