Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Haematologica ; 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37981834

ABSTRACT

Multiple myeloma (MM) is a malignancy of plasma cells whose antibody secretion creates proteotoxic stress relieved by the N-end rule pathway, a proteolytic system that degrades Narginylated proteins in the proteasome. When the proteasome is inhibited, protein cargo is alternatively targeted for autophagic degradation by binding to the ZZ-domain of p62/sequestosome-1. Here, we demonstrate that XRK3F2, a selective ligand for the ZZ-domain, dramatically improved two major responses to the proteasome inhibitor bortezomib by increasing: 1) killing of human MM cells by stimulating both bortezomib mediated apoptosis and necroptosis, a process regulated by p62; and 2) preservation of bone mass by stimulating osteoblasts differentiation and inhibiting osteoclastic bone destruction. Co-administration of bortezomib and XRK3F2 inhibited both branches of the bimodal N-end rule pathway exhibited synergistic anti-MM effects on MM cell lines and CD138+ cells from MM patients, and prevented stromal-mediated MM cell survival. In mice with established human MM, coadministration of bortezomib and XRK3F2 decreased tumor burden and prevented the progression of MM-induced osteolytic disease by inducing new bone formation more effectively than either single agent alone. The results suggest that p62-ZZ ligands enhance the anti-MM efficacy of proteasome inhibitors and can reduce MM morbidity and mortality by improving bone health.

2.
Cancers (Basel) ; 14(3)2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35159039

ABSTRACT

Multiple myeloma (MM) remains incurable for most patients due to the emergence of drug resistant clones. Here we report a p53-independent mechanism responsible for Growth Factor Independence-1 (GFI1) support of MM cell survival by its modulation of sphingolipid metabolism to increase the sphingosine-1-phosphate (S1P) level regardless of the p53 status. We found that expression of enzymes that control S1P biosynthesis, SphK1, dephosphorylation, and SGPP1 were differentially correlated with GFI1 levels in MM cells. We detected GFI1 occupancy on the SGGP1 gene in MM cells in a predicted enhancer region at the 5' end of intron 1, which correlated with decreased SGGP1 expression and increased S1P levels in GFI1 overexpressing cells, regardless of their p53 status. The high S1P:Ceramide intracellular ratio in MM cells protected c-Myc protein stability in a PP2A-dependent manner. The decreased MM viability by SphK1 inhibition was dependent on the induction of autophagy in both p53WT and p53mut MM. An autophagic blockade prevented GFI1 support for viability only in p53mut MM, demonstrating that GFI1 increases MM cell survival via both p53WT inhibition and upregulation of S1P independently. Therefore, GFI1 may be a key therapeutic target for all types of MM that may significantly benefit patients that are highly resistant to current therapies.

3.
J Hematol Oncol ; 11(1): 123, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30286780

ABSTRACT

BACKGROUND: In spite of major advances in treatment, multiple myeloma (MM) is currently an incurable malignancy due to the emergence of drug-resistant clones. We previously showed that MM cells upregulate the transcriptional repressor, growth factor independence 1 (Gfi1), in bone marrow stromal cells (BMSCs) that induces prolonged inhibition of osteoblast differentiation. However, the role of Gfi1 in MM cells is unknown. METHODS: Human primary CD138+ and BMSC were purified from normal donors and MM patients' bone marrow aspirates. Gfi1 knockdown and overexpressing cells were generated by lentiviral-mediated shRNA. Proliferation/apoptosis studies were done by flow cytometry, and protein levels were determined by Western blot and/or immunohistochemistry. An experimental MM mouse model was generated to investigate the effects of MM cells overexpressing Gfi1 on tumor burden and osteolysis in vivo. RESULTS: We found that Gfi1 expression is increased in patient's MM cells and MM cell lines and was further increased by co-culture with BMSC, IL-6, and sphingosine-1-phosphate. Modulation of Gfi1 in MM cells had major effects on their survival and growth. Knockdown of Gfi1 induced apoptosis in p53-wt, p53-mutant, and p53-deficient MM cells, while Gfi1 overexpression enhanced MM cell growth and protected MM cells from bortezomib-induced cell death. Gfi1 enhanced cell survival of p53-wt MM cells by binding to p53, thereby blocking binding to the promoters of the pro-apoptotic BAX and NOXA genes. Further, Gfi1-p53 binding could be blocked by HDAC inhibitors. Importantly, inoculation of MM cells overexpressing Gfi1 in mice induced increased bone destruction, increased osteoclast number and size, and enhanced tumor growth. CONCLUSIONS: These results support that Gfi1 plays a key role in MM tumor growth, survival, and bone destruction and contributes to bortezomib resistance, suggesting that Gfi1 may be a novel therapeutic target for MM.


Subject(s)
DNA-Binding Proteins/biosynthesis , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Osteogenesis/physiology , Transcription Factors/biosynthesis , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Cell Line, Tumor , Female , Humans , Mice
4.
Article in English | MEDLINE | ID: mdl-30008697

ABSTRACT

Multiple myeloma bone disease (MMBD) is characterized by non-healing lytic bone lesions that persist even after a patient has achieved a hematologic remission. We previously reported that p62 (sequestosome-1) in bone marrow stromal cells (BMSC) is critical for the formation of MM-induced signaling complexes that mediate OB suppression. Importantly, XRK3F2, an inhibitor of the p62-ZZ domain, blunted MM-induced Runx2 suppression in vitro, and induced new bone formation and remodeling in the presence of tumor in vivo. Additionally, we reported that MM cells induce the formation of repressive chromatin on the Runx2 gene in BMSC via direct binding of the transcriptional repressor GFI1, which recruits the histone modifiers, histone deacetylase 1 (HDAC1) and Enhancer of zeste homolog 2 (EZH2). In this study we investigated the mechanism by which blocking p62-ZZ domain-dependent signaling prevents MM-induced suppression of Runx2 in BMSC. XRK3F2 prevented MM-induced upregulation of Gfi1 and repression of the Runx2 gene when present in MM-preOB co-cultures. We also show that p62-ZZ-domain blocking by XRK3F2 also prevented MM conditioned media and TNF plus IL7-mediated Gfi1 mRNA upregulation and the concomitant Runx2 repression, indicating that XRK3F2's prevention of p62-ZZ domain signaling within preOB is involved in the response. Chromatin immunoprecipitation (ChIP) analyses revealed that XRK3F2 decreased MM-induced GFI1 occupancy at the Runx2-P1 promoter and prevented recruitment of HDAC1, thus preserving the transcriptionally permissive chromatin mark H3K9ac on Runx2 and allowing osteogenic differentiation. Furthermore, treatment of MM-exposed preOB with XRK3F2 after MM removal decreased GFI1 enrichment at Runx2-P1 and rescued MM-induced suppression of Runx2 mRNA and its downstream osteogenic gene targets together with increased osteogenic differentiation. Further, primary BMSC (hBMSC) from MM patients (MM-hBMSC) had little ability to increase H3K9ac on the Runx2 promoter in osteogenic conditions when compared to hBMSC from healthy donors (HD). XRK3F2 treatment enriched Runx2 gene H3K9ac levels in MM-hBMSC to the level observed in HD-hBMSC, but did not alter HD-hBMSC H3K9ac. Importantly, XRK3F2 treatment of long-term MM-hBMSC cultures rescued osteogenic differentiation and mineralization. Our data show that blocking p62-ZZ domain-dependent signaling with XRK3F2 can reverse epigenetic-based mechanisms of MM-induced Runx2 suppression and promote osteogenic differentiation.

5.
Mol Cancer Res ; 15(4): 405-417, 2017 04.
Article in English | MEDLINE | ID: mdl-28119431

ABSTRACT

In multiple myeloma, osteolytic lesions rarely heal because of persistent suppressed osteoblast differentiation resulting in a high fracture risk. Herein, chromatin immunoprecipitation analyses reveal that multiple myeloma cells induce repressive epigenetic histone changes at the Runx2 locus that prevent osteoblast differentiation. The most pronounced multiple myeloma-induced changes were at the Runx2-P1 promoter, converting it from a poised bivalent state to a repressed state. Previously, it was observed that multiple myeloma induces the transcription repressor GFI1 in osteoblast precursors, which correlates with decreased Runx2 expression, thus prompting detailed characterization of the multiple myeloma and TNFα-dependent GFI1 response element within the Runx2-P1 promoter. Further analyses reveal that multiple myeloma-induced GFI1 binding to Runx2 in osteoblast precursors and recruitment of the histone modifiers HDAC1, LSD1, and EZH2 is required to establish and maintain Runx2 repression in osteogenic conditions. These GFI1-mediated repressive chromatin changes persist even after removal of multiple myeloma. Ectopic GFI1 is sufficient to bind to Runx2, recruit HDAC1 and EZH2, increase H3K27me3 on the gene, and prevent osteogenic induction of endogenous Runx2 expression. Gfi1 knockdown in MC4 cells blocked multiple myeloma-induced recruitment of HDAC1 and EZH2 to Runx2, acquisition of repressive chromatin architecture, and suppression of osteoblast differentiation. Importantly, inhibition of EZH2 or HDAC1 activity in pre-osteoblasts after multiple myeloma exposure in vitro or in osteoblast precursors from patients with multiple myeloma reversed the repressive chromatin architecture at Runx2 and rescued osteoblast differentiation.Implications: This study suggests that therapeutically targeting EZH2 or HDAC1 activity may reverse the profound multiple myeloma-induced osteoblast suppression and allow repair of the lytic lesions. Mol Cancer Res; 15(4); 405-17. ©2017 AACR.


Subject(s)
Core Binding Factor Alpha 1 Subunit/genetics , DNA-Binding Proteins/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Histone Deacetylase 1/metabolism , Multiple Myeloma/genetics , Osteoblasts/cytology , Transcription Factors/metabolism , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Coculture Techniques , Core Binding Factor Alpha 1 Subunit/metabolism , DNA-Binding Proteins/genetics , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Epigenesis, Genetic , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/pharmacology , Humans , Indoles/administration & dosage , Indoles/pharmacology , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Osteoblasts/drug effects , Promoter Regions, Genetic , Pyridones/administration & dosage , Pyridones/pharmacology , Transcription Factors/genetics
6.
J Biol Chem ; 288(42): 30399-30410, 2013 Oct 18.
Article in English | MEDLINE | ID: mdl-24005670

ABSTRACT

Abnormal osteoclast formation and osteolysis are the hallmarks of multiple myeloma (MM) bone disease, yet the underlying molecular mechanisms are incompletely understood. Here, we show that the AKT pathway was up-regulated in primary bone marrow monocytes (BMM) from patients with MM, which resulted in sustained high expression of the receptor activator of NF-κB (RANK) in osteoclast precursors. The up-regulation of RANK expression and osteoclast formation in the MM BMM cultures was blocked by AKT inhibition. Conditioned media from MM cell cultures activated AKT and increased RANK expression and osteoclast formation in BMM cultures. Inhibiting AKT in cultured MM cells decreased their growth and ability to promote osteoclast formation. Of clinical significance, systemic administration of the AKT inhibitor LY294002 blocked the formation of tumor tissues in the bone marrow cavity and essentially abolished the MM-induced osteoclast formation and osteolysis in SCID mice. The level of activating transcription factor 4 (ATF4) protein was up-regulated in the BMM cultures from multiple myeloma patients. Adenoviral overexpression of ATF4 activated RANK expression in osteoclast precursors. These results demonstrate a new role of AKT in the MM promotion of osteoclast formation and bone osteolysis through, at least in part, the ATF4-dependent up-regulation of RANK expression in osteoclast precursors.


Subject(s)
Gene Expression Regulation, Neoplastic , Multiple Myeloma/enzymology , Osteoclasts/enzymology , Osteolysis/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation , Activating Transcription Factor 4/metabolism , Animals , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Female , Heterografts , Humans , Male , Mice , Mice, SCID , Morpholines/pharmacology , Multiple Myeloma/pathology , Neoplasm Transplantation , Osteoclasts/pathology , Osteolysis/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptor Activator of Nuclear Factor-kappa B/metabolism , Tumor Cells, Cultured
7.
Blood ; 118(26): 6871-80, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22042697

ABSTRACT

Protracted inhibition of osteoblast (OB) differentiation characterizes multiple myeloma (MM) bone disease and persists even when patients are in long-term remission. However, the underlying pathophysiology for this prolonged OB suppression is unknown. Therefore, we developed a mouse MM model in which the bone marrow stromal cells (BMSCs) remained unresponsive to OB differentiation signals after removal of MM cells. We found that BMSCs from both MM-bearing mice and MM patients had increased levels of the transcriptional repressor Gfi1 compared with controls and that Gfi1 was a novel transcriptional repressor of the critical OB transcription factor Runx2. Trichostatin-A blocked the effects of Gfi1, suggesting that it induces epigenetic changes in the Runx2 promoter. MM-BMSC cell-cell contact was not required for MM cells to increase Gfi1 and repress Runx2 levels in MC-4 before OBs or naive primary BMSCs, and Gfi1 induction was blocked by anti-TNF-α and anti-IL-7 antibodies. Importantly, BMSCs isolated from Gfi1(-/-) mice were significantly resistant to MM-induced OB suppression. Strikingly, siRNA knockdown of Gfi1 in BMSCs from MM patients significantly restored expression of Runx2 and OB differentiation markers. Thus, Gfi1 may have an important role in prolonged MM-induced OB suppression and provide a new therapeutic target for MM bone disease.


Subject(s)
Bone Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Multiple Myeloma/metabolism , Osteoblasts/metabolism , Stromal Cells/metabolism , Transcription Factors/metabolism , 3T3 Cells , Animals , Blotting, Western , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , DNA-Binding Proteins/genetics , Female , Gene Expression , Humans , Interleukin-7/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Osteoblasts/pathology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/pathology , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
Blood ; 115(1): 140-9, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-19887675

ABSTRACT

Increased osteoclastogenesis and angiogenesis occur in physiologic and pathologic conditions. However, it is unclear if or how these processes are linked. To test the hypothesis that osteoclasts stimulate angiogenesis, we modulated osteoclast formation in fetal mouse metatarsal explants or in adult mice and determined the effect on angiogenesis. Suppression of osteoclast formation with osteoprotegerin dose-dependently inhibited angiogenesis and osteoclastogenesis in metatarsal explants. Conversely, treatment with parathyroid hormone related protein (PTHrP) increased explant angiogenesis, which was completely blocked by osteoprotegerin. Further, treatment of mice with receptor activator of nuclear factor-kappaB ligand (RANKL) or PTHrP in vivo increased calvarial vessel density and osteoclast number. We next determined whether matrix metalloproteinase-9 (MMP-9), an angiogenic factor predominantly produced by osteoclasts in bone, was important for osteoclast-stimulated angiogenesis. The pro-angiogenic effects of PTHrP or RANKL were absent in metatarsal explants or calvaria in vivo, respectively, from Mmp9(-/-) mice, demonstrating the importance of MMP-9 for osteoclast-stimulated angiogenesis. Lack of MMP-9 decreased osteoclast numbers and abrogated angiogenesis in response to PTHrP or RANKL in explants and in vivo but did not decrease osteoclast differentiation in vitro. Thus, MMP-9 modulates osteoclast-stimulated angiogenesis primarily by affecting osteoclasts, most probably by previously reported migratory effects on osteoclasts. These results clearly demonstrate that osteoclasts stimulate angiogenesis in vivo through MMP-9.


Subject(s)
Metatarsal Bones/blood supply , Neovascularization, Physiologic , Osteoclasts/physiology , Angiogenesis Inducing Agents/metabolism , Animals , Female , Fetus/blood supply , Fetus/drug effects , Humans , Male , Matrix Metalloproteinase 9/deficiency , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Metatarsal Bones/drug effects , Metatarsal Bones/embryology , Mice , Mice, Inbred C57BL , Models, Biological , Neovascularization, Physiologic/drug effects , Osteoclasts/drug effects , Osteoclasts/enzymology , Parathyroid Hormone-Related Protein/pharmacology , RANK Ligand/pharmacology , Skull/cytology , Skull/drug effects , Skull/enzymology , Up-Regulation/drug effects
9.
Nucleic Acids Res ; 36(Database issue): D426-33, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18073189

ABSTRACT

The Worldwide Protein Data Bank (wwPDB; wwpdb.org) is the international collaboration that manages the deposition, processing and distribution of the PDB archive. The online PDB archive at ftp://ftp.wwpdb.org is the repository for the coordinates and related information for more than 47 000 structures, including proteins, nucleic acids and large macromolecular complexes that have been determined using X-ray crystallography, NMR and electron microscopy techniques. The members of the wwPDB-RCSB PDB (USA), MSD-EBI (Europe), PDBj (Japan) and BMRB (USA)-have remediated this archive to address inconsistencies that have been introduced over the years. The scope and methods used in this project are presented.


Subject(s)
Databases, Protein , Macromolecular Substances/chemistry , Archives , Crystallography, X-Ray , Databases, Protein/standards , Dictionaries, Chemical as Topic , Internet , Microscopy, Electron , Nuclear Magnetic Resonance, Biomolecular , Nucleic Acids/chemistry , Proteins/chemistry , Reproducibility of Results , Terminology as Topic
10.
J Biol Chem ; 281(41): 30542-50, 2006 Oct 13.
Article in English | MEDLINE | ID: mdl-16895901

ABSTRACT

Annexin II is a heterotetramer, consisting of two 11-kDa (p11) and two 36-kDa (p36) subunits, that is produced by osteoclasts and stimulates osteoclast formation. However, its receptor is unknown. We showed that annexin II binds to normal primary human marrow stromal cells and the Paget's marrow-derived PSV10 stromal cell line to induce osteoclast formation. 125I-Labeled annexin II binding assays with PSV10 cells demonstrated that there was a single class of annexin II receptors with a Kd of 5.79 nm and Bmax of 2.13 x 10(5) receptors/cell. Annexin III or annexin V did not bind this receptor. Using 125I-labeled annexin II binding to screen NIH3T3 transfected with a human marrow cDNA expression library, we identified a putative annexin II receptor clone, which encoded a novel 26-kDa type I membrane receptor protein when expressed in HEK 293 cells. HEK 293 cells transformed with the cloned annexin II receptor cDNA showed a similar binding affinity to annexin II as that observed in PSV10 cells. Chemical cross-linking experiments with biotinylated annexin II and intact PSV10 cells identified a 55-kDa band on Western blot analysis that reacted with both an anti-p11 antibody and streptavidin but not anti-p36 antibody. A rabbit polyclonal antibody raised against the putative recombinant annexin II receptor also recognized the same 26-kDa protein band detected in PSV10 cells. Importantly, the annexin II receptor antibody dose-dependently blocked the stimulatory effects of annexin II on human osteoclast formation, demonstrating that the receptor mediates the effects of annexin II on osteoclast formation.


Subject(s)
Annexin A2/chemistry , Bone Marrow Cells/metabolism , Receptors, Peptide/chemistry , Receptors, Peptide/genetics , Stromal Cells/metabolism , Animals , Cloning, Molecular , Cross-Linking Reagents/pharmacology , Humans , Kinetics , Mice , Molecular Sequence Data , NIH 3T3 Cells , Osteoclasts/metabolism
11.
J Med Chem ; 48(20): 6386-92, 2005 Oct 06.
Article in English | MEDLINE | ID: mdl-16190764

ABSTRACT

A series of novel aminoalkylindoles was synthesized in an effort to develop compounds that are potent agonists at the CB1 cannabinoid receptor and that are also easily labeled with radioisotopes of iodine for biochemical and imaging studies. 2-Iodophenyl-[1-(1-methylpiperidin-2-ylmethyl)-1H-indol-3-yl]methanone (8, AM2233) had a very high affinity for the rat CB1 receptor, with most of the affinity residing with the (R)-enantiomer. Radioiodinated 8, (R)-8, and (S)-8 were prepared by radioiododestannylation of the tributyltin analogues in high yields, radiochemical purities, and specific radioactivities. In a mouse hippocampal membrane preparation with [131I](R)-8 as radioligand, racemic 8 exhibited a K(i) value of 0.2 nM compared with 1.6 nM for WIN55212-2. In autoradiographic experiments with mouse brain sections, the distribution of radioiodinated 8 was consistent with that of brain CB1 receptors. Again, very little specific binding was seen with the (S)-enantiomer [131I](S)-8 and none occurred with the (R)-enantiomer [131I](R)-8 in sections from CB1 receptor knockout mice. Radioiodinated 8 thus appears to be a suitable radioligand for studies of CB1 cannabinoid receptors.


Subject(s)
Brain/metabolism , Indoles/chemical synthesis , Piperidines/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Receptor, Cannabinoid, CB1/metabolism , Animals , Autoradiography , Crystallography, X-Ray , Hippocampus/metabolism , In Vitro Techniques , Indoles/chemistry , Indoles/pharmacokinetics , Iodine Radioisotopes , Ligands , Mice , Mice, Knockout , Piperidines/chemistry , Piperidines/pharmacokinetics , Radioligand Assay , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Rats , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/metabolism , Spleen/metabolism , Stereoisomerism , Structure-Activity Relationship
12.
J Am Chem Soc ; 127(15): 5388-95, 2005 Apr 20.
Article in English | MEDLINE | ID: mdl-15826177

ABSTRACT

Here, we explore the chemistry of the previously undocumented E form of diazeniumdiolates having the structure R(1)R(2)NN(O)=NOR(3). Reported crystallographic studies have uniformly revealed the Z configuration, and our attempts to observe a Z --> E conversion through thermal equilibration or photochemical means have, until now, consistently failed to reveal a significant amount of a second conformer. As a typical example, the NMR spectrum of trimethyl derivative Me(2)NN(O)=NOMe revealed no evidence for a second configuration. Electronic structure calculations attribute this finding to a prohibitively high interconversion barrier of approximately 40 kcal/mol. A similar result was obtained when we considered the case of R(1) = Me = R(3) and R(2) = H at the same levels of theory. However, when MeHNN(O)=NOMe was ionized by dissociating the N-H bond, the barrier was calculated to be lower by approximately 20 kcal/mol, with the E form of the anion being favored over Z. This circumstance suggested that an E isomer might be isolable if a Z anion were formed and given sufficient time to assume the E configuration, then quenched by reaction with an electrophile to trap and neutralize the E form and restore the putatively high interconversion barrier. Consistent with this prediction, basifying iPrHNN(O)=NOCH(2)CH(2)Br rapidly led to a six-membered heterocycle that was crystallographically characterized as containing the -N(O)=NO- functional group in the E configuration. The results suggest an approach for generating pairs of Z and E diazeniumdiolates for systematic comparison of the rates at which the individual isomers release bioactive NO and of other physicochemical determinants of their biomedical utility.


Subject(s)
Azo Compounds/chemistry , Crystallography, X-Ray , Isomerism , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Structure , Thermodynamics
13.
Nucleic Acids Res ; 33(Database issue): D233-7, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15608185

ABSTRACT

The Protein Data Bank (PDB) is the central worldwide repository for three-dimensional (3D) structure data of biological macromolecules. The Research Collaboratory for Structural Bioinformatics (RCSB) has completely redesigned its resource for the distribution and query of 3D structure data. The re-engineered site is currently in public beta test at http://pdbbeta.rcsb.org. The new site expands the functionality of the existing site by providing structure data in greater detail and uniformity, improved query and enhanced analysis tools. A new key feature is the integration and searchability of data from over 20 other sources covering genomic, proteomic and disease relationships. The current capabilities of the re-engineered site, which will become the RCSB production site at http://www.pdb.org in late 2005, are described.


Subject(s)
Databases, Protein , Models, Molecular , Protein Conformation , Software , Systems Integration , User-Computer Interface
14.
J Org Chem ; 69(16): 5322-7, 2004 Aug 06.
Article in English | MEDLINE | ID: mdl-15287777

ABSTRACT

The synthesis of the ortho- and para-e isomers in the oxide-bridged 5-phenylmorphan series of rigid tetracyclic compounds was accomplished via rac-5-(2-fluoro-5-nitrophenyl)-2-methyl-2-azabicyclo[3.3.1]nonan-9beta-ol ((+/-)-10), an intermediate containing an aromatic nitro-activated fluorine atom. The fluorine atom was used as the leaving group for the formation of the strained tetracyclic trans-fused 5,6-ring system in rac-(1alpha,4aalpha,9aalpha)-1,3,4,9a-tetrahydro-2-methyl-6-nitro-2H-1,4a-propanobenzofuro[2,3-c]pyridine ((+/-)-11), although preference for cis ring fusion during the formation of tricyclic tetra- and hexahydrodibenzofurans has been well-documented. Single-crystal X-ray crystallographic study of the desired para-e isomer ((+/-)-2), as well as of two intermediates in its synthesis, provided assurance of the correct structures. The e-isomers are among the last of the 12 oxide-bridged 5-phenylmorphans to be synthesized. We envisioned the syntheses of these rigid, tetracyclic compounds in order to determine the three-dimensional pattern of a ligand that would enable interaction with opioid receptors as agonists or antagonists.


Subject(s)
Bridged-Ring Compounds , Molecular Probes/chemical synthesis , Morphinans/chemical synthesis , Receptors, Opioid/metabolism , Bridged-Ring Compounds/chemistry , Crystallography, X-Ray , Fluorine/chemistry , Molecular Structure , Stereoisomerism
15.
J Med Chem ; 47(10): 2624-34, 2004 May 06.
Article in English | MEDLINE | ID: mdl-15115403

ABSTRACT

In our efforts toward developing a nonselective ligand that would block the effects of stimulants such as methamphetamine at dopamine (DA), serotonin (5-HT), and norepinephrine (NE) transporters, we synthesized a series of 3-(3,4-dichlorophenyl)-1-indanamine derivatives. Two of the examined higher affinity compounds had a phenolic hydroxyl group enabling preparation of a medium to long chain carboxylic acid ester that might eventually be useful for a long-acting depot formulation. The in vitro data indicated that (-)-(1R,3S)-trans-3-(3,4-dichlorophenyl)-6-hydroxy-N-methyl-1-indanamine ((-)-(1R,3S)-11) displays high-affinity binding and potent inhibition of uptake at all three biogenic amine transporters. In vivo microdialysis experiments demonstrated that intravenous administration of (-)-(1R,3S)-11 to rats elevated extracellular DA and 5-HT in the nucleus accumbens in a dose-dependent manner. Pretreating rats with 0.5 mg/kg (-)-(1R,3S)-11 elevated extracellular DA and 5-HT by approximately 150% and reduced methamphetamine-induced neurotransmitter release by about 50%. Ex vivo autoradiography, however, demonstrated that iv administration of (-)-(1R,3S)-11 produced a dose-dependent, persistent occupation of 5-HT transporter binding sites but not DA transporter sites.


Subject(s)
Biogenic Amines/metabolism , Indans/chemical synthesis , Membrane Transport Proteins/metabolism , Animals , Autoradiography , Carrier Proteins/metabolism , Crystallography, X-Ray , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins , Extracellular Fluid/metabolism , In Vitro Techniques , Indans/chemistry , Indans/pharmacology , Ligands , Membrane Glycoproteins/metabolism , Molecular Structure , Nerve Tissue Proteins/metabolism , Norepinephrine Plasma Membrane Transport Proteins , Nucleus Accumbens/metabolism , Rats , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins , Stereoisomerism , Structure-Activity Relationship , Symporters/metabolism
16.
Org Biomol Chem ; 2(3): 330-6, 2004 Feb 07.
Article in English | MEDLINE | ID: mdl-14747861

ABSTRACT

In an attempt to obtain the para-f isomer, rac-(1R,4aR,9aR)-2-methyl-1,3,4,9a-tetrahydro-2H-1,4a-propanobenzofuro[2,3-c]pyridin-6-ol, via mesylation of an intermediate 9[small alpha]-hydroxyphenylmorphan, we obtained, instead, a rearranged chloro compound with a 5-membered nitrogen ring, 7-chloro-3a-(2,5-dimethoxyphenyl)-1-methyl-octahydroindole. This indole underwent a second rearrangement to give us the desired para-f isomer. The structures of the intermediate indole and the final product were unequivocally established by X-ray crystallography. A resynthesis of the known rac-(1R,4aR,9aR)-2-methyl-1,3,4,9a-tetrahydro-2H-1,4a-propanobenzofuro[2,3-c]pyridin-8-ol, the ortho-f isomer, was achieved using the reaction conditions for the para-f isomer, as well as under Mitsunobu reaction conditions where, unusually, the oxide-bridge ring in the 5-phenylmorphan was closed to obtain the desired product. The synthesis of the para-f isomer adds an additional compound to those oxide-bridged phenylmorphans that were initially visualized and synthesized; the establishment of the structure and configuration of 8 of the theoretically possible 12 racemates has now been achieved. The X-ray crystallographic structure analysis of the para-f isomer provides essential data that will be needed to establish the configuration of a ligand necessary to interact with an opioid receptor.


Subject(s)
Benzofurans/chemistry , Benzofurans/chemical synthesis , Morphinans/chemistry , Morphinans/chemical synthesis , Oxides/chemistry , Pyridines/chemistry , Pyridines/chemical synthesis , Crystallography, X-Ray , Isomerism , Molecular Conformation , Molecular Structure
17.
Bioorg Med Chem ; 11(22): 4761-8, 2003 Nov 03.
Article in English | MEDLINE | ID: mdl-14556791

ABSTRACT

We have explored the synthesis of compounds that have good affinity for both mu- and delta-opioid receptors from the (alphaR,2S,5S) class of diaryldimethylpiperazines. These non-selective compounds were related to opioids that have been found to interact selectively with mu- or delta-opioid receptors as agonists or antagonists. In our initial survey, we found two compounds, (+)-4-[(alphaR)-alpha-(4-allyl-(2S,5S)-dimethylpiperazin-1-yl)-(3-hydroxyphenyl)methyl]-N-ethyl-N-phenylbenzamide (14) and its N-H relative, (-)-4-[(alphaR)-alpha-(2S,5S)-dimethylpiperazin-1-yl)-(3-hydroxyphenyl)methyl]-N-ethyl-N-phenylbenzamide (15), that interacted with delta-receptors with good affinity, and, as we hoped, with much higher affinity at mu-receptors than SNC80. The relative configuration of the benzylic position in (+)-4-[(alphaR)-alpha-(4-allyl-(2S,5S)-dimethyl-1-piperazinyl)-(3-methoxyphenyl)methyl]-benzyl alcohol (10) was determined by X-ray crystallographic analysis of a crystal that was an unresolved twin. The absolute stereochemistry of that benzylic stereogenic center was unequivocally derived by the X-ray crystallographic analysis from the two other centers of asymmetry in the molecule that were known. Those were established from the synthesis via a dipeptide cyclo-L-Ala-L-Ala in which the absolute stereochemistry was established.


Subject(s)
Benzamides/chemical synthesis , Benzamides/metabolism , Piperazines/chemical synthesis , Piperazines/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Animals , Benzamides/chemistry , Benzamides/pharmacology , Brain/metabolism , Cell Membrane/metabolism , Crystallography, X-Ray , Guinea Pigs , Ligands , Molecular Conformation , Molecular Structure , Piperazines/chemistry , Piperazines/pharmacology , Radioligand Assay , Rats , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Stereoisomerism , Structure-Activity Relationship
18.
J Org Chem ; 68(5): 1929-32, 2003 Mar 07.
Article in English | MEDLINE | ID: mdl-12608812

ABSTRACT

Treatment of 5-trimethylsilylthebaine with L-Selectride gave rise to a rearrangement to 10-trimethylsilylbractazonine through migration of the phenyl group, whereas treatment of thebaine with strong Lewis acids is known to lead to a similar rearrangement through migration of the alkyl bridge to give, after reduction, (+)-neodihydrothebaine. It is suggested that the rearrangement of the alkyl group of thebaine is favored due to the formation of a tertiary benzylic cation. However, for 5-trimethylsilylthebaine, the lithium ion of L-Selectride acts as the Lewis acid and the beta-silyl effect dominates in the stabilization of any positive charge. This rearrangement provides a clear example of the greater relative migratory aptitude of phenyl groups over alkyl groups, and provides an efficient synthesis of (+)-bractazonine from thebaine.


Subject(s)
Alkaloids , Thebaine , Thebaine/chemical synthesis , Alkaloids/chemical synthesis , Alkaloids/chemistry , Catalysis , Combinatorial Chemistry Techniques , Indicators and Reagents , Magnetic Resonance Spectroscopy , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Stereoisomerism , Thebaine/analogs & derivatives , Thebaine/chemistry
19.
J Org Chem ; 68(5): 2010-3, 2003 Mar 07.
Article in English | MEDLINE | ID: mdl-12608825

ABSTRACT

A practical method for the conversion of tetrahydrothebaine to dihydromorphine in 92% yield is described. The procedure should allow more efficient production of opium products and may be easily modified for large-scale synthesis. The conversion of codeine to (8S)-8-bromomorphide, a potentially valuable intermediate to 6-demethoxyoripavine and derivatives, is also described. The absolute configuration of (8S)-8-bromomorphide was determined by a single-crystal X-ray diffraction study of the hydrobromide salt.


Subject(s)
Dihydromorphine/chemical synthesis , Morphine Derivatives/chemical synthesis , Catalysis , Chromatography, Thin Layer , Crystallography, X-Ray , Dihydromorphine/analysis , Indicators and Reagents , Molecular Structure , Morphine Derivatives/analysis , Stereoisomerism , Thebaine/analogs & derivatives , Thebaine/chemistry
20.
Bioorg Med Chem ; 11(6): 1123-36, 2003 Mar 20.
Article in English | MEDLINE | ID: mdl-12614900

ABSTRACT

There is considerable interest in developing dopamine transporter (DAT) inhibitors as potential therapies for the treatment of cocaine abuse. We report herein our pharmacophore-based discovery and molecular modeling-assisted rational design of 2,3-disubstituted quinuclidines as potent DAT inhibitors with a novel chemical scaffold. Through 3-D-database pharmacophore searching, compound 12 was identified as a very weak DAT inhibitor with K(i) values of 7.3 and 8.9 microM in [3H]mazindol binding and in inhibition of dopamine reuptake, respectively. Molecular modeling-assisted rational design and chemical modifications led to identification of potent analogues (-)-29 and 34 with K(i) values of 14 and 32 nM for both compounds in binding affinity and inhibition of dopamine reuptake, respectively. Behavioral pharmacological evaluations in rodents showed that 34 has a profile very different from cocaine. While 34 is substantially more potent than cocaine as a DAT inhibitor, it is approximately four times less potent than cocaine in mimicking the discriminative stimulus properties of cocaine in rat. On the other hand, 34 (3-30 mg/kg) lacks either the locomotor stimulant or stereotypic properties of cocaine in mice. Importantly, 34 blocks locomotor stimulant activity induced by 20 mg/kg cocaine in mice, with an estimated ED(50) of 19 mg/kg. Taken together, our data suggest that 34 represents a class of potent DAT inhibitors with a novel chemical scaffold and a behavioral pharmacological profile different from that of cocaine in rodents. Thus, 34 may serve as a novel lead compound in the ultimate development of therapeutic entities for cocaine abuse and/or addiction.


Subject(s)
Membrane Glycoproteins , Membrane Transport Modulators , Membrane Transport Proteins/antagonists & inhibitors , Nerve Tissue Proteins , Quinuclidines/chemical synthesis , Quinuclidines/pharmacology , Animals , Behavior, Animal/drug effects , Binding, Competitive/drug effects , Cocaine/pharmacology , Discrimination, Psychological/drug effects , Dopamine Plasma Membrane Transport Proteins , Dopamine Uptake Inhibitors/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Interactions , Indicators and Reagents , Magnetic Resonance Spectroscopy , Mazindol/metabolism , Mice , Models, Molecular , Motor Activity/drug effects , Protein Binding , Protein Conformation , Rats , Structure-Activity Relationship , Synaptosomes/drug effects , Synaptosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...