Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters











Publication year range
1.
ACS Chem Biol ; 16(3): 548-556, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33621466

ABSTRACT

We report a general approach to engineering multivalent d-proteins with antibody-like activities in vivo. Mirror-image phage display and structure-guided design were utilized to create a d-protein that uses receptor mimicry to antagonize vascular endothelial growth factor A (VEGF-A). Selections against the d-protein form of VEGF-A using phage-displayed libraries of two different domain scaffolds yielded two proteins that bound distinct receptor interaction sites on VEGF-A. X-ray crystal structures of the d-protein/VEGF-A complexes were used to guide affinity maturation and to construct a heterodimeric d-protein VEGF-A antagonist with picomolar activity. The d-protein VEGF-A antagonist prevented vascular leakage in a rabbit eye model of wet age-related macular degeneration and slowed tumor growth in the MC38 syngeneic mouse tumor model with efficacies comparable to those of approved antibody drugs, and in contrast with antibodies, the d-protein was non-immunogenic during treatment and following subcutaneous immunizations.


Subject(s)
Antineoplastic Agents/chemistry , Neoplasms/drug therapy , Peptides/chemistry , Receptors, Vascular Endothelial Growth Factor/chemistry , Retinal Vessels/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Amino Acid Sequence , Animals , Antineoplastic Agents/pharmacology , Bevacizumab/pharmacology , Binding Sites , Drug Evaluation, Preclinical , Eye/drug effects , Female , Humans , Mice , Models, Molecular , Peptide Library , Peptides/pharmacology , Protein Binding , Protein Conformation , Protein Multimerization , Rabbits , Receptors, Vascular Endothelial Growth Factor/metabolism
2.
Mol Cell ; 69(4): 551-565.e7, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29452636

ABSTRACT

Inflammatory responses mediated by NOD2 rely on RIP2 kinase and ubiquitin ligase XIAP for the activation of nuclear factor κB (NF-κB), mitogen-activated protein kinases (MAPKs), and cytokine production. Herein, we demonstrate that selective XIAP antagonism blocks NOD2-mediated inflammatory signaling and cytokine production by interfering with XIAP-RIP2 binding, which removes XIAP from its ubiquitination substrate RIP2. We also establish that the kinase activity of RIP2 is dispensable for NOD2 signaling. Rather, the conformation of the RIP2 kinase domain functions to regulate binding to the XIAP-BIR2 domain. Effective RIP2 kinase inhibitors block NOD2 signaling by disrupting RIP2-XIAP interaction. Finally, we identify NOD2 signaling and XIAP-dependent ubiquitination sites on RIP2 and show that mutating these lysine residues adversely affects NOD2 pathway signaling. Overall, these results reveal a critical role for the XIAP-RIP2 interaction in NOD2 inflammatory signaling and provide a molecular basis for the design of innovative therapeutic strategies based on XIAP antagonists and RIP2 kinase inhibitors.


Subject(s)
Aminoquinolines/pharmacology , Inflammation/prevention & control , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Protein Interaction Domains and Motifs/drug effects , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Sulfones/pharmacology , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , Cells, Cultured , Humans , Inflammation/metabolism , Inflammation/pathology , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinase 2/antagonists & inhibitors , Signal Transduction , Ubiquitin/metabolism , Ubiquitination , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors
3.
Oncotarget ; 7(31): 49539-49551, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27385100

ABSTRACT

Apoptosis is deregulated in most, if not all, cancers, including hematological malignancies. Smac mimetics that antagonize Inhibitor of Apoptosis (IAP) proteins have so far largely been investigated in acute myeloid leukemia (AML) cell lines; however, little is yet known on the therapeutic potential of Smac mimetics in primary AML samples. In this study, we therefore investigated the antileukemic activity of the Smac mimetic BV6 in diagnostic samples of 67 adult AML patients and correlated the response to clinical, cytogenetic and molecular markers and gene expression profiles. Treatment with cytarabine (ara-C) was used as a standard chemotherapeutic agent. Interestingly, about half (51%) of primary AML samples are sensitive to BV6 and 21% intermediate responsive, while 28% are resistant. Notably, 69% of ara-C-resistant samples show a good to fair response to BV6. Furthermore, combination treatment with ara-C and BV6 exerts additive effects in most samples. Whole-genome gene expression profiling identifies cell death, TNFR1 and NF-κB signaling among the top pathways that are activated by BV6 in BV6-sensitive, but not in BV6-resistant cases. Furthermore, sensitivity of primary AML blasts to BV6 correlates with significantly elevated expression levels of TNF and lower levels of XIAP in diagnostic samples, as well as with NPM1 mutation. In a large set of primary AML samples, these data provide novel insights into factors regulating Smac mimetic response in AML and have important implications for the development of Smac mimetic-based therapies and related diagnostics in AML.


Subject(s)
Cell Death/drug effects , Gene Expression Regulation, Leukemic , Intracellular Signaling Peptides and Proteins/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Mitochondrial Proteins/pharmacology , Adult , Aged , Aged, 80 and over , Apoptosis , Apoptosis Regulatory Proteins , Cell Line, Tumor , Cell Survival , Cytarabine/pharmacology , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Mutation , NF-kappa B/metabolism , Nucleophosmin , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Treatment Outcome , Young Adult
4.
J Mol Biol ; 428(3): 603-617, 2016 Feb 13.
Article in English | MEDLINE | ID: mdl-26778617

ABSTRACT

Context-independent anti-hypusine antibodies that bind to the post-translational modification (PTM), hypusine, with minimal dependence on flanking amino acid sequences, were identified. The antibodies bind to both hypusine and deoxyhypusine or selectively to hypusine but not to deoxyhypusine. Phage display was used to further enhance the affinity of the antibodies. Affinity maturation of these anti-hypusine antibodies improved their performance in affinity capture of the only currently known hypusinated protein, eukaryotic translation initiation factor 5A. These anti-hypusine antibodies may have utility in the identification of novel hypusinated proteins. Crystal structures of the corresponding Fab fragments were determined in complex with hypusine- or deoxyhypusine-containing peptides. The hypusine or deoxyhypusine moiety was found to reside in a deep pocket formed between VH and VL domains of the Fab fragments. Interaction between the antibodies and hypusine includes an extensive hydrogen bond network. These are, to our knowledge, the first reported structures of context-independent anti-PTM antibodies in complex with the corresponding PTM.


Subject(s)
Antibodies, Monoclonal/immunology , Lysine/analogs & derivatives , Peptide Initiation Factors/chemistry , RNA-Binding Proteins/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibody Affinity , Crystallography, X-Ray , Humans , Lysine/analysis , Lysine/immunology , Models, Molecular , Molecular Sequence Data , Peptide Initiation Factors/immunology , Protein Processing, Post-Translational , RNA-Binding Proteins/immunology , Rabbits , Eukaryotic Translation Initiation Factor 5A
5.
J Org Chem ; 80(7): 3677-81, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25769022

ABSTRACT

An orthogonally protected hypusine reagent was developed for solid-phase synthesis of hypusinated peptides using the Fmoc/t-Bu protection strategy. The reagent was synthesized in an overall yield of 27% after seven steps from Cbz-Lys-OBzl and (R)-3-hydroxypyrrolidin-2-one. The side-chain protecting groups (Boc and t-Bu) are fully compatible with standard Fmoc chemistry and can be readily removed during the peptide cleavage step. The utility of the reagent was demonstrated by solid-phase synthesis of hypusinated peptides.


Subject(s)
Amino Acids/chemical synthesis , Lysine/analogs & derivatives , Peptides/chemical synthesis , Pyrrolidinones/chemistry , Amino Acid Sequence , Amino Acids/chemistry , Indicators and Reagents/chemistry , Lysine/chemical synthesis , Lysine/chemistry , Molecular Structure , Peptides/chemistry , Solid-Phase Synthesis Techniques
7.
ACS Med Chem Lett ; 5(10): 1088-93, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25313317

ABSTRACT

A-1155463, a highly potent and selective BCL-XL inhibitor, was discovered through nuclear magnetic resonance (NMR) fragment screening and structure-based design. This compound is substantially more potent against BCL-XL-dependent cell lines relative to our recently reported inhibitor, WEHI-539, while possessing none of its inherent pharmaceutical liabilities. A-1155463 caused a mechanism-based and reversible thrombocytopenia in mice and inhibited H146 small cell lung cancer xenograft tumor growth in vivo following multiple doses. A-1155463 thus represents an excellent tool molecule for studying BCL-XL biology as well as a productive lead structure for further optimization.

9.
Nat Chem Biol ; 9(6): 390-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23603658

ABSTRACT

The prosurvival BCL-2 family protein BCL-X(L) is often overexpressed in solid tumors and renders malignant tumor cells resistant to anticancer therapeutics. Enhancing apoptotic responses by inhibiting BCL-X(L) will most likely have widespread utility in cancer treatment and, instead of inhibiting multiple prosurvival BCL-2 family members, a BCL-X(L)-selective inhibitor would be expected to minimize the toxicity to normal tissues. We describe the use of a high-throughput screen to discover a new series of small molecules targeting BCL-X(L) and their structure-guided development by medicinal chemistry. The optimized compound, WEHI-539 (7), has high affinity (subnanomolar) and selectivity for BCL-X(L) and potently kills cells by selectively antagonizing its prosurvival activity. WEHI-539 will be an invaluable tool for distinguishing the roles of BCL-X(L) from those of its prosurvival relatives, both in normal cells and notably in malignant tumor cells, many of which may prove to rely upon BCL-X(L) for their sustained growth.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/chemistry , Animals , Apoptosis , Benzothiazoles/chemistry , Cell Line, Tumor , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Humans , Hydrazones/chemistry , Kinetics , Mice , Models, Chemical , Myeloid Cell Leukemia Sequence 1 Protein , Protein Binding , Proto-Oncogene Proteins c-bcl-2/genetics
10.
EMBO J ; 32(8): 1103-14, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23524849

ABSTRACT

The cellular inhibitor of apoptosis (c-IAP) proteins are E3 ubiquitin ligases that are critical regulators of tumour necrosis factor (TNF) receptor (TNFR)-mediated signalling. Through their E3 ligase activity c-IAP proteins promote ubiquitination of receptor-interaction protein 1 (RIP1), NF-κB-inducing kinase (NIK) and themselves, and regulate the assembly of TNFR signalling complexes. Consequently, in the absence of c-IAP proteins, TNFR-mediated activation of NF-κB and MAPK pathways and the induction of gene expression are severely reduced. Here, we describe the identification of OTUB1 as a c-IAP-associated deubiquitinating enzyme that regulates c-IAP1 stability. OTUB1 disassembles K48-linked polyubiquitin chains from c-IAP1 in vitro and in vivo within the TWEAK receptor-signalling complex. Downregulation of OTUB1 promotes TWEAK- and IAP antagonist-stimulated caspase activation and cell death, and enhances c-IAP1 degradation. Furthermore, knockdown of OTUB1 reduces TWEAK-induced activation of canonical NF-κB and MAPK signalling pathways and modulates TWEAK-induced gene expression. Finally, suppression of OTUB1 expression in zebrafish destabilizes c-IAP (Birc2) protein levels and disrupts fish vasculature. These results suggest that OTUB1 regulates NF-κB and MAPK signalling pathways and TNF-dependent cell death by modulating c-IAP1 stability.


Subject(s)
Cysteine Endopeptidases/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Signal Transduction , Ubiquitin/metabolism , Animals , Blood Vessels/embryology , Cell Line , Deubiquitinating Enzymes , Humans , Hydrolysis , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Zebrafish/embryology
11.
ACS Chem Biol ; 8(2): 297-302, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23151250

ABSTRACT

An attractive approach for developing therapeutic peptides is to enhance binding to their targets by stabilizing their α-helical conformation, for example, stabilized BimBH3 peptides (BimSAHB) designed to induce apoptosis. Unexpectedly, we found that such modified peptides have reduced affinity for their targets, the pro-survival Bcl-2 proteins. We attribute this loss in affinity to disruption of a network of stabilizing intramolecular interactions present in the bound state of the native peptide. Altering this network may compromise binding affinity, as in the case of the BimBH3 stapled peptide studied here. Moreover, cells exposed to these peptides do not readily undergo apoptosis, strongly indicating that BimSAHB is not inherently cell permeable.


Subject(s)
Apoptosis , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Animals , Humans , Mice , Models, Molecular , Protein Structure, Secondary
12.
Biochem J ; 447(3): 427-36, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22853455

ABSTRACT

ML-IAP [melanoma IAP (inhibitor of apoptosis)] is an anti-apoptotic protein that is expressed highly in melanomas where it contributes to resistance to apoptotic stimuli. The anti-apoptotic activity and elevated expression of IAP family proteins in many human cancers makes IAP proteins attractive targets for inhibition by cancer therapeutics. Small-molecule IAP antagonists that bind with high affinities to select BIR (baculovirus IAP repeat) domains have been shown to stimulate auto-ubiquitination and rapid proteasomal degradation of c-IAP1 (cellular IAP1) and c-IAP2 (cellular IAP2). In the present paper, we report ML-IAP proteasomal degradation in response to bivalent, but not monovalent, IAP antagonists. This degradation required ML-IAP ubiquitin ligase activity and was independent of c-IAP1 or c-IAP2. Although ML-IAP is best characterized in melanoma cells, we show that ML-IAP expression in normal mammalian tissues is restricted largely to the eye, being most abundant in ciliary body epithelium and retinal pigment epithelium. Surprisingly, given this pattern of expression, gene-targeted mice lacking ML-IAP exhibited normal intraocular pressure as well as normal retinal structure and function. The results of the present study indicate that ML-IAP is dispensable for both normal mouse development and ocular homoeostasis.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Eye/metabolism , Inhibitor of Apoptosis Proteins/physiology , Neoplasm Proteins/physiology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line, Tumor , Eye/blood supply , Female , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/genetics , Intraocular Pressure , Male , Melanoma , Mice , Mice, Mutant Strains , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Organ Specificity , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Protein Structure, Tertiary , Retina/anatomy & histology , Retina/physiology , Ubiquitin-Protein Ligases/metabolism
13.
J Med Chem ; 55(9): 4101-13, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22413863

ABSTRACT

A series of compounds were designed and synthesized as antagonists of cIAP1/2, ML-IAP, and XIAP based on the N-terminus, AVPI, of mature Smac. Compound 1 (GDC-0152) has the best profile of these compounds; it binds to the XIAP BIR3 domain, the BIR domain of ML-IAP, and the BIR3 domains of cIAP1 and cIAP2 with K(i) values of 28, 14, 17, and 43 nM, respectively. These compounds promote degradation of cIAP1, induce activation of caspase-3/7, and lead to decreased viability of breast cancer cells without affecting normal mammary epithelial cells. Compound 1 inhibits tumor growth when dosed orally in the MDA-MB-231 breast cancer xenograft model. Compound 1 was advanced to human clinical trials, and it exhibited linear pharmacokinetics over the dose range (0.049 to 1.48 mg/kg) tested. Mean plasma clearance in humans was 9 ± 3 mL/min/kg, and the volume of distribution was 0.6 ± 0.2 L/kg.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Thiadiazoles/chemical synthesis , Thiadiazoles/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Baculoviral IAP Repeat-Containing 3 Protein , Binding, Competitive , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Clinical Trials, Phase I as Topic , Female , Humans , Inhibitor of Apoptosis Proteins/metabolism , Male , Thiadiazoles/chemistry , Thiadiazoles/pharmacokinetics , Ubiquitin-Protein Ligases
14.
Sci Signal ; 5(216): ra22, 2012 Mar 20.
Article in English | MEDLINE | ID: mdl-22434933

ABSTRACT

Tumor necrosis factor (TNF) family members are essential for the development and proper functioning of the immune system. TNF receptor (TNFR) signaling is mediated through the assembly of protein signaling complexes that activate the nuclear factor κB (NF-κB) and mitogen-activated protein kinase (MAPK) pathways in a ubiquitin-dependent manner. The cellular inhibitor of apoptosis (c-IAP) proteins c-IAP1 and c-IAP2 are E3 ubiquitin ligases that are recruited to TNFR signaling complexes through their constitutive association with the adaptor protein TNFR-associated factor 2 (TRAF2). We demonstrated that c-IAP1 and c-IAP2 were required for canonical activation of NF-κB and MAPK by members of the TNFR family. c-IAPs were required for the recruitment of inhibitor of κB kinase ß (IKKß), the IKK regulatory subunit NF-κB essential modulator (NEMO), and RBCK1/Hoil1-interacting protein (HOIP) to TNFR signaling complexes and the induction of gene expression by TNF family members. In contrast, TNFRs that stimulated the noncanonical NF-κB pathway triggered translocation of c-IAPs, TRAF2, and TRAF3 from the cytosol to membrane fractions, which led to their proteasomal and lysosomal degradation. Finally, we established that signaling by B cell-activating factor receptor 3 induced the cytosolic depletion of TRAF3, which enabled noncanonical NF-κB activation. These results define c-IAP proteins as critical regulators of the activation of NF-κB and MAPK signaling pathways by members of the TNFR superfamily.


Subject(s)
Inhibitor of Apoptosis Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Signal Transduction/immunology , Tumor Necrosis Factors/metabolism , Blotting, Western , Carrier Proteins/metabolism , Cell Line, Tumor , Gene Silencing , Humans , I-kappa B Kinase/metabolism , Inhibitor of Apoptosis Proteins/immunology , Protein Transport , RNA, Small Interfering/genetics , Receptors, Interleukin-4/metabolism , TNF Receptor-Associated Factor 2/metabolism , Tumor Necrosis Factors/immunology , Ubiquitin/metabolism , Ubiquitin-Protein Ligases
15.
Science ; 334(6054): 376-80, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-22021857

ABSTRACT

Inhibitor of apoptosis (IAP) proteins are negative regulators of cell death. IAP family members contain RING domains that impart E3 ubiquitin ligase activity. Binding of endogenous or small-molecule antagonists to select baculovirus IAP repeat (BIR) domains within cellular IAP (cIAP) proteins promotes autoubiquitination and proteasomal degradation and so releases inhibition of apoptosis mediated by cIAP. Although the molecular details of antagonist-BIR domain interactions are well understood, it is not clear how this binding event influences the activity of the RING domain. Here biochemical and structural studies reveal that the unliganded, multidomain cIAP1 sequesters the RING domain within a compact, monomeric structure that prevents RING dimerization. Antagonist binding induces conformational rearrangements that enable RING dimerization and formation of the active E3 ligase.


Subject(s)
Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/chemistry , Amino Acid Sequence , Animals , Cell Line , Cell Line, Tumor , Cloning, Molecular , Humans , Hydrophobic and Hydrophilic Interactions , Inhibitor of Apoptosis Proteins/metabolism , Mice , Models, Biological , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Proteasome Endopeptidase Complex/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Secondary , Scattering, Small Angle , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitinated Proteins/chemistry , Ubiquitinated Proteins/metabolism , Ubiquitination
16.
Neoplasia ; 13(10): 971-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22028622

ABSTRACT

Searching for new strategies to bypass apoptosis resistance, we investigated the potential of the Smac mimetic BV6 in Jurkat leukemia cells deficient in key molecules of the death receptor pathway. Here, we demonstrate for the first time that Smac mimetic primes apoptosis-resistant, FADD- or caspase-8-deficient leukemia cells for TNFα-induced necroptosis in a synergistic manner. In contrast to TNFα, Smac mimetic significantly enhances CD95-induced apoptosis in wild-type but not in FADD-deficient cells. Interestingly, Smac mimetic- and TNFα-mediated cell death occurs without characteristic features of apoptosis (i.e., caspase activation, DNA fragmentation) in FADD-deficient cells. By comparison, Smac mimetic and TNFα trigger activation of caspase-8, -9, and -3 and DNA fragmentation in wild-type cells. Consistently, the caspase inhibitor zVAD.fmk fails to block Smac mimetic- and TNFα-triggered cell death in FADD- or caspase-8-deficient cells, while it confers protection in wild-type cells. By comparison, necrostatin-1, an RIP1 kinase inhibitor, abolishes Smac mimetic- and TNFα-induced cell death in FADD- or caspase-8-deficient. Thus, Smac mimetic enhances TNFα-induced cell death in leukemia cells via two distinct pathways in a context-dependent manner: it primes apoptosis-resistant cells lacking FADD or caspase-8 to TNFα-induced, RIP1-dependent and caspase-independent necroptosis, whereas it sensitizes apoptosis-proficient cells to TNFα-mediated, caspase-dependent apoptosis. These findings have important implications for the therapeutic exploitation of necroptosis as an alternative cell death program to overcome apoptosis resistance.


Subject(s)
Apoptosis/drug effects , Biomimetic Materials/pharmacology , Caspase 8/metabolism , Fas-Associated Death Domain Protein/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis Regulatory Proteins , Blotting, Western , Caspase 3/metabolism , Caspase 9/metabolism , DNA Fragmentation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Enzyme Activation/drug effects , Fas-Associated Death Domain Protein/deficiency , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , Microscopy, Fluorescence , Mitochondrial Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism
17.
Chem Biol ; 18(7): 839-45, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21802005

ABSTRACT

Limitations to the application of molecularly targeted cancer therapies are the inability to accurately match patient with effective treatment and the absence of a prompt readout of posttreatment response. Noninvasive agents that rapidly report vascular endothelial growth factor (VEGF) levels using positron emission tomography (PET) have the potential to enhance anti-angiogenesis therapies. Using phage display, two distinct classes of peptides were identified that bind to VEGF with nanomolar affinity and high selectivity. Co-crystal structures of these different peptide classes demonstrate that both bind to the receptor-binding region of VEGF. (18)F-radiolabelling of these peptides facilitated the acquisition of PET images of tumor VEGF levels in a HM7 xenograph model. The images obtained from one 59-residue probe, (18)F-Z-3B, 2 hr postinjection are comparable to those obtained with anti-VEGF antibody B20 72 hr postinjection. Furthermore, VEGF levels in growing SKOV3 tumors were followed using (18)F-Z-3B as a PET probe with VEGF levels increasing with tumor size.


Subject(s)
Neoplasms/diagnostic imaging , Peptides/chemistry , Peptides/metabolism , Positron-Emission Tomography/methods , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/metabolism , Amino Acid Sequence , Animals , Click Chemistry , Crystallography, X-Ray , Humans , Mice , Mice, Nude , Models, Molecular , Molecular Sequence Data , Neoplasms/metabolism , Peptide Library , Peptides/chemical synthesis , Protein Binding , Protein Multimerization , Vascular Endothelial Growth Factor A/chemistry
18.
Mol Cell Proteomics ; 10(5): M110.003756, 2011 May.
Article in English | MEDLINE | ID: mdl-21048196

ABSTRACT

Ubiquitinated substrates can be recruited to macromolecular complexes through interactions between their covalently bound ubiquitin (Ub) signals and Ub receptor proteins. To develop a functional understanding of the Ub system in vivo, methods are needed to determine the composition of Ub signals on individual substrates and in protein mixtures. Mass spectrometry has emerged as an important tool for characterizing the various forms of Ub. In the Ubiquitin-AQUA approach, synthetic isotopically labeled internal standard peptides are used to quantify unbranched peptides and the branched -GG signature peptides generated by trypsin digestion of Ub signals. Here we have built upon existing methods and established a comprehensive platform for the characterization of Ub signals. Digested peptides and isotopically labeled standards are analyzed either by selected reaction monitoring on a QTRAP mass spectrometer or by narrow window extracted ion chromatograms on a high resolution LTQ-Orbitrap. Additional peptides are now monitored to account for the N terminus of ubiquitin, linear polyUb chains, the peptides surrounding K33 and K48, and incomplete digestion products. Using this expanded battery of peptides, the total amount of Ub in a sample can be determined from multiple loci within the protein, minimizing possible confounding effects of complex Ub signals, digestion abnormalities, or use of mutant Ub in experiments. These methods have been useful for the characterization of in vitro, multistage ubiquitination and have now been extended to reactions catalyzed by multiple E2 enzymes. One question arising from in vitro studies is whether individual protein substrates in cells may be modified by multiple forms of polyUb. Here we have taken advantage of recently developed polyubiquitin linkage-specific antibodies recognizing K48- and K63-linked polyUb chains, coupled with these mass spectrometry methods, to further evaluate the abundance of mixed linkage Ub substrates in cultured mammalian cells. By combining these two powerful tools, we show that polyubiquitinated substrates purified from cells can be modified by mixtures of K48, K63, and K11 linkages.


Subject(s)
Mutant Proteins/chemistry , Ubiquitin/chemistry , Ubiquitinated Proteins/metabolism , Amino Acid Sequence , HEK293 Cells , Humans , Immunoprecipitation , Jurkat Cells , Leupeptins/pharmacology , Lysine/chemistry , Methionine/chemistry , Molecular Sequence Data , Oxidation-Reduction , Peptide Fragments/chemistry , Proteasome Inhibitors , Tandem Mass Spectrometry , Ubiquitinated Proteins/chemistry , Ubiquitination
19.
Neoplasia ; 13(12): 1162-70, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22241962

ABSTRACT

Defects in apoptosis contribute to treatment resistance and poor outcome of pancreatic cancer, calling for novel therapeutic strategies. Here, we provide the first evidence that nuclear factor (NF) κB is required for Smac mimetic-mediated sensitization of pancreatic carcinoma cells for gemcitabine-induced apoptosis. The Smac mimetic BV6 cooperates with gemcitabine to reduce cell viability and to induce apoptosis. In addition, BV6 significantly enhances the cytotoxicity of several anticancer drugs against pancreatic carcinoma cells, including doxorubicin, cisplatin, and 5-fluorouracil. Molecular studies reveal that BV6 stimulates NF-κB activation, which is further increased in the presence of gemcitabine. Importantly, inhibition of NF-κB by overexpression of the dominant-negative IκBα superrepressor significantly decreases BV6- and gemcitabine-induced apoptosis, demonstrating that NF-κB exerts a proapoptotic function in this model of apoptosis. In support of this notion, inhibition of tumor necrosis factor α (TNFα) by the TNFα blocking antibody Enbrel reduces BV6- and gemcitabine-induced activation of caspase 8 and 3, loss of mitochondrial membrane potential, and apoptosis. By demonstrating that BV6 and gemcitabine trigger a NF-κB-dependent, TNFα-mediated loop to activate apoptosis signaling pathways and caspase-dependent apoptotic cell death, our findings have important implications for the development of Smac mimetic-based combination protocols in the treatment of pancreatic cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Deoxycytidine/analogs & derivatives , Intracellular Signaling Peptides and Proteins/chemistry , Mitochondrial Proteins/chemistry , Molecular Mimicry , NF-kappa B/metabolism , Pancreatic Neoplasms/metabolism , Animals , Antineoplastic Agents/administration & dosage , Apoptosis Regulatory Proteins , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Humans , Mice , Tumor Necrosis Factor-alpha/metabolism , Gemcitabine
20.
J Am Chem Soc ; 132(46): 16423-31, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21038913

ABSTRACT

A water-soluble octacarboxyhemicarcerand was used as a shuttle to transport redox-active substrates across the aqueous medium and deliver them to the target protein. The results show that weak multivalent interactions and conformational flexibility can be exploited to reversibly bind complex supramolecular assemblies to biological molecules. Hydrophobic electron donors and acceptors were encapsulated within the hemicarcerand, and photoinduced electron transfer (ET) between the Zn-substituted cytochrome c (MW = 12.3 kD) and the host-guest complexes (MW = 2.2 kD) was used to probe the association between the negatively charged hemicarceplex and the positively charged protein. The behavior of the resulting ternary protein-hemicarcerand-guest assembly was investigated in two binding limits: (1) when K(encaps) ≫ K(assoc), the hemicarcerand transports the ligand to the protein while protecting it from the aqueous medium; and (2) when K(assoc) > K(encaps), the hemicarcerand-protein complex is formed first, and the hemicarcerand acts as an artificial receptor site that intercepts ligands from solution and positions them close to the active site of the metalloenzyme. In both cases, ET mediated by the protein-bound hemicarcerand is much faster than that due to diffusional encounters with the respective free donor or acceptor in solution. The measured ET rates suggest that the dominant binding region of the host-guest complex on the surface of the protein is consistent with the docking area of the native redox partner of cytochrome c. The strong association with the protein is attributed to the flexible conformation and adaptable charge distribution of the hemicarcerand, which allow for surface-matching with the cytochrome.


Subject(s)
Cytochromes c/chemistry , Light , Crystallography, X-Ray , Electron Transport , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Solubility , Thermodynamics , Water/chemistry , Zinc/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL