Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
J Biol Chem ; 293(26): 10026-10040, 2018 06 29.
Article in English | MEDLINE | ID: mdl-29739850

ABSTRACT

Asthma is a chronic inflammatory disease of the lungs and airways and one of the most burdensome of all chronic maladies. Previous studies have established that expression of experimental and human asthma requires the IL-4/IL-13/IL-4 receptor α (IL-4Rα) signaling pathway, which activates the transcription factor STAT6. However, no small molecules targeting this important pathway are currently in clinical development. To this end, using a preclinical asthma model, we sought to develop and test a small-molecule inhibitor of the Src homology 2 domains in mouse and human STAT6. We previously developed multiple peptidomimetic compounds on the basis of blocking the docking site of STAT6 to IL-4Rα and phosphorylation of Tyr641 in STAT6. Here, we expanded the scope of our initial in vitro structure-activity relationship studies to include central and C-terminal analogs of these peptides to develop a lead compound, PM-43I. Conducting initial dose range, toxicity, and pharmacokinetic experiments with PM-43I, we found that it potently inhibits both STAT5- and STAT6-dependent allergic airway disease in mice. Moreover, PM-43I reversed preexisting allergic airway disease in mice with a minimum ED50 of 0.25 µg/kg. Of note, PM-43I was efficiently cleared through the kidneys with no long-term toxicity. We conclude that PM-43I represents the first of a class of small molecules that may be suitable for further clinical development against asthma.


Subject(s)
Asthma/drug therapy , Molecular Targeted Therapy , STAT5 Transcription Factor/chemistry , STAT5 Transcription Factor/metabolism , STAT6 Transcription Factor/chemistry , STAT6 Transcription Factor/metabolism , Small Molecule Libraries/pharmacology , Animals , Asthma/immunology , Asthma/metabolism , Cell Line , Drug Evaluation, Preclinical , Female , Humans , Lung/drug effects , Lung/immunology , Lung/metabolism , Mice , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use , Structure-Activity Relationship , src Homology Domains
2.
Int J Radiat Oncol Biol Phys ; 100(4): 1034-1043, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29485045

ABSTRACT

PURPOSE: To determine the role of macrophage polarization on the response of inflammatory breast cancer (IBC) cells to radiation and whether modulation of macrophage plasticity can alter radiation response. METHODS AND MATERIALS: The human THP-1 monocyte cell line and primary human monocytes isolated from peripheral blood mononuclear cells were differentiated into macrophages and polarized to either an "antitumor" (M1) or a "protumor" (M2) phenotype. These polarized macrophages were co-cultured with IBC cells (SUM149, KPL4, MDA-IBC3, or SUM190) without direct contact for 24 hours, then subjected to irradiation (0, 2, 4, or 6 Gy). Interleukin (IL)4/IL13-induced activation of STAT6 signaling was measured by Western blotting of phospho-STAT6 (Tyr641), and expression of M2 polarization gene markers (CD206, fibronectin, and CCL22) was measured by quantitative polymerase chain reaction. RESULTS: Expression of M2 polarization markers was higher in M2-polarized macrophages after IL4/IL13 treatment than in control (M0) or M1-polarized macrophages. Co-culture of IBC cell lines with M1-polarized THP-1 macrophages mediated radiosensitivity of IBC cells, whereas co-culture with M2-polarized macrophages mediated radioresistance. Phosphopeptide mimetic PM37, targeting the SH2 domain of STAT6, prevented and reversed IL4/IL13-mediated STAT6 phosphorylation (Tyr641) and decreased the expression of M2 polarization markers. Pretreatment of M2-THP1 macrophages with PM37 reduced the radioresistance they induced in IBC cells after co-culture. Targeted proteomics analysis of IBC KPL4 cells using a kinase antibody array revealed induction of protein kinase C zeta (PRKCZ) in these cells only after co-culture with M2-THP1 macrophages, which was prevented by PM37 pretreatment. KPL4 cells with stable short hairpin RNA knockdown of PRKCZ exhibited lower radioresistance after M2-THP1 co-culture. CONCLUSIONS: These data suggest that inhibition of M2 polarization of macrophages by PM37 can prevent radioresistance of IBC by down-regulating PRKCZ.


Subject(s)
Cell Polarity/drug effects , Inflammatory Breast Neoplasms/radiotherapy , Interleukin-13/antagonists & inhibitors , Interleukin-4/antagonists & inhibitors , Macrophages/drug effects , Protein Kinase C/metabolism , Radiation Tolerance , STAT6 Transcription Factor/antagonists & inhibitors , Biomimetic Materials/pharmacology , Cell Line, Tumor , Cell Polarity/physiology , Chemokine CCL22/genetics , Chemokine CCL22/metabolism , Coculture Techniques/methods , Enzyme Induction , Female , Fibronectins/genetics , Fibronectins/metabolism , Genetic Markers , Humans , Inflammatory Breast Neoplasms/metabolism , Inflammatory Breast Neoplasms/pathology , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Macrophages/cytology , Macrophages/physiology , Macrophages/radiation effects , Mannose Receptor , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/metabolism , Molecular Mimicry , Phenotype , Phosphopeptides/pharmacology , Phosphorylation/drug effects , Protein Kinase C/genetics , RNA, Small Interfering/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , STAT6 Transcription Factor/metabolism , THP-1 Cells , Tumor Microenvironment , src Homology Domains/drug effects
3.
PLoS One ; 11(4): e0153933, 2016.
Article in English | MEDLINE | ID: mdl-27100181

ABSTRACT

Mutational processes and signatures that drive early tumorigenesis are centrally important for early cancer prevention. Yet, to date, biomarkers and risk factors for polyps (adenomas) that inordinately and rapidly develop into colon cancer remain poorly defined. Here, we describe surprisingly high mutational profiles through whole-genome sequence (WGS) analysis in 2 of 4 pairs of benign colorectal adenoma tissue samples. Unsupervised hierarchical clustered transcriptomic analysis of a further 7 pairs of adenomas reveals distinct mutational signatures regardless of adenoma size. Transitional single nucleotide substitutions of C:G>T:A predominate in the adenoma mutational spectrum. Strikingly, we observe mutations in the TGF-ß pathway and CEA-associated genes in 4 out of 11 adenomas, overlapping with the Wnt pathway. Immunohistochemical labeling reveals a nearly 5-fold increase in CEA levels in 23% of adenoma samples with a concomitant loss of TGF-ß signaling. We also define a functional role by which the CEA B3 domain interacts with TGFBR1, potentially inactivating the tumor suppressor function of TGF-ß signaling. Our study uncovers diverse mutational processes underlying the transition from early adenoma to cancer. This has broad implications for biomarker-driven targeting of CEA/TGF-ß in high-risk adenomas and may lead to early detection of aggressive adenoma to CRC progression.


Subject(s)
Adenoma/genetics , Carcinoembryonic Antigen/genetics , Colon/metabolism , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mutation/genetics , Transforming Growth Factor beta/genetics , Adenoma/metabolism , Adenoma/pathology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoembryonic Antigen/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Colon/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Progression , High-Throughput Nucleotide Sequencing/methods , Humans , Immunoenzyme Techniques , Immunoprecipitation , Signal Transduction , Transforming Growth Factor beta/metabolism
4.
Oncotarget ; 7(3): 3461-76, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26658105

ABSTRACT

The resistance of apoptosis in cancer cells is pivotal for their survival and is typically ruled by mutations or dysregulation of core apoptotic cascade. Mantle cell lymphoma (MCL) is a non-Hodgkin's B-cell malignancy expressing higher anti-apoptotic proteins providing survival advantage. B-PAC-1, a procaspase activating compound, induces apoptosis by sequestering Zn bound to procaspase-3, but the amino acids holding Zn in Caspase-3 is not known. Here we show that reintroduction of WT caspase-3 or 7 in Caspase3-7 double knock-out (DKO) mouse embryonic fibroblasts (MEF) promoted B-PAC-1 to induce apoptosis (27-43%), but not in DKO MEFs or MEFs expressing respective Casp3-7 catalytic mutants (12-13%). Using caspase-6 and -9 exosite analysis, we identified and mutated predicted Zn-ligands in caspase-3 (H108A, C148S and E272A) and overexpressed into DKO MEFs. Mutants carrying E272A abrogated Zn-reversal of apoptosis induced by B-PAC-1 via higher XIAP and smac expressions but not in H108A or C148S mutants. Co-immunoprecipitation analysis revealed stronger XIAP-caspase-3 interaction suggesting a novel mechanism of impulsive apoptosis resistance by disrupting predicted Zn-ligands in caspase-3. B-PAC-1 sponsored apoptosis in MCL cell lines (30-73%) via caspase-3 and PARP cleavages accompanied by loss of Mcl-1 and IAPs including XIAP while Zn substantially abrogated B-PAC-1-driven apoptosis (18-36%). In contrary, Zn is dispensable to inhibit staurosporin, bendamustine, ABT199 or MK206-induced apoptosis. Consistent to cell lines, B-PAC-1 stimulated cell death in primary B-lymphoma cells via caspase-3 cleavage with decline in both Mcl-1 and XIAP. This study underscores the first genetic evidence that B-PAC-1 driven apoptosis is mediated via Zn chelation.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Chelating Agents/metabolism , Hydrazones/pharmacology , Lymphoma, B-Cell/pathology , Lymphoma, Mantle-Cell/pathology , Piperazines/pharmacology , Zinc/metabolism , Adult , Aged , Animals , Blotting, Western , Caspases/chemistry , Caspases/genetics , Cell Proliferation/drug effects , Cells, Cultured , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Flow Cytometry , Fluorescent Antibody Technique , Humans , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/metabolism , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/metabolism , Male , Mice , Middle Aged , Neoplasm Staging , Prognosis , Protein Conformation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
5.
PLoS One ; 10(11): e0142212, 2015.
Article in English | MEDLINE | ID: mdl-26605551

ABSTRACT

Asthma is one of the most common of medical illnesses and is treated in part by drugs that activate the beta-2-adrenoceptor (ß2-AR) to dilate obstructed airways. Such drugs include long acting beta agonists (LABAs) that are paradoxically linked to excess asthma-related mortality. Here we show that LABAs such as salmeterol and structurally related ß2-AR drugs such as formoterol and carvedilol, but not short-acting agonists (SABAs) such as albuterol, promote exaggerated asthma-like allergic airway disease and enhanced airway constriction in mice. We demonstrate that salmeterol aberrantly promotes activation of the allergic disease-related transcription factor signal transducer and activator of transcription 6 (STAT6) in multiple mouse and human cells. A novel inhibitor of STAT6, PM-242H, inhibited initiation of allergic disease induced by airway fungal challenge, reversed established allergic airway disease in mice, and blocked salmeterol-dependent enhanced allergic airway disease. Thus, structurally related ß2-AR ligands aberrantly activate STAT6 and promote allergic airway disease. This untoward pharmacological property likely explains adverse outcomes observed with LABAs, which may be overcome by agents that antagonize STAT6.


Subject(s)
Adrenergic beta-2 Receptor Agonists/adverse effects , Anti-Asthmatic Agents/adverse effects , Aspergillosis, Allergic Bronchopulmonary/drug therapy , Asthma/chemically induced , Peptidomimetics/pharmacology , STAT6 Transcription Factor/antagonists & inhibitors , Albuterol/therapeutic use , Animals , Arrestins/deficiency , Arrestins/genetics , Aspergillosis, Allergic Bronchopulmonary/genetics , Aspergillosis, Allergic Bronchopulmonary/metabolism , Aspergillosis, Allergic Bronchopulmonary/pathology , Aspergillus niger/physiology , Asthma/drug therapy , Asthma/genetics , Asthma/metabolism , Bronchoconstriction/drug effects , Carbazoles/adverse effects , Carvedilol , Disease Models, Animal , Female , Formoterol Fumarate/adverse effects , Gene Expression , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , Propanolamines/adverse effects , Receptors, Adrenergic, beta-2/deficiency , Receptors, Adrenergic, beta-2/genetics , STAT6 Transcription Factor/agonists , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Salmeterol Xinafoate/adverse effects , beta-Arrestins
6.
J Med Chem ; 58(22): 8970-84, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26506089

ABSTRACT

Signal transducer and activator of transcription 6 (STAT6) transmits signals from cytokines IL-4 and IL-13 and is activated in allergic airway disease. We are developing phosphopeptide mimetics targeting the SH2 domain of STAT6 to block recruitment to phosphotyrosine residues on IL-4 or IL-13 receptors and subsequent Tyr641 phosphorylation to inhibit the expression of genes contributing to asthma. Structure-affinity relationship studies showed that phosphopeptides based on Tyr631 from IL-4Rα bind with weak affinity to STAT6, whereas replacing the pY+3 residue with simple aryl and alkyl amides resulted in affinities in the mid to low nM range. A set of phosphatase-stable, cell-permeable prodrug analogues inhibited cytokine-stimulated STAT6 phosphorylation in both Beas-2B human airway cells and primary mouse T-lymphocytes at concentrations as low as 100 nM. IL-13-stimulated expression of CCL26 (eotaxin-3) was inhibited in a dose-dependent manner, demonstrating that targeting the SH2 domain blocks both phosphorylation and transcriptional activity of STAT6.


Subject(s)
Phosphopeptides/pharmacology , STAT6 Transcription Factor/drug effects , src Homology Domains/drug effects , Animals , Asthma/genetics , CD4-Positive T-Lymphocytes/drug effects , Cell Line , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Interleukin-13/biosynthesis , Interleukin-4/biosynthesis , Mice , Mice, Inbred C57BL , Models, Molecular , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Prodrugs , Rats , Receptors, Interleukin-3/drug effects , Receptors, Interleukin-4/drug effects , Structure-Activity Relationship , Transcriptional Activation/drug effects , Tyrosine/chemistry , Tyrosine/metabolism
7.
Nat Commun ; 6: 7997, 2015 Aug 17.
Article in English | MEDLINE | ID: mdl-26278622

ABSTRACT

Apart from T helper (Th)-2 cells, T follicular helper (Tfh) cells are a major class of IL-4-producing T cells, required for regulation of type 2 humoral immunity; however, transcriptional control of IL-4 production in Tfh cells remains mainly unknown. Here, we show that the basic leucine zipper transcription factor ATF-like, Batf is important for IL-4 expression in Tfh cells rather than in canonical Th2 cells. Functionally, Batf in cooperation with interferon regulatory factor (IRF) 4 along with Stat3 and Stat6 trigger IL-4 production in Tfh cells by directly binding to and activation of the CNS2 region in the IL-4 locus. In addition, Batf-to-c-Maf signalling is an important determinant of IL-4 expression in Tfh cells. Batf deficiency impairs the generation of IL-4-producing Tfh cells that results in protection against allergic asthma. Our results thus indicate a positive role of Batf in promoting the generation of pro-allergic IL-4-producing Tfh cells.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation/physiology , Interleukin-4/metabolism , T-Lymphocytes, Helper-Inducer/physiology , Adoptive Transfer , Animals , Asthma/immunology , Basic-Leucine Zipper Transcription Factors/genetics , Bone Marrow Cells , Cell Differentiation , Chromatin Immunoprecipitation , Interleukin-4/genetics , Male , Mice , Mice, Knockout , Ovalbumin/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism
8.
Int J Pept Res Ther ; 20(1): 1-12, 2014 Mar.
Article in English | MEDLINE | ID: mdl-25530741

ABSTRACT

Identifying novel allosteric inhibitors of G protein-coupled receptor kinases (GRKs) would be of considerable use in limiting both the extent of desensitization of GPCRs as well as downstream positive regulation through GRKs. Several peptides have previously been identified as inhibitors of specific GRKs, but to date there have been few comparisons of the selectivities of these materials on the seven GRKs, modifications to allow cell penetration, or off-target activities. The goal of this study was to determine if a panel of peptides mimicking domains on either GPCRs or GRKs would exhibit selective inhibition of GRKs 2, 5, 6 and 7 phosphorylation of rhodopsin. Peptides included sequences from GRK5; helices 3, 9, and 10 (α3, α9, and α10) in the RH domain, and the N-terminal peptide (N-Ter), as well as the intracellular loop 1 (iL1) of the ß2-adrenergic receptor (ß2AR), and the Gα transducin C-tail (TCT). While some selectivity for individual GRKs was found, overall selectivity was limited and often not reflective of structural predictions. Off-target effects were probed by determining peptide inhibition of adenylyl cyclase (AC) and PKA, and while peptides had no effect on AC activity, N-Ter, iL1, and α10 were potent inhibitors of PKA. To probe inhibition of GRK activity in intact cells, we synthesized TAT-tagged peptides, and found that TAT-α9-R169A and TAT-TCT inhibited isoproterenol-stimulated GRK phosphorylation of the ß2AR; however, the TAT peptides also inhibited isoproterenol and forskolin stimulation of AC activity. Our findings demonstrate potent peptide inhibition of GRK activities in vitro, highlight the differences in the environments of biochemical and cell-based assays, and illustrate the care that must be exercised in interpreting results of either assay alone.

9.
Nat Commun ; 5: 5798, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25503582

ABSTRACT

The transcriptional regulator STAT3 curbs pro-inflammatory cytokine production mediated by NF-κB signalling in innate immune cells, yet the mechanism by which this occurs has been unclear. Here we identify STAT3 as a pivotal negative regulator of Ubc13, an E2 ubiquitin-conjugating enzyme that facilitates TRAF6 K63-linked ubiquitination and NF-κB activation. Ubc13 accumulates intracellularly in the absence of STAT3. Depletion of Ubc13 in Stat3-deficient macrophages subdues excessive RANKL- or LPS-dependent gene expression, indicating that Ubc13 overexpression mediates enhanced transcriptional responses in the absence of STAT3. In RANKL-activated macrophages, STAT3 is stimulated by autocrine IL-6 and inhibits accrual of Ets-1, Set1 methyltransferase and trimethylation of histone H3 lysine 4 (H3K4me3) at the Ube2n (Ubc13) promoter. These results delineate a mechanism by which STAT3 operates as a transcriptional repressor on Ube2n, thus modulating NF-κB activity by regulation of Ubc13 abundance. Our data suggest that this pathway plays important roles in bone homeostasis and restraint of inflammation.


Subject(s)
Fibroblasts/metabolism , Macrophages/metabolism , RANK Ligand/pharmacology , STAT3 Transcription Factor/genetics , Toll-Like Receptor 4/genetics , Ubiquitin-Conjugating Enzymes/genetics , Animals , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Regulation , HEK293 Cells , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Male , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Primary Cell Culture , Proto-Oncogene Protein c-ets-1 , RANK Ligand/genetics , RANK Ligand/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Toll-Like Receptor 4/metabolism , Transcription, Genetic , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination
10.
Future Med Chem ; 6(17): 1909-26, 2014.
Article in English | MEDLINE | ID: mdl-25495984

ABSTRACT

Breast cancer is among the most commonly diagnosed cancer types in women worldwide and is the second leading cause of cancer-related disease in the USA. SH2 domains recruit signaling proteins to phosphotyrosine residues on aberrantly activated growth factor and cytokine receptors and contribute to cancer cell cycling, metastasis, angiogenesis and so on. Herein we review phosphopeptide mimetic and small-molecule approaches targeting the SH2 domains of Grb2, Grb7 and STAT3 that inhibit their targets and reduce proliferation in in vitro breast cancer models. Only STAT3 inhibitors have been evaluated in in vivo models and have led to tumor reduction. Taken together, these studies suggest that targeting SH2 domains is an important approach to the treatment of breast cancer.


Subject(s)
GRB2 Adaptor Protein/antagonists & inhibitors , GRB7 Adaptor Protein/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , src Homology Domains , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , GRB2 Adaptor Protein/metabolism , GRB7 Adaptor Protein/metabolism , Humans , Phosphopeptides/chemistry , Phosphopeptides/metabolism , Phosphopeptides/therapeutic use , Protein Binding , STAT3 Transcription Factor/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/therapeutic use
11.
J Org Chem ; 79(17): 8422-7, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25116734

ABSTRACT

We report a very mild synthesis of N-protected α-methylamines from the corresponding amino acids. Carboxyl groups of amino acids are reduced to iodomethyl groups via hydroxymethyl intermediates. Reductive deiodination to methyl groups is achieved by hydrogenation or catalytic transfer hydrogenation under alkaline conditions. Basic hydrodehalogenation is selective for the iodomethyl group over hydrogenolysis-labile protecting groups, such as benzyloxycarbonyl, benzyl ester, benzyl ether, and 9-fluorenyloxymethyl, thus allowing the conversion of virtually any protected amino acid into the corresponding N-protected α-methylamine.


Subject(s)
Alkalies/chemistry , Iodides/chemistry , Methylamines/chemistry , Methylamines/chemical synthesis , Catalysis , Hydrogenation , Magnetic Resonance Spectroscopy
12.
ACS Med Chem Lett ; 5(1): 69-72, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24900775

ABSTRACT

An improved synthesis of a phosphopeptidomimetic prodrug targeting the Src Homology 2 (SH2) domain of signal transducer and activator of transcription 6 (STAT6) is reported. In our convergent methodology, we employed a phosphotyrosine surrogate active ester harboring pivaloyloxymethyl groups, which efficiently coupled to tert-butylglycinyl proline diarylamide. Biological evaluation of 1 has not been reported. We show that it inhibits STAT6 phosphorylation in intact human bronchial epithelial cells, suggesting potential application in the treatment of asthma.

13.
PLoS Genet ; 10(5): e1004336, 2014 May.
Article in English | MEDLINE | ID: mdl-24784729

ABSTRACT

Secondary metabolites are produced by numerous organisms and can either be beneficial, benign, or harmful to humans. Genes involved in the synthesis and transport of these secondary metabolites are frequently found in gene clusters, which are often coordinately regulated, being almost exclusively dependent on transcription factors that are located within the clusters themselves. Gliotoxin, which is produced by a variety of Aspergillus species, Trichoderma species, and Penicillium species, exhibits immunosuppressive properties and has therefore been the subject of research for many laboratories. There have been a few proteins shown to regulate the gliotoxin cluster, most notably GliZ, a Zn2Cys6 binuclear finger transcription factor that lies within the cluster, and LaeA, a putative methyltransferase that globally regulates secondary metabolism clusters within numerous fungal species. Using a high-copy inducer screen in A. fumigatus, our lab has identified a novel C2H2 transcription factor, which plays an important role in regulating the gliotoxin biosynthetic cluster. This transcription factor, named GipA, induces gliotoxin production when present in extra copies. Furthermore, loss of gipA reduces gliotoxin production significantly. Through protein binding microarray and mutagenesis, we have identified a DNA binding site recognized by GipA that is in extremely close proximity to a potential GliZ DNA binding site in the 5' untranslated region of gliA, which encodes an efflux pump within the gliotoxin cluster. Not surprisingly, GliZ and GipA appear to work in an interdependent fashion to positively control gliA expression.


Subject(s)
Aspergillus fumigatus/metabolism , Fungal Proteins/metabolism , Transcription Factors/physiology , 5' Untranslated Regions , Binding Sites , Fungal Proteins/genetics , Gene Dosage , Promoter Regions, Genetic
14.
PLoS One ; 8(8): e71508, 2013.
Article in English | MEDLINE | ID: mdl-24013655

ABSTRACT

WNT signaling plays a key role in the self-renewal of tumor initiation cells (TICs). In this study, we used pyrvinium pamoate (PP), an FDA-approved antihelmintic drug that inhibits WNT signaling, to test whether pharmacologic inhibition of WNT signaling can specifically target TICs of aggressive breast cancer cells. SUM-149, an inflammatory breast cancer cell line, and SUM-159, a metaplastic basal-type breast cancer cell line, were used in these studies. We found that PP inhibited primary and secondary mammosphere formation of cancer cells at nanomolar concentrations, at least 10 times less than the dose needed to have a toxic effect on cancer cells. A comparable mammosphere formation IC50 dose to that observed in cancer cell lines was obtained using malignant pleural effusion samples from patients with IBC. A decrease in activity of the TIC surrogate aldehyde dehydrogenase was observed in PP-treated cells, and inhibition of WNT signaling by PP was associated with down-regulation of a panel of markers associated with epithelial-mesenchymal transition. In vivo, intratumoral injection was associated with tumor necrosis, and intraperitoneal injection into mice with tumor xenografts caused significant tumor growth delay and a trend toward decreased lung metastasis. In in vitro mammosphere-based and monolayer-based clonogenic assays, we found that PP radiosensitized cells in monolayer culture but not mammosphere culture. These findings suggest WNT signaling inhibition may be a feasible strategy for targeting aggressive breast cancer. Investigation and modification of the bioavailability and toxicity profile of systemic PP are warranted.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Pyrvinium Compounds/pharmacology , Radiation-Sensitizing Agents/pharmacology , Animals , Anthelmintics/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation/drug effects , Epithelial-Mesenchymal Transition , Female , Humans , Inhibitory Concentration 50 , Lung Neoplasms/secondary , Mice , Mice, Nude , Mice, SCID , Neoplastic Stem Cells/drug effects , Radiation Tolerance/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/physiology , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , beta Catenin/metabolism
15.
J Exp Med ; 210(2): 321-37, 2013 Feb 11.
Article in English | MEDLINE | ID: mdl-23359069

ABSTRACT

Although aberrant Notch activation contributes to leukemogenesis in T cells, its role in acute myelogenous leukemia (AML) remains unclear. Here, we report that human AML samples have robust expression of Notch receptors; however, Notch receptor activation and expression of downstream Notch targets are remarkably low, suggesting that Notch is present but not constitutively activated in human AML. The functional role of these Notch receptors in AML is not known. Induced activation through any of the Notch receptors (Notch1-4), or through the Notch target Hairy/Enhancer of Split 1 (HES1), consistently leads to AML growth arrest and caspase-dependent apoptosis, which are associated with B cell lymphoma 2 (BCL2) loss and enhanced p53/p21 expression. These effects were dependent on the HES1 repressor domain and were rescued through reexpression of BCL2. Importantly, activated Notch1, Notch2, and HES1 all led to inhibited AML growth in vivo, and Notch inhibition via dnMAML enhanced proliferation in vivo, thus revealing the physiological inhibition of AML growth in vivo in response to Notch signaling. As a novel therapeutic approach, we used a Notch agonist peptide that led to significant apoptosis in AML patient samples. In conclusion, we report consistent Notch-mediated growth arrest and apoptosis in human AML, and propose the development of Notch agonists as a potential therapeutic approach in AML.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Receptors, Notch/metabolism , Adolescent , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Child , DNA-Binding Proteins/genetics , Gene Expression , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Infant , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mutation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptors, Notch/agonists , Receptors, Notch/genetics , Signal Transduction , Transcription Factor HES-1 , Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism
16.
BMC Struct Biol ; 13 Suppl 1: S11, 2013.
Article in English | MEDLINE | ID: mdl-24564952

ABSTRACT

BACKGROUND: Using the popular program AutoDock, computer-aided docking of small ligands with 6 or fewer rotatable bonds, is reasonably fast and accurate. However, docking large ligands using AutoDock's recommended standard docking protocol is less accurate and computationally slow. RESULTS: In our earlier work, we presented a novel AutoDock-based incremental protocol (DINC) that addresses the limitations of AutoDock's standard protocol by enabling improved docking of large ligands. Instead of docking a large ligand to a target protein in one single step as done in the standard protocol, our protocol docks the large ligand in increments. In this paper, we present three detailed examples of docking using DINC and compare the docking results with those obtained using AutoDock's standard protocol. We summarize the docking results from an extended docking study that was done on 73 protein-ligand complexes comprised of large ligands. We demonstrate not only that DINC is up to 2 orders of magnitude faster than AutoDock's standard protocol, but that it also achieves the speed-up without sacrificing docking accuracy. We also show that positional restraints can be applied to the large ligand using DINC: this is useful when computing a docked conformation of the ligand. Finally, we introduce a webserver for docking large ligands using DINC. CONCLUSIONS: Docking large ligands using DINC is significantly faster than AutoDock's standard protocol without any loss of accuracy. Therefore, DINC could be used as an alternative protocol for docking large ligands. DINC has been implemented as a webserver and is available at http://dinc.kavrakilab.org. Applications such as therapeutic drug design, rational vaccine design, and others involving large ligands could benefit from DINC and its webserver implementation.


Subject(s)
Ligands , Proteins/metabolism , Algorithms , Molecular Conformation , Molecular Docking Simulation , Proteins/chemistry , Software , User-Computer Interface
17.
Int J Pept Res Ther ; 19(1): 3-12, 2013 Mar.
Article in English | MEDLINE | ID: mdl-24707243

ABSTRACT

Signal transducer and activator of transcription 3 (Stat3) transmits signals from growth factors and interleukin-6 family cytokines by binding to their receptors via its Src homology 2 (SH2) domain. This results in phosphorylation of Tyr705, dimerization, translocation to the nucleus, and regulation of transcription of downstream genes. Stat3 is constitutively activated in several human cancers and is a target for anti-cancer drug design. We have shown previously phosphorylation of Tyr705 in intact cancer cells can be inhibited with prodrugs of phosphopeptide mimics targeting the SH2 domain. In a series of prodrugs consisting of bis-pivaloyloxymethyl esters of 4'-phosphonodifluoromethyl cinnamoyl-Haic-Gln-NHBn, appending methyl group to the ß-position of the cinnamate increased potency ca. twofold, which paralleled the increase in affinity of the corresponding phosphopeptide models. However, dramatic increases in potency were observed when the C-terminal C(O)NHBn of Gln-NHBn was replaced with a simple methyl group. In this communication we continue to explore the effects of structural modifications of prodrugs on their ability to inhibit Tyr705 phosphorylation. A set of 4-substituted prolines incorporated into ß-methyl-4-phosphocinnamoyl-leucinyl-Xaa-4-aminopentamide model peptides exhibited affinities of 88-317 nM by fluorescence polarization (Pro IC50 = 156 nM). In corresponding prodrugs, Pro inhibited constitutive Stat3 phosphorylation at 10 µM in MDA-MB-468 breast tumor cells. However, 4,4-difluoroproline and 4,4-dimethylproline resulted in complete inhibition at 0.5 µM. These results suggest that the prodrug with native proline undergoes metabolism that those with substituted prolines do not. In conclusion, changes in structure with minimal impact on intrinsic affinity can nevertheless have profound effects on the cellular potency of prodrug inhibitors of Stat3.

18.
PLoS One ; 7(12): e51603, 2012.
Article in English | MEDLINE | ID: mdl-23251591

ABSTRACT

STAT3 is a transcription factor that has been found to be constitutively activated in a number of human cancers. Dimerization of STAT3 via its SH2 domain and the subsequent translocation of the dimer to the nucleus leads to transcription of anti-apoptotic genes. Prevention of the dimerization is thus an attractive strategy for inhibiting the activity of STAT3. Phosphotyrosine-based peptidomimetic inhibitors, which mimic pTyr-Xaa-Yaa-Gln motif and have strong to weak binding affinities, have been previously investigated. It is well-known that structures of protein-inhibitor complexes are important for understanding the binding interactions and designing stronger inhibitors. Experimental structures of inhibitors bound to the SH2 domain of STAT3 are, however, unavailable. In this paper we describe a computational study that combined molecular docking and molecular dynamics to model structures of 12 peptidomimetic inhibitors bound to the SH2 domain of STAT3. A detailed analysis of the modeled structures was performed to evaluate the characteristics of the binding interactions. We also estimated the binding affinities of the inhibitors by combining MMPB/GBSA-based energies and entropic cost of binding. The estimated affinities correlate strongly with the experimentally obtained affinities. Modeling results show binding modes that are consistent with limited previous modeling studies on binding interactions involving the SH2 domain and phosphotyrosine(pTyr)-based inhibitors. We also discovered a stable novel binding mode that involves deformation of two loops of the SH2 domain that subsequently bury the C-terminal end of one of the stronger inhibitors. The novel binding mode could prove useful for developing more potent inhibitors aimed at preventing dimerization of cancer target protein STAT3.


Subject(s)
Drug Design , Peptidomimetics/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Cluster Analysis , Humans , Hydrogen Bonding/drug effects , Models, Molecular , Peptidomimetics/chemistry , Peptidomimetics/metabolism , Protein Binding/drug effects , Protein Conformation , STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/metabolism
19.
J Exp Ther Oncol ; 10(2): 155-62, 2012.
Article in English | MEDLINE | ID: mdl-23350355

ABSTRACT

Signal transducer and activator of transcription 3 (Stat3) is constitutively activated in a number of human cancers and cancer cell lines. Via its Src homology 2 (SH2) domain, Stat3 is recruited to phosphotyrosine residues on intracellular domains of cytokine and growth factor receptors, whereupon it is phosphorylated on Tyr705, dimerizes, translocates to the nucleus and is reported to participate in the expression of genes related to angiogenesis, metastasis, growth and survival. To block this process, we are developing cell-permeable, phosphatase-stable phosphopeptide mimics, targeted to the SH2 domain of Stat3, that inhibit the phosphorylation of Tyr705 of Stat3 in cultured tumor cells (Mandal et al., J. Med. Chem. 54, 3549-5463, 2011). At concentrations that inhibit tyrosine phosphorylation, these materials were not cytotoxic, similar to recent reports on JAK inhibitors. At higher concentrations, cytotoxicity was accompanied by off-target effects. We report that treatment of MDA-MB-468 human breast cancer xenografts in mice with peptidomimetic PM-73G significantly inhibited tumor growth, which was accompanied by reduction in VEGF production and microvessel density. No evidence of apoptosis or changes in the expression of the canonical genes cyclin D1 or survivin were observed. Thus selective inhibition of Stat3 Tyr705 phosphorylation may be a novel anti-angiogenesis strategy for the treatment of cancer.


Subject(s)
Biomimetics , Breast Neoplasms/prevention & control , Neovascularization, Pathologic/prevention & control , Phosphopeptides/pharmacology , Prodrugs/pharmacology , STAT3 Transcription Factor/metabolism , src Homology Domains/drug effects , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Female , Humans , Mice , Mice, Nude , Phosphorylation/drug effects , Phosphotyrosine/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
JAKSTAT ; 1(4): 263-347, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-24058783

ABSTRACT

Herein we review our progress on the development of phosphopeptide-based prodrugs targeting the SH2 domain of STAT3 to prevent recruitment to cytokine and growth factor receptors, activation, nuclear translocation and transcription of genes involved in cancer. We developed high affinity phosphopeptides (K I = 46-200 nM). Corresponding prodrugs inhibited constitutive and IL-6 induced Tyr705 phosphorylation at 0.5-1 µM in a variety of human cancer cell lines. They were not cytotoxic at 5 µM in vitro but they inhibited tumor growth in a human xenograft breast cancer model in mice, accompanied by reduced VEGF expression and angiogenesis.

SELECTION OF CITATIONS
SEARCH DETAIL
...