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










Publication year range
2.
Oncotarget ; 9(40): 26072-26085, 2018 May 25.
Article in English | MEDLINE | ID: mdl-29899843

ABSTRACT

Resistance to chemotherapy is a major cause of treatment failure and poor overall survival in patients with lung cancer. Identification of molecular targets present in resistant cancer cells is essential for addressing therapeutic resistance and prolonging lung cancer patient survival. Members of the B-cell lymphoma 2 (Bcl-2) family of proteins are associated with chemotherapeutic resistance. In this study, we found that pro-survival protein Bcl-2 is upregulated in paclitaxel resistant cells, potentially contributing to chemotherapy resistance. To exploit the increase in Bcl-2 expression for targeting therapy resistance, we investigated the effects of a peptide derived from the nuclear receptor Nur77 that converts Bcl-2 from an anti-apoptotic protein to a pro-apoptotic protein. The Nur77 derived peptide preferentially induced apoptosis in paclitaxel-resistant cancer cells with high expression of Bcl-2. This peptide also induced apoptosis of multidrug resistant H69AR lung cancer cells that express Bcl-2 and inhibited their growth in 3D spheroids. The Nur77 peptide strongly suppressed the growth of paclitaxel-resistant lung cancer cells in a zebrafish xenograft tumor model. Taken together, our data supports a new strategy for treating lung cancers that acquire resistance to chemotherapy through overexpression of Bcl-2.

3.
Proc Natl Acad Sci U S A ; 110(19): 7808-13, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23603272

ABSTRACT

Host innate immune responses to DNA viruses involve members of the nucleotide-binding domain, leucine-rich repeat and pyrin domain containing protein (NLRP) family, which form "inflammasomes" that activate caspase-1, resulting in proteolytic activation of cytokines interleukin (IL)-1ß and IL-18. We hypothesized that DNA viruses would target inflammasomes to overcome host defense. A Vaccinia virus (VACV) B-cell CLL/lymphoma 2 (Bcl-2) homolog, F1L, was demonstrated to bind and inhibit the NLR family member NLRP1 in vitro. Moreover, infection of macrophages in culture with virus lacking F1L (ΔF1L) caused increased caspase-1 activation and IL-1ß secretion compared with wild-type virus. Virulence of ΔF1L virus was attenuated in vivo, causing altered febrile responses, increased proteolytic processing of caspase-1, and more rapid inflammation in lungs of infected mice without affecting cell death or virus replication. Furthermore, we found that a hexapeptide from F1L is necessary and sufficient for inhibiting the NLRP1 inflammasome in vitro, thus identifying a peptidyl motif required for binding and inhibiting NLRP1. The functional importance of this NLRP1-binding motif was further confirmed by studies of recombinant ΔF1L viruses reconstituted either with the wild-type F1L or a F1L mutant that fails to bind NLRP1. Cellular infection with wild-type F1L reconstituted virus-suppressed IL-1ß production, whereas mutant F1L did not. In contrast, both wild-type and mutant versions of F1L equally suppressed apoptosis. In vivo, the NLR nonbinding F1L mutant virus exhibited an attenuated phenotype similar to ΔF1L virus, thus confirming the importance of F1L interactions with NLRP1 for viral pathogenicity in mice. Altogether, these findings reveal a unique viral mechanism for evading host innate immune responses.


Subject(s)
Gene Expression Regulation, Viral , Immunity, Innate , Inflammasomes/metabolism , Vaccinia virus/metabolism , Viral Proteins/metabolism , Amino Acid Motifs , Animals , Caspases/metabolism , Chlorocebus aethiops , Cytokines/metabolism , HEK293 Cells , HeLa Cells , Humans , Interleukin-1beta/metabolism , Mice , Mice, Inbred BALB C , Mutation , Phenotype , Recombinant Proteins/metabolism , Vero Cells , Virulence
4.
J Biol Chem ; 287(47): 39470-9, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23012361

ABSTRACT

The K7L gene product of the smallpox virus is a protease implicated in the maturation of viral proteins. K7L belongs to protease Clan CE, which includes distantly related cysteine proteases from eukaryotes, pathogenic bacteria, and viruses. Here, we describe its recombinant high level expression, biochemical mechanism, substrate preference, and regulation. Earlier studies inferred that the orthologous I7L vaccinia protease cleaves at an AG-X motif in six viral proteins. Our data for K7L suggest that the AG-X motif is necessary but not sufficient for optimal cleavage activity. Thus, K7L requires peptides extended into the P7 and P8 positions for efficient substrate cleavage. Catalytic activity of K7L is substantially enhanced by homodimerization, by the substrate protein P25K as well as by glycerol. RNA and DNA also enhance cleavage of the P25K protein but not of synthetic peptides, suggesting that nucleic acids augment the interaction of K7L with its protein substrate. Library-based peptide preference analyses enabled us to design an activity-based probe that covalently and selectively labels K7L in lysates of transfected and infected cells. Our study thus provides proof-of-concept for the design of inhibitors and probes that may contribute both to a better understanding of the role of K7L in the virus life cycle and the design of novel anti-virals.


Subject(s)
Antiviral Agents/chemistry , Molecular Probes/chemistry , Peptide Hydrolases/chemistry , Peptide Library , Protease Inhibitors/chemistry , Variola virus/enzymology , Viral Proteins/antagonists & inhibitors , Amino Acid Motifs , Animals , Cell Line , Cricetinae , Drug Design , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Smallpox/drug therapy , Smallpox/enzymology , Smallpox/genetics , Variola virus/genetics , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
5.
Assay Drug Dev Technol ; 9(5): 465-74, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21561376

ABSTRACT

The human Bcl-2 family includes six antiapoptotic members (Bcl-2, Bcl-B, Bcl-W, Bcl-X(L), Bfl-1, and Mcl-1) and many proapoptotic members, wherein a balance between the two determines cell life or death in many physiological and disease contexts. Elevated expression of various antiapoptotic Bcl-2 members is commonly observed in cancers, and chemical inhibitors of these proteins have been shown to promote apoptosis of malignant cells in culture, in animal models, and in human clinical trials. All six antiapoptotic members bind a helix from the proapoptotic family member Bim, thus quenching Bim's apoptotic signal. Here, we describe the use of a multiplex, high-throughput flow cytometry assay for the discovery of small molecule modulators that disrupt the interaction between the antiapoptotic members of the Bcl-2 family and Bim. The six antiapoptotic Bcl-2 family members were expressed as glutathione-S-transferase fusion proteins and bound individually to six glutathione bead sets, with each set having a different intensity of red fluorescence. A fluorescein-conjugated Bcl-2 homology region 3 (BH3) peptide from Bim was employed as a universal ligand. Flow cytometry measured the amount of green peptide bound to each bead set in a given well, with inhibitory compounds resulting in a decrease of green fluorescence on one or more bead set(s). Hits and cheminformatically selected analogs were retested in a dose-response series, resulting in three "active" compounds for Bcl-B. These three compounds were validated by fluorescence polarization and isothermal titration calorimetry. We discuss some of the lessons learned about screening a chemical library provided by the National Institutes of Health Small Molecule Repository (∼195,000 compounds) using high-throughput flow cytometry.


Subject(s)
Apoptosis Regulatory Proteins/antagonists & inhibitors , Drug Discovery/methods , High-Throughput Screening Assays/methods , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Small Molecule Libraries/analysis , Animals , Apoptosis , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Binding, Competitive , Calorimetry/methods , Clinical Trials as Topic , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Flow Cytometry , Fluorescence Polarization/methods , Glutathione/metabolism , Green Fluorescent Proteins , Humans , Membrane Proteins/antagonists & inhibitors , Models, Chemical , Molecular Targeted Therapy , Protein Binding , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/analysis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , Reproducibility of Results
6.
Proc Natl Acad Sci U S A ; 106(10): 3935-40, 2009 Mar 10.
Article in English | MEDLINE | ID: mdl-19223583

ABSTRACT

NLRP1 (NLR family, pyrin domain-containing 1) is a contributor to innate immunity involved in intracellular sensing of pathogens, as well as danger signals related to cell injury. NLRP1 is one of the core components of caspase-1-activating platforms termed "inflammasomes," which are involved in proteolytic processing of interleukin-1beta (IL-1beta) and in cell death. We previously discovered that anti-apoptotic proteins Bcl-2 and Bcl-X(L) bind to and inhibit NLRP1 in cells. Using an in vitro reconstituted system employing purified recombinant proteins, we studied the mechanism by which Bcl-2 and Bcl-X(L) inhibit NLRP1. Bcl-2 and Bcl-X(L) inhibited caspase-1 activation induced by NLRP1 in a concentration-dependent manner, with K(i) approximately 10 nM. Bcl-2 and Bcl-X(L) were also determined to inhibit ATP binding to NLRP1, which is required for oligomerization of NLRP1, and Bcl-X(L) was demonstrated to interfere with NLRP1 oligomerization. Deletion of the flexible loop regions of Bcl-2 and Bcl-X(L), which are located between the first and second alpha-helices of these anti-apoptotic proteins and which were previously shown to be required for binding NLRP1, abrogated ability to inhibit caspase-1 activation, ATP binding and oligomerization of NLRP1. Conversely, synthetic peptides corresponding to the loop region of Bcl-2 were sufficient to potently inhibit NLRP1. These findings thus demonstrate that the loop domain is necessary and sufficient to inhibit NLRP1, providing insights into the mechanism by which anti-apoptotic proteins Bcl-2 and Bcl-X(L) inhibit NLRP1.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Inflammation/metabolism , bcl-X Protein/chemistry , bcl-X Protein/metabolism , Caspase 1/metabolism , Enzyme Activation , Kinetics , Peptides/chemistry , Peptides/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary , Structure-Activity Relationship
7.
Cancer Cell ; 14(4): 285-98, 2008 Oct 07.
Article in English | MEDLINE | ID: mdl-18835031

ABSTRACT

Bcl-2 can be converted into a proapoptotic molecule by nuclear receptor Nur77. However, the development of Bcl-2 converters as anticancer therapeutics has not been explored. Here we report the identification of a Nur77-derived Bcl-2-converting peptide with 9 amino acids (NuBCP-9) and its enantiomer, which induce apoptosis of cancer cells in vitro and in animals. The apoptotic effect of NuBCPs and their activation of Bax are not inhibited but rather potentiated by Bcl-2. NuBCP-9 and its enantiomer bind to the Bcl-2 loop, which shares the characteristics of structurally adaptable regions with many cancer-associated and signaling proteins. NuBCP-9s act as molecular switches to dislodge the Bcl-2 BH4 domain, exposing its BH3 domain, which in turn blocks the activity of antiapoptotic Bcl-X(L).


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , DNA-Binding Proteins/metabolism , Neoplasms, Experimental/drug therapy , Oligopeptides/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Steroid/metabolism , Animals , Antineoplastic Agents/chemistry , BH3 Interacting Domain Death Agonist Protein/metabolism , Binding Sites , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Female , HeLa Cells , Humans , Jurkat Cells , Mice , Mice, Knockout , Mice, SCID , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1 , Oligopeptides/chemistry , Peptide Fragments/pharmacology , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Proto-Oncogene Proteins/pharmacology , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, Steroid/genetics , Stereoisomerism , Time Factors , Transfection , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
8.
J Biomol Screen ; 13(7): 665-73, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18626112

ABSTRACT

Nuclear receptor TR3/Nur77/NR4A1 binds several antiapoptotic Bcl-2-family proteins (Bcl-B, Bcl-2, Bfl-1) in a non-BH3-dependent manner. A 9-amino-acid peptide derived from full-length TR3 with polyarginine tail (TR3-r8) recapitulates TR3's binding specificity, displaying high affinity for Bcl-B. TR3-r8 peptide was used to screen for small molecule Bcl-B inhibitors. A fluorescence polarization assay (FPA) employing fluorescein isothiocyanate (FITC)-labeled TR3-r8 peptide (FITC-TR3-r8) and Bcl-B protein was optimized, with nonfluorescent TR3-r8 serving to demonstrate reversible, competitive binding. Approximately 50,000 compounds were screened at 3.75 mg/L, yielding 145 reproducible hits with > or =50% FITC-TR3-r8 displacement (a confirmed hit rate of 0.29%). After dose-response analyses and counterscreening with an unrelated FITC-based FPA, 6 candidate compounds remained. Nuclear magnetic resonance (NMR) showed that 2 of these compounds bound Bcl-B, but not glutathione S-transferase (GST) control protein. One Bcl-B-binding compound was unable to displace FITClabeled BH3 peptides from Bcl-B, confirming a unique binding mechanism compared with traditional antagonists of antiapoptotic Bcl-2-family proteins. This compound bound Bcl-B with Kd 1.94 +/- 0.38 microM, as determined by isothermal titration calorimetry. Experiments using Bcl-B overexpressing HeLa cells demonstrated that this compound induced Bcl-B-dependent cell death. The current FPA represents a screen that can identify noncanonical inhibitors of Bcl-2-family proteins.


Subject(s)
DNA-Binding Proteins/chemistry , Drug Evaluation, Preclinical/methods , Fluorescence Polarization/methods , Peptides/chemistry , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Receptors, Steroid/chemistry , Amino Acid Sequence , Calorimetry , Drug Evaluation, Preclinical/instrumentation , Fluorescein-5-isothiocyanate/pharmacology , Glutathione Transferase/metabolism , HeLa Cells , Humans , Kinetics , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Nuclear Receptor Subfamily 4, Group A, Member 1
9.
Mol Cancer Ther ; 7(6): 1639-46, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18566235

ABSTRACT

The natural product gambogic acid (GA) has been reported to have cytotoxic activity against tumor cells in culture and was identified as an active compound in a cell-based high-throughput screening assay for activators of caspases, proteases involved in apoptosis. Using the antiapoptotic Bcl-2 family protein, Bfl-1, as a target for screening of a library of natural products, we identified GA as a competitive inhibitor that displaced BH3 peptides from Bfl-1 in a fluorescence polarization assay. Analysis of competition for BH3 peptide binding revealed that GA inhibits all six human Bcl-2 family proteins to various extents, with Mcl-1 and Bcl-B the most potently inhibited [concentrations required for 50% inhibition (IC(50)), < 1 micromol/L]. Competition for BH3 peptide binding was also confirmed using a time-resolved fluorescence resonance energy transfer assay. GA functionally inhibited the antiapoptotic Bcl-2 family proteins as shown by experiments using isolated mitochondria in which recombinant purified Bcl-2 family proteins suppress SMAC release in vitro, showing that GA neutralizes their suppressive effects on mitochondria in a concentration-dependent manner. GA killed tumor cell lines via an apoptotic mechanism, whereas analogues of GA with greatly reduced potency at BH3 peptide displacement showed little or no cytotoxic activity. However, GA retained cytotoxic activity against bax-/-bak-/- cells in which antiapoptotic Bcl-2 family proteins lack a cytoprotective phenotype, implying that GA also has additional targets that contribute to its cytotoxic mechanism. Altogether, the findings suggest that suppression of antiapoptotic Bcl-2 family proteins may be among the cytotoxic mechanisms by which GA kills tumor cells.


Subject(s)
Apoptosis/drug effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Xanthones/pharmacology , Animals , BH3 Interacting Domain Death Agonist Protein/metabolism , Binding Sites , Binding, Competitive , Cell Line, Tumor , Cytoprotection/drug effects , Drug Screening Assays, Antitumor , Fluorescein-5-isothiocyanate/metabolism , Fluorescence Resonance Energy Transfer , Humans , Mice , Minor Histocompatibility Antigens , Mitochondria/drug effects , Mitochondria/metabolism , Peptides/pharmacology , Time Factors , Xanthones/chemistry , bcl-2-Associated X Protein/metabolism
10.
J Biol Chem ; 283(15): 9580-6, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18178565

ABSTRACT

The pro-apoptotic members of the Bcl-2 family include initiator proteins that contain only BH3 domains and downstream effector multi-BH domain-containing proteins, including Bax and Bak. In this report, we compared the ability of the six human anti-apoptotic Bcl-2 family members to suppress apoptosis induced by overexpression of Bax or Bak, correlating findings with protein interactions measured by three different methods: co-immunoprecipitation, glutathione S-transferase pulldown, and fluorescence polarization assays employing synthetic BH3 peptides from Bax and Bak. Bcl-B and Mcl-1 showed strong preferences for binding to and suppression of Bax and Bak, respectively. In contrast, the other anti-apoptotic Bcl-2 family proteins (Bcl-2, Bcl-X(L), Bcl-W, and Bfl-1) suppressed apoptosis induced by overexpression of either Bax or Bak, and they displayed an ability to bind both Bax and Bak by at least one of the three protein interaction methods. Interestingly, however, full-length Bax and Bak proteins and synthetic Bax and Bak BH3 peptides exhibited discernible differences in their interactions with some anti-apoptotic members of the Bcl-2 family, cautioning against reliance on a single method for detecting protein interactions of functional significance. Altogether, the findings reveal striking distinctions in the behaviors of Bcl-B and Mcl-1 relative to the other anti-apoptotic Bcl-2 family members, where Bcl-B and Mcl-1 display reciprocal abilities to bind and neutralize Bax and Bak.


Subject(s)
Apoptosis/physiology , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Gene Expression , HeLa Cells , Humans , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/genetics , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/genetics
11.
Biochem Biophys Res Commun ; 366(3): 807-13, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18082620

ABSTRACT

RIZ1 is a transcriptional regulator and tumor suppressor that catalyzes methylation of lysine 9 of histone H3. It contains a distinct SET domain, sometimes referred to as PR (PRDI-BF1 and RIZ1 homology) domain, that is responsible for its catalytic activity. We determined the solution structure of the PR domain from RIZ1 and characterized its interaction with S-adenosyl-l-homocysteine (SAH) and a peptide from histone H3. Despite low sequence identity with canonical SET domains, the PR domain displays a typical SET fold including a pseudo-knot at the C-terminus. The N-flanking sequence of RIZ1 PR domain adopts a novel conformation and interacts closely with the SET fold. The C-flanking sequence contains an alpha-helix that points away from the protein face that harbors active site in other SET domains. The SET fold of RIZ1 does not have detectable affinity for SAH but it interacts with a synthetic peptide comprising residues 1-20 of histone H3.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/ultrastructure , Models, Chemical , Models, Molecular , Nuclear Proteins/chemistry , Nuclear Proteins/ultrastructure , Transcription Factors/chemistry , Transcription Factors/ultrastructure , Amino Acid Sequence , Computer Simulation , Histone-Lysine N-Methyltransferase , Molecular Sequence Data , Protein Conformation , Protein Structure, Tertiary
12.
Chem Biol Drug Des ; 70(5): 383-92, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17927731

ABSTRACT

Humanin is a short endogenous peptide, which can provide protection from cell death through its association with various receptors, including the pro-apoptotic Bcl-2 family proteins Bid, Bim, and Bax. By using NMR chemical shift mapping experiments, we demonstrate that the interaction between Humanin-derived peptides and Bid is specific, and we localize the binding site to a region on the surface of Bid, which includes residues from the conserved helical BH3 domain of the protein. The BH3 domain mediates the association of Bid with other Bcl-2 family members and is essential for the protein's cytotoxic activity. The data suggest that Humanin exerts its cytoprotective activity by engaging the Bid BH3 domain; this would hinder the association of Bid with other Bcl-2 family proteins, thereby mitigating its toxicity. The identification of a Humanin-specific binding site on the surface of Bid reinforces its importance as a direct modulator of programmed cell death, and suggests a strategy for the design of cytoprotective peptide inhibitors of Bid.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein/chemistry , Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/chemistry , Proto-Oncogene Proteins/chemistry , bcl-2-Associated X Protein/chemistry , Amino Acid Sequence , Animals , Bcl-2-Like Protein 11 , Cell Death/physiology , Conserved Sequence , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Alignment , Sequence Homology, Amino Acid
13.
Blood ; 109(9): 3849-55, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17227826

ABSTRACT

Defects in apoptosis mechanisms play important roles in malignancy and autoimmunity. Orphan nuclear receptor Nur77/TR3 has been demonstrated to bind antiapoptotic protein Bcl-2 and convert it from a cytoprotective to a cytodestructive protein, representing a phenotypic conversion mechanism. Of the 6 antiapoptotic human Bcl-2 family members, we found that Nur77/TR3 binds strongest to Bcl-B, showing selective reactivity with Bcl-B, Bcl-2, and Bfl-1 but not Bcl-X(L), Mcl-1, or Bcl-W. Nur77 converts the phenotype of Bcl-B from antiapoptotic to proapoptotic. Bcl-B is prominently expressed in plasma cells and multiple myeloma. Endogenous Bcl-B associates with endogenous Nur77 in RPMI 8226 myeloma cells, where RNA interference experiments demonstrated dependence on Bcl-B for Nur77-induced apoptosis. Furthermore, a Nur77-mimicking peptide killed RPMI 8226 myeloma cells through a Bcl-B-dependent mechanism. Because Bcl-B is abundantly expressed in plasma cells and some myelomas, these findings raise the possibility of exploiting the Nur77/Bcl-B mechanism for apoptosis for eradication of autoimmune plasma cells or myeloma.


Subject(s)
DNA-Binding Proteins/pharmacology , Gene Expression Regulation, Neoplastic , Multiple Myeloma/metabolism , Peptides/pharmacology , Plasma Cells/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription Factors/pharmacology , Animals , Apoptosis Regulatory Proteins/immunology , Apoptosis Regulatory Proteins/metabolism , Autoimmunity/drug effects , COS Cells , Cell Death/drug effects , Cell Death/immunology , Chlorocebus aethiops , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/immunology , HeLa Cells , Humans , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1 , Peptides/immunology , Peptides/metabolism , Plasma Cells/immunology , Plasma Cells/pathology , Proto-Oncogene Proteins c-bcl-2/immunology , Receptors, Cytoplasmic and Nuclear/immunology , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/immunology , Receptors, Steroid/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism
14.
J Med Chem ; 49(1): 27-30, 2006 Jan 12.
Article in English | MEDLINE | ID: mdl-16392787

ABSTRACT

We have recently identified a series of compounds that efficiently inhibit anthrax lethal factor (LF) metallo-protease. Here we present further structure-activity relationship and CoMFA (comparative molecular field analysis) studies on newly derived inhibitors. The obtained 3D QSAR model was subsequently compared with the X-ray structure of the complex between LF and a representative compound. Our studies form the basis for the rational design of additional compounds with improved activity and selectivity.


Subject(s)
Bacillus anthracis/enzymology , Bacterial Toxins/antagonists & inhibitors , Metalloendopeptidases/antagonists & inhibitors , Protease Inhibitors , Rhodanine , Antigens, Bacterial , Crystallography, X-Ray , Models, Molecular , Molecular Structure , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Rhodanine/analogs & derivatives , Rhodanine/chemical synthesis , Rhodanine/pharmacology , Structure-Activity Relationship
15.
J Biol Chem ; 280(39): 33620-6, 2005 Sep 30.
Article in English | MEDLINE | ID: mdl-16009714

ABSTRACT

Epstein-Barr virus is a human herpesvirus that causes infectious mononucleosis and lymphoproliferative malignancies. LMP1 (latent membrane protein-1), which is encoded by this virus and which is essential for transformation of B lymphocytes, acts as a constitutively active mimic of the tumor necrosis factor receptor (TNFR) CD40. LMP1 is an integral membrane protein containing six transmembrane segments and a cytoplasmic domain at the C terminus that binds to intracellular TNFR-associated factors (TRAFs). TRAFs are intracellular co-inducers of downstream signaling from CD40 and other TNFRs, and TRAF3 is required for activation of B lymphocytes by LMP1. Cytoplasmic C-terminal activation region 1 of LMP1 bears a motif (PQQAT) that conforms to the TRAF recognition motif PVQET in CD40. In this study, we report the crystal structure of this portion of LMP1 C-terminal activation region-1 (204PQQATDD210) bound in complex with TRAF3. The PQQAT motif is bound in the same binding crevice on TRAF3 where CD40 is bound, providing a molecular mechanism for LMP1 to act as a CD40 decoy for TRAF3. The LMP1 motif is presented in the TRAF3 crevice as a close structural mimic of the PVQET motif in CD40, and the intermolecular contacts are similar. However, the viral protein makes a unique contact: a hydrogen bond network formed between Asp210 in LMP1 and Tyr395 and Arg393 in TRAF3. This intermolecular contact is not made in the CD40-TRAF3 complex. The additional hydrogen bonds may stabilize the complex and strengthen the binding to permit LMP1 to compete with CD40 for binding to the TRAF3 crevice, influencing downstream signaling to B lymphocytes and contributing to dysregulated signaling by LMP1.


Subject(s)
B-Lymphocytes/metabolism , CD40 Antigens/chemistry , Herpesvirus 4, Human/chemistry , TNF Receptor-Associated Factor 3/metabolism , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/metabolism , Alanine/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Animals , B-Lymphocytes/virology , Binding Sites , Blotting, Western , CD40 Antigens/metabolism , Cell Transformation, Viral , Crystallography, X-Ray , Humans , Lymphocyte Activation , Mice , Microscopy, Fluorescence , Models, Molecular , Precipitin Tests , Transfection , Viral Matrix Proteins/genetics
16.
J Biol Chem ; 280(16): 15825-35, 2005 Apr 22.
Article in English | MEDLINE | ID: mdl-15661735

ABSTRACT

Humanin (HN) is a recently identified endogenous peptide that protects cells against cytotoxicity induced by various stimuli. Recently, we showed that HN binds to and inhibits Bax, a proapoptotic Bcl-2 family protein, suggesting a mechanism for HN action. In this study, we identified Bim, a Bcl-2 homology 3-only member of the Bcl-2/Bax family, as an additional HN target protein. Using in vitro protein binding, immunoprecipitation, and coimmunolocalization assays, we demonstrated that HN binds directly to the extra long isoform of Bim (BimEL) but not the long (BimL) or short (BimS) isoforms. HN also protects cells against apoptosis induced by BimEL but not BimL and BimS in gene transfection studies. In contrast, mutants of HN which failed to bind BimEL failed to protect from BimEL-induced cell death. Moreover, HN inhibited BimEL-induced release of SMAC and cytochrome c from mitochondria isolated from bax-/-cells, indicating that HN can suppress BimEL independently of its effect on Bax. Finally, we demonstrate that HN prevents BimEL-induced oligomerization of Bak using isolated mitochondria. Taken together, our results indicate that the inhibition of BimEL may contribute to the antiapoptotic properties of the HN peptide.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/physiology , Proteins/metabolism , Animals , Apoptosis Regulatory Proteins , Bcl-2-Like Protein 11 , COS Cells , Carrier Proteins/metabolism , Cytochromes c/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Peptide Fragments/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2 Homologous Antagonist-Killer Protein , bcl-2-Associated X Protein
17.
J Biol Chem ; 280(16): 15815-24, 2005 Apr 22.
Article in English | MEDLINE | ID: mdl-15661737

ABSTRACT

Recently, we discovered that Humanin (HN), a small endogenous peptide of 24 amino acids, binds to and inhibits the proapoptotic protein Bax. We show here that HN also interacts with the BH3-only Bcl-2/Bax family protein, Bid, as well as a truncated form of Bid (tBid) associated with protease-mediated activation of this proapoptotic protein. Synthetic HN peptide binds purified Bid and tBid in vitro and blocks tBid-induced release of cytochrome c and SMAC from isolated mitochondria, whereas mutant peptides that fail to bind Bid or tBid lack this activity. Moreover, HN peptide also retained protective activity on bax-/-mitochondria, indicating that HN can block tBid-induced release of apoptogenic proteins from these organelles in a Bax-independent manner. HN peptide inhibits tBid-induced oligomerization of Bax and Bak in mitochondrial membranes, as shown by experiments with chemical cross-linkers or gel filtration. Gene transfection experiments showed that HN (but not an inactive mutant of HN) also protects intact cells from apoptosis induced by overexpression of tBid. We conclude that Bid represents an additional cellular target of HN, and we propose that HN-mediated suppression of Bid contributes to the antiapoptotic activity of this endogenous peptide.


Subject(s)
Carrier Proteins/metabolism , Membrane Proteins/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mitochondria/metabolism , Peptides/metabolism , Structure-Activity Relationship , bcl-2 Homologous Antagonist-Killer Protein , bcl-2-Associated X Protein
18.
J Immunol ; 173(12): 7394-400, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15585864

ABSTRACT

B cell-activating factor belonging to the TNF family receptor (BAFF-R), a member of the TNFR superfamily, plays a role in autoimmunity after ligation with BAFF ligand (also called TALL-1, BLyS, THANK, or zTNF4). BAFF/BAFF-R interactions are critical for B cell regulation, and signaling from this ligand-receptor complex results in NF-kappaB activation. Most TNFRs transmit signals intracellularly by recruitment of adaptor proteins called TNFR-associated factors (TRAFs). However, BAFF-R binds only one TRAF adaptor, TRAF3, and this interaction negatively regulates activation of NF-kappaB. In this study, we report the crystal structure of a 24-residue fragment of the cytoplasmic portion of BAFF-R bound in complex with TRAF3. The recognition motif (162)PVPAT(166) in BAFF-R is accommodated in the same binding crevice on TRAF3 that binds two related TNFRs, CD40 and LTbetaR, but is presented in a completely different structural framework. This region of BAFF-R assumes an open conformation with two extended strands opposed at right angles that each make contacts with TRAF3. The recognition motif is located in the N-terminal arm and intermolecular contacts mediate TRAF recognition. In the C-terminal arm, key stabilizing contacts are made, including critical hydrogen bonds with Gln(379) in TRAF3 that define the molecular basis for selective binding of BAFF-R solely to this member of the TRAF family. A dynamic conformational adjustment of Tyr(377) in TRAF3 occurs forming a new intermolecular contact with BAFF-R that stabilizes the complex. The structure of the complex provides a molecular explanation for binding affinities and selective protein interactions in TNFR-TRAF interactions.


Subject(s)
Intracellular Fluid/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/immunology , TNF Receptor-Associated Factor 3/chemistry , TNF Receptor-Associated Factor 3/metabolism , Amino Acid Motifs , Amino Acid Sequence , B-Cell Activation Factor Receptor , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Line , Crystallography, X-Ray , Cytoplasm/chemistry , Cytoplasm/genetics , Cytoplasm/metabolism , DNA Mutational Analysis , Humans , Membrane Proteins/genetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding/genetics , Protein Binding/immunology , Protein Conformation , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction/genetics , TNF Receptor-Associated Factor 3/genetics , Thermodynamics
19.
J Biol Chem ; 278(50): 50523-9, 2003 Dec 12.
Article in English | MEDLINE | ID: mdl-14517219

ABSTRACT

Lymphotoxin-beta receptor (LTbetaR) and CD40 are members of the tumor necrosis factor family of signaling receptors that regulate cell survival or death through activation of NF-kappaB. These receptors transmit signals through downstream adaptor proteins called tumor necrosis factor receptor-associated factors (TRAFs). In this study, the crystal structure of a region of the cytoplasmic domain of LTbetaR bound to TRAF3 has revealed an unexpected new recognition motif, 388IPEEGD393, for TRAF3 binding. Although this motif is distinct in sequence and structure from the PVQET motif in CD40 and PIQCT in the regulator TRAF-associated NF-kappaB activator (TANK), recognition is mediated in the same binding crevice on the surface of TRAF3. The results reveal structurally adaptive "hot spots" in the TRAF3-binding crevice that promote molecular interactions driving specific signaling after contact with LTbetaR, CD40, or the downstream regulator TANK.


Subject(s)
Adaptor Proteins, Signal Transducing , CD40 Antigens/biosynthesis , Receptors, Tumor Necrosis Factor/chemistry , Signal Transduction , Amino Acid Motifs , Amino Acid Sequence , CD40 Antigens/chemistry , Cell Line , Cell Survival , Crystallography, X-Ray , DNA, Complementary/metabolism , Electrons , Glutathione Transferase/metabolism , Humans , Lymphotoxin beta Receptor , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Proteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Fusion Proteins/metabolism
20.
Nature ; 423(6938): 456-61, 2003 May 22.
Article in English | MEDLINE | ID: mdl-12732850

ABSTRACT

Bax (Bcl2-associated X protein) is an apoptosis-inducing protein that participates in cell death during normal development and in various diseases. Bax resides in an inactive state in the cytosol of many cells. In response to death stimuli, Bax protein undergoes conformational changes that expose membrane-targeting domains, resulting in its translocation to mitochondrial membranes, where Bax inserts and causes release of cytochrome c and other apoptogenic proteins. It is unknown what controls conversion of Bax from the inactive to active conformation. Here we show that Bax interacts with humanin (HN), an anti-apoptotic peptide of 24 amino acids encoded in mammalian genomes. HN prevents the translocation of Bax from cytosol to mitochondria. Conversely, reducing HN expression by small interfering RNAs sensitizes cells to Bax and increases Bax translocation to membranes. HN peptides also block Bax association with isolated mitochondria, and suppress cytochrome c release in vitro. Notably, the mitochondrial genome contains an identical open reading frame, and the mitochondrial version of HN can also bind and suppress Bax. We speculate therefore that HN arose from mitochondria and transferred to the nuclear genome, providing a mechanism for protecting these organelles from Bax.


Subject(s)
Apoptosis , Peptides/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/antagonists & inhibitors , Amino Acid Sequence , Cell Line , Cell Membrane/metabolism , Cell Nucleus/genetics , Cytochrome c Group/metabolism , Cytosol/metabolism , DNA, Mitochondrial/genetics , Humans , Intracellular Signaling Peptides and Proteins , Mitochondria/genetics , Mitochondria/metabolism , Molecular Sequence Data , Peptides/chemistry , Peptides/genetics , Protein Binding , Protein Transport , Proteins/chemistry , Proteins/genetics , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...