Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 6(6): e20845, 2011.
Article in English | MEDLINE | ID: mdl-21695258

ABSTRACT

BACKGROUND: Missing in metastasis (MIM) is a member of the inverse BAR-domain protein family, and in vitro studies have implied MIM plays a role in deforming membrane curvature into filopodia-like protrusions and cell dynamics. Yet, the physiological role of the endogenous MIM in mammalian cells remains undefined. PRINCIPAL FINDINGS: We have examined mouse embryonic fibroblasts (MEFs) derived from mice in which the MIM locus was targeted by a gene trapping vector. MIM(-/-) MEFs showed a less polarized architecture characterized by smooth edges and fewer cell protrusions as compared to wild type cells, although the formation of filopodia-like microprotrusions appeared to be normal. Immunofluorescent staining further revealed that MIM(-/-) cells were partially impaired in the assembly of stress fibers and focal adhesions but were enriched with transverse actin filaments at the periphery. Poor assembly of stress fibers was apparently correlated with attenuation of the activity of Rho GTPases and partially relieved upon overexpressing of Myc-RhoA(Q63L), a constitutively activated RhoA mutant. MIM(-/-) cells were also spread less effectively than wild type cells during attachment to dishes and substratum. Upon treatment with PDGF MIM(-/-) cells developed more prominent dorsal ruffles along with increased Rac1 activity. Compared to wild type cells, MIM(-/-) cells had a slower motility in the presence of a low percentage of serum-containing medium but migrated normally upon adding growth factors such as 10% serum, PDGF or EGF. MIM(-/-) cells were also partially impaired in the internalization of transferrin, fluorescent dyes, foreign DNAs and PDGF receptor alpha. On the other hand, the level of tyrosine phosphorylation of PDGF receptors was more elevated in MIM depleted cells than wild type cells upon PDGF treatment. CONCLUSIONS: Our data suggests that endogenous MIM protein regulates globally the cell architecture and endocytosis that ultimately influence a variety of cellular behaviors, including cell polarity, motility, receptor signaling and membrane ruffling.


Subject(s)
Cell Movement/drug effects , Cell Polarity/drug effects , Microfilament Proteins/metabolism , Neoplasm Proteins/metabolism , Platelet-Derived Growth Factor/pharmacology , Actins/metabolism , Animals , Cell Adhesion/drug effects , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Shape/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Extracellular Space/drug effects , Extracellular Space/metabolism , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Knockout Techniques , Genetic Vectors/genetics , Male , Mice , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Protein Transport/drug effects , Transferrin/metabolism
2.
Biochem Biophys Res Commun ; 401(4): 527-32, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-20875796

ABSTRACT

Abba is a member of the I-BAR-domain protein family that is characterized by a convex-shaped membrane-binding motif. Overexpression of GFP-tagged Abba in murine fibroblasts potentiated PDGF-mediated formation of membrane ruffles and lamellipodia. Immunofluorescent microscopy and pull-down analysis revealed that GFP-Abba colocalized with an active form of Rac1 in the membrane ruffles and enhanced the Rac GTPase activity in response to PDGF stimulation. Further immunoprecipitation assays demonstrated that GFP-Abba bound to both wild-type and constitutively active Rac1 and that the binding to either of the Rac1 forms was significantly enhanced upon PDGF stimulation. On the other hand, an Abba mutant deficient in Rac1 binding failed to promote membrane ruffling and Rac1 activation in response to PDGF. However, the cells overexpressing a truncated mutant carrying the I-BAR domain alone displayed numerous filopodia-like microspikes in a manner independent of growth factors. Also, the Rac-binding activity of the mutant was not affected by PDGF treatment. Our data indicates that the interaction between full-length Abba and Rac1 is implicated in membrane deformation and subjected to a growth factor-mediated regulation through the C-terminal sequence.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/metabolism , Platelet-Derived Growth Factor/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Humans , Membrane Proteins/genetics , Mice , Microfilament Proteins , NIH 3T3 Cells , Platelet-Derived Growth Factor/pharmacology , Protein Structure, Tertiary
3.
Bioorg Med Chem ; 17(20): 7344-52, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19762246

ABSTRACT

A novel series of 17-modified and 2,17-modified analogs of 2-methoxyestradiol (2ME2) were synthesized and characterized. These analogs were designed to retain or potentiate the biological activities of 2ME2 and have diminished metabolic liability. The analogs were evaluated for antiproliferative activity against MDA-MB-231 breast tumor cells, antiangiogenic activity in HUVEC, and estrogenic activity on MCF-7 cell proliferation. Several analogs were evaluated for metabolic stability in human liver microsomes and in vivo in a rat cassette dosing model. This study lead to several 17-modified analogs of 2ME2 that have similar or improved antiproliferative and antiangiogenic activity, lack estrogenic properties and have improved metabolic stability compared to 2ME2.


Subject(s)
Cell Proliferation/drug effects , Estradiol/analogs & derivatives , Estrone/chemical synthesis , Estrone/pharmacology , 2-Methoxyestradiol , Animals , Cell Line , Estradiol/pharmacology , Estrone/chemistry , Humans , Rats , Structure-Activity Relationship
4.
Bioorg Med Chem Lett ; 19(22): 6459-62, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19782568

ABSTRACT

The syntheses of 2-methoxyestradiol analogs with modifications at the 3-position are described. The analogs were assessed for their antiproliferative, antiangiogenic, and estrogenic activities. Several lead substituents were identified with similar or improved antitumor activities and reduced metabolic liability compared to 2-methoxyestradiol.


Subject(s)
Estradiol/analogs & derivatives , Spindle Apparatus/drug effects , 2-Methoxyestradiol , Animals , Cell Proliferation/drug effects , Cells, Cultured , Drug Screening Assays, Antitumor , Estradiol/pharmacology , Estradiol/therapeutic use , Humans , Male , Mice , Mice, Nude , Models, Molecular , Xenograft Model Antitumor Assays/methods , Xenograft Model Antitumor Assays/statistics & numerical data
5.
Angiogenesis ; 7(2): 115-22, 2004.
Article in English | MEDLINE | ID: mdl-15516832

ABSTRACT

The medicinal plant Withania somnifera is widely researched for its anti-inflammatory, cardioactive and central nervous system effects. In Ayurveda , the major Traditional Indian medicine system, extracts from W. somnifera are distinctively employed for the treatment of arthritis and menstrual disorders. Because these conditions involve angiogenic processes we hypothesized that the W. somnifera extracts might contain angiogenesis inhibitors. We employed an endothelial cell-sprouting assay to monitor the purification of substances from W. somnifera root extracts and isolated as the active principle the previously known natural product withaferin A. We show that withaferin A inhibits human umbilical vein endothelial cell (HUVEC) sprouting in three-dimensional collagen-I matrix at doses which are relevant to NF-kappa B-inhibitory activity. Withaferin A inhibits cell proliferation in HUVECs (IC50 =12 nM) at doses that are significantly lower than those required for tumor cell lines through a process associated with inhibition of cyclin D1 expression. We propose that the inhibition of NF-kappa B by withaferin A in HUVECs occurs by interference with the ubiquitin-mediated proteasome pathway as suggested by the increased levels of poly-ubiquitinated proteins. Finally, withaferin A is shown to exert potent anti-angiogenic activity in vivo at doses that are 500-fold lower than those previously reported to exert anti-tumor activity in vivo. In conclusion, our findings identify a novel mode of action of withaferin A, which highlights the potential use of this natural product for cancer treatment or prevention.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Ergosterol/analogs & derivatives , Ergosterol/pharmacology , Neovascularization, Pathologic/drug therapy , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Mice , Mice, Inbred C57BL , NF-kappa B/antagonists & inhibitors , Plants, Medicinal/chemistry , Proteasome Endopeptidase Complex/metabolism , Spheroids, Cellular , Ubiquitin/metabolism , Umbilical Veins , Withanolides
6.
Cancer Res ; 63(2): 468-75, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12543804

ABSTRACT

2-Methoxyestradiol (2ME2), a natural metabolite of estradiol, is a potent antitumor and antiangiogenic agent. In vitro, 2ME2 inhibits the proliferation of a wide variety of cell lines and primary cultures, and in numerous models in vivo, it has been shown to be an effective inhibitor of tumor growth and angiogenesis. 2ME2 is currently in several Phase I and Phase II clinical trials under the name Panzem. Although various molecular targets have been proposed for this compound, the mechanism by which 2ME2 exerts its effects is still uncertain. This study shows that 2ME2 uses the extrinsic pathway for induction of apoptosis. 2ME2 treatment results in up-regulation of death receptor 5 (DR5) protein expression in vitro and in vivo and renders cells more sensitive to the cytotoxic activities of the DR5 ligand tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). 2ME2-induced apoptosis requires caspase activation and kinetic studies show the sequential activation of caspase-8, caspase-9, and caspase-3. Blockage of death receptor signaling by expression of dominant-negative Fas-associated death domain severely attenuates the ability of 2ME2 to induce apoptosis. Because 2ME2 administration has not manifested dose-limiting toxicity in the clinic, DR5 expression may serve as a surrogate marker for biological response.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/drug effects , Estradiol/pharmacology , Receptors, Tumor Necrosis Factor/physiology , 2-Methoxyestradiol , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Carrier Proteins/physiology , Caspases/metabolism , Cell Division/drug effects , Drug Synergism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Activation , Estradiol/analogs & derivatives , Fas-Associated Death Domain Protein , Female , Humans , Isoenzymes/metabolism , Membrane Glycoproteins/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction/physiology , TNF-Related Apoptosis-Inducing Ligand , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
7.
Cancer Res ; 62(13): 3691-7, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12097276

ABSTRACT

2-Methoxyestradiol (2ME(2)) is an endogenous metabolite of 17beta-estradiol (E(2)) that arises from the hydroxylation and subsequent methylation at the 2-position. In vitro 2ME(2) inhibits a large variety of tumor and nontumor cell lines from diverse origins, as well as several stages of the angiogenic cascade. In vivo it has been shown to be a very effective inhibitor of tumor growth and angiogenesis in numerous models. Although various molecular targets have been proposed for this compound, the mechanism of action is still uncertain. As this molecule emerges as a drug candidate it is important to assess the estrogen receptors (ERs) as molecular targets for 2ME(2). The purpose of this study was to investigate whether 2ME(2) is able to engage ERs as an agonist and whether its antiproliferative activities are mediated through ERs. We confirm that 2ME(2) has a lower binding affinity for ERalpha as compared with E(2) and other E(2) metabolites and antagonists, and we demonstrate that the affinity of 2ME(2) for ERbeta is even lower. When assessed in the presence of galangin, a cytochrome P450 enzyme inhibitor, at concentrations at which 2ME(2) interacts with ERalpha in an in vitro binding assay, it does not stimulate the proliferation of an estrogen-dependent breast carcinoma cell line. Similar IC(50) values for inhibition of proliferation and induction of apoptosis are obtained in estrogen-dependent and estrogen-independent human breast cancer cell lines, irrespective of the expression of ERalpha and ERbeta. Moreover, the estrogen antagonist ICI 182,780 does not inhibit the antiproliferative activity of 2ME(2). In E(2)-responsive cells such as MCF-7 and human umbilical vascular endothelial cells, high levels of E(2) inhibit the antiproliferative activity of ICI 182,780 but not of 2ME(2). Collectively, these results suggest that 2ME(2) is distinct among estradiol metabolites because of its inability to engage ERs as an agonist, and its unique antiproliferative and apoptotic activities are mediated independently of ERalpha and ERbeta.


Subject(s)
Apoptosis/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Receptors, Estrogen/physiology , 2-Methoxyestradiol , Apoptosis/physiology , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Division/drug effects , Cytochrome P-450 Enzyme Inhibitors , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Estradiol/metabolism , Estrogen Receptor alpha , Estrogen Receptor beta , Flavonoids/pharmacology , Fulvestrant , Humans , Receptors, Estrogen/agonists , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...