Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 130(1): 152-163, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27199371

ABSTRACT

Although it is known that a stiffening of the stroma and the rearrangement of collagen fibers within the extracellular matrix facilitate the movement of tumor cells away from the primary lesion, the underlying mechanisms responsible are not fully understood. We now show that this invasion, which can be initiated by applying tensional loads to a three-dimensional collagen gel matrix in culture, is dependent on the Rap1 GTPases (Rap1a and Rap1b, referred to collectively as Rap1). Under these conditions Rap1 activity stimulates the formation of focal adhesion structures that align with the tensional axis as single tumor cells move into the matrix. These effects are mediated by the ability of Rap1 to induce the polarized polymerization and retrograde flow of actin, which stabilizes integrins and recruits vinculin to preformed adhesions, particularly those near the leading edge of invasive cells. Rap1 activity also contributes to the tension-induced collective invasive elongation of tumor cell clusters and it enhances tumor cell growth in vivo Thus, Rap1 mediates the effects of increased extracellular tension in multiple ways that are capable of contributing to tumor progression when dysregulated.


Subject(s)
Stress, Mechanical , rap1 GTP-Binding Proteins/metabolism , Actins/metabolism , Animals , Biomechanical Phenomena , Cell Aggregation , Cell Line, Tumor , Cell Proliferation , Collagen/metabolism , Crk-Associated Substrate Protein/metabolism , Extracellular Matrix/metabolism , Focal Adhesions/metabolism , Gels , Guanosine Triphosphate/metabolism , Humans , Integrins/metabolism , Intercellular Junctions/metabolism , Mice , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphorylation , Polymerization , Protein Stability , Pseudopodia/metabolism , Signal Transduction , Vinculin/metabolism , rac1 GTP-Binding Protein/metabolism
2.
Dis Model Mech ; 9(9): 1039-49, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27519690

ABSTRACT

A hallmark of all primary and metastatic tumours is their high rate of glucose uptake and glycolysis. A consequence of the glycolytic phenotype is the accumulation of metabolic acid; hence, tumour cells experience considerable intracellular acid stress. To compensate, tumour cells upregulate acid pumps, which expel the metabolic acid into the surrounding tumour environment, resulting in alkalization of intracellular pH and acidification of the tumour microenvironment. Nevertheless, we have only a limited understanding of the consequences of altered intracellular pH on cell physiology, or of the genes and pathways that respond to metabolic acid stress. We have used yeast as a genetic model for metabolic acid stress with the rationale that the metabolic changes that occur in cancer that lead to intracellular acid stress are likely fundamental. Using a quantitative systems biology approach we identified 129 genes required for optimal growth under conditions of metabolic acid stress. We identified six highly conserved protein complexes with functions related to oxidative phosphorylation (mitochondrial respiratory chain complex III and IV), mitochondrial tRNA biosynthesis [glutamyl-tRNA(Gln) amidotransferase complex], histone methylation (Set1C-COMPASS), lysosome biogenesis (AP-3 adapter complex), and mRNA processing and P-body formation (PAN complex). We tested roles for two of these, AP-3 adapter complex and PAN deadenylase complex, in resistance to acid stress using a myeloid leukaemia-derived human cell line that we determined to be acid stress resistant. Loss of either complex inhibited growth of Hap1 cells at neutral pH and caused sensitivity to acid stress, indicating that AP-3 and PAN complexes are promising new targets in the treatment of cancer. Additionally, our data suggests that tumours may be genetically sensitized to acid stress and hence susceptible to acid stress-directed therapies, as many tumours accumulate mutations in mitochondrial respiratory chain complexes required for their proliferation.


Subject(s)
Genes, Fungal , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/therapy , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Stress, Physiological/genetics , Cell Line, Tumor , Cell Proliferation , Gene Knockout Techniques , Genetic Testing , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Protein Subunits/metabolism , Vacuolar Proton-Translocating ATPases/metabolism
3.
Breast Cancer Res ; 18(1): 11, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26796961

ABSTRACT

BACKGROUND: Overexpression of the transmembrane sialomucin podocalyxin, which is known to play a role in lumen formation during polarized epithelial morphogenesis, is an independent indicator of poor prognosis in a number of epithelial cancers, including those that arise in the breast. Therefore, we set out to determine if podocalyxin plays a functional role in breast tumor progression. METHODS: MCF-7 breast cancer cells, which express little endogenous podocalyxin, were stably transfected with wild type podocalyxin for forced overexpression. 4T1 mammary tumor cells, which express considerable endogenous podocalyxin, were retrovirally transduced with a short hairpin ribonucleic acid (shRNA) targeting podocalyxin for stable knockdown. In vitro, the effects of podocalyxin on collective cellular migration and invasion were assessed in two-dimensional monolayer and three-dimensional basement membrane/collagen gel culture, respectively. In vivo, local invasion was assessed after orthotopic transplantation in immunocompromised mice. RESULTS: Forced overexpression of podocalyxin caused cohesive clusters of epithelial MCF-7 breast tumor cells to bud off from the primary tumor and collectively invade the stroma of the mouse mammary gland in vivo. This budding was not associated with any obvious changes in histoarchitecture, matrix deposition or proliferation in the primary tumour. In vitro, podocalyxin overexpression induced a collective migration of MCF-7 tumor cells in two-dimensional (2-D) monolayer culture that was dependent on the activity of the actin scaffolding protein ezrin, a cytoplasmic binding partner of podocalyxin. In three-dimensional (3-D) culture, podocalyxin overexpression induced a collective budding and invasion that was dependent on actomyosin contractility. Interestingly, the collectively invasive cell aggregates often contained expanded microlumens that were also observed in vivo. Conversely, when endogenous podocalyxin was removed from highly metastatic, but cohesive, 4T1 mammary tumor cells there was a decrease in collective invasion in three-dimensional culture. CONCLUSIONS: Podocalyxin is a tumor cell-intrinsic regulator of experimental collective tumor cell invasion and tumor budding.


Subject(s)
Breast Neoplasms/genetics , Cell Movement/genetics , Neoplasm Invasiveness/genetics , Sialoglycoproteins/biosynthesis , Animals , Breast Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , MCF-7 Cells , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mice , Sialoglycoproteins/genetics
4.
Stem Cells ; 31(7): 1434-45, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23592435

ABSTRACT

The incidence of refractory acute myeloid leukemia (AML) is on the increase due in part to an aging population that fails to respond to traditional therapies. High throughput genomic analysis promises better diagnosis, prognosis, and therapeutic intervention based on improved patient stratification. Relevant preclinical models are urgently required to advance drug development in this area. The collaborating oncogenes, HOXA9 and MEIS1, are frequently co-overexpressed in cytogenetically normal AML (CN-AML), and a conditional transplantation mouse model was developed that demonstrated oncogene dependency and expression levels comparable to CN-AML patients. Integration of gene signatures obtained from the mouse model and a cohort of CN-AML patients using statistically significant connectivity map analysis identified Entinostat as a drug with the potential to alter the leukemic condition toward the normal state. Ex vivo treatment of leukemic cells, but not age-matched normal bone marrow controls, with Entinostat validated the gene signature and resulted in reduced viability in liquid culture, impaired colony formation, and loss of the leukemia initiating cell. Furthermore, in vivo treatment with Entinostat resulted in prolonged survival of leukemic mice. This study demonstrates that the HDAC inhibitor Entinostat inhibits disease maintenance and prolongs survival in a clinically relevant murine model of cytogenetically normal AML.


Subject(s)
Benzamides/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Pyridines/pharmacology , Animals , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Immunophenotyping , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL
5.
Mol Cancer Res ; 11(5): 530-40, 2013 May.
Article in English | MEDLINE | ID: mdl-23399642

ABSTRACT

During metastatic progression, an aberrant epithelial-to-mesenchymal transformation (EMT) that is most often driven by the loss of the cell-cell adhesion molecule E-cadherin generates noncohesive tumor cells that are highly invasive. We used mesenchymally transformed, E-cadherin-negative MDA-MB-231 breast carcinoma cells in a natural product screen and determined that the triterpenoid saponin sarasinoside A1 inhibited their invasion and the invasion of a number of other tumor cell lines. Sarasinoside A1 also caused MDA-MB-231 cells to become cohesive in a three-dimensional basement membrane and collagen gel cultures. In two-dimensional culture, sarasinoside A1 initiated a morphologic re-epithelialization of MDA-MB-231 cells wherein preexisting nonepithelial cadherins and the junction-associated proteins ß-catenin and ZO-1 all relocalized to sites of cell-cell contact. In addition, the intercellular space between neighboring cells narrowed considerably, the stability of polymerized actin at cell-cell contact sites increased, and there was a recruitment and stabilization of nectin-based adhesion complexes to these sites, all of which strongly suggested that functional cell-cell junctions had formed. Importantly, sarasinoside A1 induced nascent cell-cell junction formation that did not require changes in gene expression and was not associated with an induction of E-cadherin but resulted in increased activation of Rap GTPases. Therefore, our findings with sarasinoside A1 suggest that it may be possible to re-epithelialize metastatic tumor cells with phenotypic consequence even when E-cadherin is completely absent.


Subject(s)
Breast Neoplasms/drug therapy , Cadherins/metabolism , Epithelial-Mesenchymal Transition/drug effects , Glycosides/pharmacology , Triterpenes/pharmacology , Actins/metabolism , Breast Neoplasms/pathology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic , Female , Glycosides/chemistry , Glycosides/isolation & purification , Humans , Intercellular Junctions/drug effects , Neoplasm Invasiveness , Triterpenes/chemistry , Triterpenes/isolation & purification , Tumor Cells, Cultured
6.
Org Biomol Chem ; 11(9): 1476-81, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23334605

ABSTRACT

The tricyclic peptides neopetrosiamides A and B, isolated from the marine sponge Neopetrosia sp., are potential antimetastatic agents that inhibit tumour cell invasion by both amoeboid and mesenchymal migration pathways. They differ in the stereochemistry of the methionine sulfoxide at position 24. Our previously reported syntheses using an orthogonal sulfur protection strategy established the critical connectivity of the three disulfide bonds. In this report, fifteen analogues of neopetrosiamide A and B, six which replace selected disulfide bonds and nine which replace the diastereomeric methionine sulfoxide, have been prepared using Fmoc solid-phase peptide chemistry. Disulfide replacement analogues were shown to lose activity, and only one of the methionine sulfoxide analogues retained full bioactivity in morphological studies.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasm Invasiveness/prevention & control , Peptides, Cyclic/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Neoplasm Invasiveness/pathology , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/chemistry , Structure-Activity Relationship
7.
Exp Cell Res ; 319(6): 908-17, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23333560

ABSTRACT

Individual tumor cells utilize one of two modes of motility to invade the extracellular matrix, mesenchymal or amoeboid. We have determined that the diterpenoid genkwanine M (GENK) enhances the mesenchymal mode of cell motility that is intrinsic to HT-1080 osteosarcoma cells, stimulates a mesenchymal mode of motility in stationary MDA-MB-453 breast carcinoma cells, and induces a shift to a mesenchymal mode of cell motility in LS174T colorectal adenocarcinoma cells that normally utilize the alternate amoeboid mode of motility. The ability of GENK to stimulate or induce mesenchymal motility was preceded by a rapid cell spreading, elongation and polarization that did not require new gene expression. However, these initial morphologic changes were integrin dependent and they were associated with a reorganization of focal contacts and focal adhesions as well as an activation of the focal adhesion kinase. Therefore, GENK induces a mesenchymal mode of cell motility in a wide variety of tumor cell types that may be mediated, at least in part, by an activation of integrin-associated signaling.


Subject(s)
Cell Movement/drug effects , Cell Shape/drug effects , Flavones/pharmacology , Cell Enlargement/drug effects , Cell Line, Tumor , Cell Polarity , Flavones/chemistry , Focal Adhesions , Humans , Integrin alpha Chains/chemistry , Integrin beta Chains/chemistry , Neoplasm Invasiveness/pathology , Osteosarcoma/chemistry , Osteosarcoma/pathology , Protein Biosynthesis/drug effects , Transcription, Genetic , Wikstroemia/chemistry
8.
Oncol Rep ; 27(2): 311-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22076128

ABSTRACT

Na+/H+ exchangers (NHEs) are a group of secondary active antiporters that regulate cellular pH, cell volume and ion homeostasis. In humans, nine isoforms (NHE1-NHE9) were identified and characterized as functional NHEs. While a growing body of evidence indicates that NHE1 generates an acidic tumor environment and thereby contributes to tumor invasion, little is known about the role of other NHE isoforms in tumor progression. NHE7 is a unique member of the NHE gene family that dynamically shuttles between the trans-Golgi network, endosomes and the plasma membrane, and regulates the luminal pH of these organelles. Here we show that NHE7-overexpression in breast cancer MDA-MB-231 cells enhances cell overlay, cell-cell adhesion, invasion, anchorage-independent tumor growth and tumor formation in vivo. In contrast, NHE1-overexpression enhances tumor invasion, but it has little effect on cell adhesion or anchorage-independent tumor growth. Pathological examinations of the tumor samples derived from NHE7-overexpressing cells showed a similar appearance to aggressive tumors. Together, these results suggest that NHE7 enhances tumor progression. This is the first report to show the involvement of an organellar NHE in oncogenic processes.


Subject(s)
Breast Neoplasms/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Adhesion/genetics , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Neoplasm Invasiveness/genetics , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/genetics , Xenograft Model Antitumor Assays
9.
Blood ; 117(2): e27-38, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-20980679

ABSTRACT

The molecular mechanisms regulating self-renewal of leukemia stem cells remain poorly understood. Here we report the generation of 2 closely related leukemias created through the retroviral overexpression of Meis1 and Hoxa9. Despite their apparent common origin, these clonal leukemias exhibit enormous differences in stem cell frequency (from 1 in 1.4, FLA2; to 1 in 347, FLB1), suggesting that one of these leukemias undergoes nearly unlimited self-renewal divisions. Using next-generation RNA-sequencing, we characterized the transcriptomes of these phenotypically similar, but biologically distinct, leukemias, identifying hundreds of differentially expressed genes and a large number of structural differences (eg, alternative splicing and promoter usage). Focusing on ligand-receptor pairs, we observed high expression levels of Sdf1-Cxcr4; Jagged2-Notch2/1; Osm-Gp130; Scf-cKit; and Bmp15-Tgfb1/2. Interestingly, the integrin beta 2-like gene (Itgb2l) is both highly expressed and differentially expressed between our 2 leukemias (∼ 14-fold higher in FLA2 than FLB1). In addition, gene ontology analysis indicated G-protein-coupled receptor had a much higher proportion of differential expression (22%) compared with other classes (∼ 5%), suggesting a potential role regulating subtle changes in cellular behavior. These results provide the first comprehensive transcriptome analysis of a leukemia stem cell and document an unexpected level of transcriptome variation between phenotypically similar leukemic cells.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Neoplastic Stem Cells , Animals , Clone Cells , Flow Cytometry , Genetic Vectors , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Microarray Analysis , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/genetics , Retroviridae , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA
10.
PLoS One ; 5(5): e10836, 2010 May 26.
Article in English | MEDLINE | ID: mdl-20520768

ABSTRACT

BACKGROUND: Neopetrosiamide A (NeoA) is a 28-amino acid tricyclic peptide originally isolated from a marine sponge as a tumor cell invasion inhibitor whose mechanism of action is unknown. METHODOLOGY/PRINCIPAL FINDINGS: We show that NeoA reversibly inhibits tumor cell adhesion, disassembles focal adhesions in pre-attached cells, and decreases the level of beta1 integrin subunits on the cell surface. NeoA also induces the formation of dynamic, membrane-bound protrusions on the surface of treated cells and the release of membrane-bound vesicles into the culture medium. Proteomic analysis indicates that the vesicles contain EGF and transferrin receptors as well as a number of proteins involved in adhesion and migration including: beta1 integrin and numerous alpha integrin subunits; actin and actin-binding proteins such as cofilin, moesin and myosin 1C; and membrane modulating eps15 homology domain (EHD) proteins. Surface labeling, trafficking inhibition, and real-time imaging experiments all suggest that beta1 integrin-containing vesicles are released directly from NeoA-induced cell surface protrusions rather than from vesicles generated intracellularly. The biological activity of NeoA is dependent on its disulfide bond pattern and NMR spectroscopy indicates that the peptide is globular with a continuous ridge of hydrophobic groups flanked by charged amino acid residues that could facilitate a simultaneous interaction with lipids and proteins in the membrane. CONCLUSIONS/SIGNIFICANCE: NeoA is an anti-adhesive peptide that decreases cell surface integrin levels through a novel, yet to be elucidated, mechanism that involves the release of adhesion molecule-containing vesicles from the cell surface.


Subject(s)
Cell Membrane/drug effects , Cell Membrane/metabolism , Cytoplasmic Vesicles/drug effects , Cytoplasmic Vesicles/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Peptides, Cyclic/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Surface Extensions/drug effects , Cell Surface Extensions/ultrastructure , Focal Adhesions/drug effects , Focal Adhesions/metabolism , Humans , Integrin beta1/metabolism , Membrane Proteins/metabolism , Neoplasm Invasiveness , Peptides, Cyclic/chemistry , Protein Subunits/metabolism , Temperature
11.
Cancer Res ; 70(11): 4590-601, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20484042

ABSTRACT

The Rap1 GTPase is a master regulator of cell adhesion, polarity, and migration. We show that both blocking Rap1 activation and expressing a constitutively active form of Rap1 reduced the ability of B16F1 melanoma cells to extravasate from the microvasculature and form metastatic lesions in the lungs. This correlated with a decreased ability of the tumor cells to undergo transendothelial migration (TEM) in vitro and form dynamic, F-actin-rich pseudopodia that penetrate capillary endothelial walls in vivo. Using multiple tumor cell lines, we show that the inability to form these membrane protrusions, which likely promote TEM and extravasation, can be explained by altered adhesion dynamics and impaired cell polarization that result when Rap1 activation or cycling is perturbed. Thus, targeting Rap1 could be a useful approach for reducing the metastatic dissemination of tumor cells that undergo active TEM.


Subject(s)
Lung Neoplasms/enzymology , Lung Neoplasms/secondary , Melanoma, Experimental/enzymology , Melanoma, Experimental/secondary , rap1 GTP-Binding Proteins/metabolism , Animals , Cell Adhesion/physiology , Cell Communication/physiology , Cytoskeleton/enzymology , Cytoskeleton/pathology , Endothelial Cells/cytology , Endothelial Cells/enzymology , Enzyme Activation , Focal Adhesions/enzymology , Focal Adhesions/pathology , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/prevention & control , Melanoma, Experimental/blood supply , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL
12.
J Am Chem Soc ; 132(5): 1486-7, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-20073461

ABSTRACT

Neopetrosiamides A and B (2) from the marine sponge Neopetrosia sp. are two diastereomeric tricyclic peptides that inhibit tumor cell invasion associated with metastasis. The reported structures were chemically synthesized using solid-phase peptide synthesis and sequential stepwise disulfide bond formation in solution. The disulfide bond connectivity of the originally proposed structures was revised and confirmed by chemical synthesis together with a combination of HPLC analysis, disulfide mapping, and biological activity testing. This methodology was also utilized to generate analogues containing methionine or norleucine in place of the methionine sulfoxide at position 24. Compounds 4 and 6 demonstrated potent bioactivity comparable to that of the parent peptides.


Subject(s)
Peptides, Cyclic/chemical synthesis , Porifera/chemistry , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Movement/drug effects , Disulfides/chemistry , Humans , Molecular Sequence Data , Neoplasms/drug therapy , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology
13.
J Am Chem Soc ; 130(36): 12045-55, 2008 Sep 10.
Article in English | MEDLINE | ID: mdl-18700764

ABSTRACT

The use of a boronic ester as a captor of aqueous [(18)F]-fluoride has been previously suggested as a means of labeling biomolecules in one step for positron emission tomography (PET) imaging. For this approach to be seriously considered, the [(18)F]-labeled trifluoroborate should be humorally stable such that it neither leaches free [(18)F]-fluoride to the bone nor accumulates therein. Herein, we have synthesized a biotinylated boronic ester that is converted to the corresponding trifluoroborate salt in the presence of aqueous [(18)F]-fluoride. In keeping with its in vitro aqueous kinetic stability at pH 7.5, the trifluoroborate appears to clear in vivo quite rapidly to the bladder as the stable trifluoroborate salt with no detectable leaching of free [(18)F]-fluoride to the bone. When this labeled biotin is preincubated with avidin, the pharmacokinetic clearance of the resulting complex is visibly altered. This work validates initial claims that boronic esters are potentially useful as readily labeled precursors to [(18)F]-PET reagents.


Subject(s)
Borates/chemistry , Borates/pharmacokinetics , Fluorine Radioisotopes , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Animals , Borates/chemical synthesis , Female , Fluorine Radioisotopes/chemistry , Isotope Labeling/methods , Kinetics , Mice , Mice, Inbred BALB C , Positron-Emission Tomography/methods , Radiopharmaceuticals/chemical synthesis , Thermodynamics
14.
J Nat Prod ; 70(5): 736-40, 2007 May.
Article in English | MEDLINE | ID: mdl-17407351

ABSTRACT

The absolute configuration of strongylophorine-26 (1) was determined to be 4S, 5R, 8R, 9S, 10S, 13S, 14S by single-crystal X-ray diffraction analysis of the derivative 7 prepared from the co-occurring metabolite strongylophorine-8 (4) and chemical interconversion to the bislactone 8. Synthetic analogues (+)- and (-)-3 have been prepared in order to explore the structure-activity relationship for the anti-invasion pharmacophore of stronglylophorine-26. These studies revealed the unanticipated importance of the A ring lactone moiety for the anti-invasion activity of 1.


Subject(s)
Antineoplastic Agents/chemistry , Diterpenes/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Diterpenes/chemical synthesis , Diterpenes/pharmacology , Drug Screening Assays, Antitumor , Molecular Structure , Stereoisomerism , Structure-Activity Relationship , X-Ray Diffraction
15.
Org Lett ; 8(17): 3749-52, 2006 Aug 17.
Article in English | MEDLINE | ID: mdl-16898808

ABSTRACT

[structure: see text] The new meroterpenoids avinosol (1), 3'-aminoavarone (2), and 3'-phenethylaminoavarone (3) have been isolated from the marine sponge Dysidea sp. collected in Papua New Guinea, and their structures were elucidated by analysis of spectroscopic data. Avinosol (1), which is apparently the first example of a naturally occurring meroterpenoid-nucleoside conjugate, showed antiinvasion activity in a cell-based assay.


Subject(s)
Antineoplastic Agents , Dysidea/chemistry , Terpenes , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Female , Humans , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Papua New Guinea , Terpenes/chemistry , Terpenes/isolation & purification , Terpenes/pharmacology , Tumor Cells, Cultured
16.
Org Lett ; 7(19): 4173-6, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16146380

ABSTRACT

[structure: see text] Neopetrosiamdes A (1) and B (2), two diastereomeric tricyclic peptides that inhibit amoeboid invasion of human tumor cells, have been isolated from the marine sponge Neopetrosia sp. collected in Papua New Guinea. The structures of the neopetrosiamides were elucidated by analysis of MS and NMR data and confirmed by chemical degradation.


Subject(s)
Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Peptides/chemistry , Peptides/pharmacology , Porifera/chemistry , Animals , Cell Line, Tumor , Humans , Magnetic Resonance Spectroscopy , Methionine/analogs & derivatives , Methionine/chemistry , Molecular Structure , Neoplasm Invasiveness/prevention & control , Peptides/isolation & purification , Peptides, Cyclic/isolation & purification , Safrole/analogs & derivatives , Safrole/chemistry , Stereoisomerism
17.
Nat Med ; 9(11): 1428-32, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14578881

ABSTRACT

Hematopoietic stem cells (HSCs) can self-renew extensively after transplantation. The conditions supporting their in vitro expansion are still being defined. Retroviral overexpression of the human homeobox B4 (HOXB4) gene in mouse bone marrow cells enables over 40-fold expansion of HSCs in vitro. To circumvent the requirement for retroviral infection, we used recombinant human TAT-HOXB4 protein carrying the protein transduction domain of the HIV transactivating protein (TAT) as a potential growth factor for stem cells. HSCs exposed to TAT-HOXB4 for 4 d expanded by about four- to sixfold and were 8-20 times more numerous than HSCs in control cultures, indicating that HSC expansion induced by TAT-HOXB4 was comparable to that induced by the human HOXB4 retrovirus during a similar period of observation. Our results also show that TAT-HOXB4-expanded HSC populations retain their normal in vivo potential for differentiation and long-term repopulation. It is thus feasible to exploit recombinant HOXB4 protein for rapid and significant ex vivo expansion of normal HSCs.


Subject(s)
Cell Division/physiology , Gene Products, tat/genetics , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Recombinant Fusion Proteins/genetics , Transcription Factors/genetics , Animals , Gene Products, tat/metabolism , Gene Transfer Techniques , Genetic Vectors , Homeodomain Proteins/metabolism , Humans , Mice , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Retroviridae , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...