Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ray M. Lee is active.

Publication


Featured researches published by Ray M. Lee.


Biochimica et Biophysica Acta | 2009

Mitochondrial kinases and their molecular interaction with cardiolipin.

Uwe Schlattner; Malgorzata Tokarska-Schlattner; Sacnicte Ramirez; Anna Brückner; Laurence Kay; Cécile Polge; Raquel F. Epand; Ray M. Lee; Marie-Lise Lacombe; Richard M. Epand

Mitochondrial isoforms of creatine kinase (MtCK) and nucleoside diphosphate kinase (NDPK-D) are not phylogenetically related but share functionally important properties. They both use mitochondrially generated ATP with the ultimate goal of maintaining proper nucleotide pools, are located in the intermembrane/cristae space, have symmetrical oligomeric structures, and show high affinity binding to anionic phospholipids, in particular cardiolipin. The structural basis and functional consequences of the cardiolipin interaction have been studied and are discussed in detail in this review. They mainly result in a functional interaction of MtCK and NDPK-D with inner membrane adenylate translocator, probably by forming proteolipid complexes. These interactions allow for privileged exchange of metabolites (channeling) that ultimately regulate mitochondrial respiration. Further functions of the MtCK/membrane interaction include formation of cardiolipin membrane patches, stabilization of mitochondria and a role in apoptotic signaling, as well as in case of both kinases, a role in facilitating lipid transfer between two membranes. Finally, disturbed cardiolipin interactions of MtCK, NDPK-D and other proteins like cytochrome c and truncated Bid are discussed more generally in the context of apoptosis and necrosis.


Biochemistry | 2008

Role of Phospholipid Scramblase 3 in the Regulation of Tumor Necrosis Factor-α-Induced Apoptosis†

Jihua Liu; Raquel F. Epand; David Durrant; Douglas Grossman; Nai Wen Chi; Richard M. Epand; Ray M. Lee

In tumor necrosis factor-alpha (TNF-alpha)-induced apoptosis, tBid is targeted to mitochondria and causes cytochrome c release. We investigated the regulation of tBid-induced cytochrome c release and apoptosis by phospholipid scramblase 3 (PLS3). Overexpression of PLS3 enhanced, whereas downregulation of PLS3 delayed, TNF-alpha-induced apoptosis and targeting of tBid to mitochondria. On the basis of the theory that tBid targets mitochondrial cardiolipin, we hypothesize that PLS3 enhances translocation of cardiolipin to the mitochondrial surface to facilitate tBid targeting. NAO, a cardiolipin binding dye, was first used to quantify the distribution of cardiolipin. Overexpression of PLS3 increases, whereas downregulation of PLS3 decreases, the percentage of cardiolipin on the mitochondrial surface. Determination of the tBid binding capacity on the mitochondrial surface by FITC-labeled tBid(G94E) also confirmed that tBid binding capacity increased upon PLS3 overexpression and decreased with downregulation of PLS3. PLS3 activity, determined by a lipid flip-flop assay, was activated by calcium and tBid but inhibited by Bcl-2. Mutation of the calcium binding motif abolishes the lipid flip-flop activity of PLS3. PLS3 and tBid may form a bidirectional positive feedback loop that is antagonized by Bcl-2. Overexpression of PLS3 does not affect mitochondrial potential but does interfere with mitochondrial respiration and production of reactive oxygen species. These studies thus establish PLS3 as an important downstream effector of Bcl-2 and tBid in apoptosis.


Journal of Cellular Biochemistry | 2007

Phosphorylation of mitochondrial phospholipid scramblase 3 by protein kinase C-δ induces its activation and facilitates mitochondrial targeting of tBid

Yongwen He; Jihua Liu; Douglas Grossman; David Durrant; Trevor W. Sweatman; Leonard Lothstein; Raquel F. Epand; Richard M. Epand; Ray M. Lee

Phospholipid scramblase 3 (PLS3) is a member of the phospholipid scramblase family present in mitochondria. PLS3 plays an important role in regulation of mitochondrial morphology, respiratory function, and apoptotic responses. PLS3 is phosphorylated by PKC‐δ at Thr21 and is the mitochondrial target of PKC‐δ‐induced apoptosis. Cells with overexpression of PLS3, but not the phosphoinhibitory mutant PLS3(T21A), are more susceptible to apoptosis induced by AD198, an extranuclear targeted anthracycline that activates PKC‐δ. Here we report that the phosphomimetic mutant of PLS3(T21D) by itself can induce apoptosis in HeLa cells. Using proteoliposomes with addition of pyrene‐labeled phosphatidylcholine (PC) at the outer leaflet, we measured the lipid flip‐flop activity of PLS3 and its phosphorylation mutant. PLS3(T21D) is more potent than wild‐type PLS3 or PLS3(T21A) to transfer pyrene‐PC from the outer leaflet to the inner leaflet of liposomes. Based on our previous finding that PLS3 enhances tBid‐induced mitochondrial damages, we tested the hypothesis that PLS3 enhances cardiolipin translocation to mitochondrial surface and facilitates tBid targeting. Fluorescein‐labeled tBid(G94E) was used as a probe to quantify cardiolipin on the surface of mitochondria. Mitochondria from cells treated with AD198 or cells expressing PLS3(T21D) had a higher level of tBid‐binding capacity than control cells or cells expressing wild‐type PLS3. These findings indicate that phosphorylation of PLS3 by PKC‐δ induces PLS3 activation to facilitate mitochondrial targeting of tBid and apoptosis. J. Cell. Biochem. 101:1210–1221, 2007.


Biochemical Journal | 2006

Tankyrase recruitment to the lateral membrane in polarized epithelial cells: regulation by cell–cell contact and protein poly(ADP-ribosyl)ation

Tsung-Yin J. Yeh; Tobias N. Meyer; Catherine Schwesinger; Zhi-Yang Tsun; Ray M. Lee; Nai-Wen Chi

PARsylation [poly(ADP-ribosyl)ation] of proteins is implicated in the regulation of diverse physiological processes. Tankyrase is a molecular scaffold with this catalytic activity and has been proposed as a regulator of vesicular trafficking on the basis, in part, of its Golgi localization in non-polarized cells. Little is known about tankyrase localization in polarized epithelial cells. Using MDCK (Madin-Darby canine kidney) cells as a model, we found that E-cadherin-mediated intercellular adhesion recruits tankyrase from the cytoplasm to the lateral membrane (including the tight junction), where it stably associates with detergent-insoluble structures. This recruitment is mostly completed within 8 h of calcium-induced formation of cell-cell contact. Conversely, when intercellular adhesion is disrupted by calcium deprivation, tankyrase returns from the lateral membrane to the cytoplasm and becomes more soluble in detergents. The PARsylating activity of tankyrase promotes its dissociation from the lateral membrane as well as its ubiquitination and proteasome-mediated degradation, resulting in an apparent protein half-life of approximately 2 h. Inhibition of tankyrase autoPARsylation using H2O2-induced NAD+ depletion or PJ34 [N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-N,N-dimethylacetamide hydrochloride] treatment results in tankyrase stabilization and accumulation at the lateral membrane. By contrast, stabilization through proteasome inhibition results in tankyrase accumulation in the cytoplasm. These data suggest that cell-cell contact promotes tankyrase association with the lateral membrane, whereas PARsylating activity promotes translocation to the cytosol, which is followed by ubiquitination and proteasome-mediated degradation. Since the lateral membrane is a sorting station that ensures domain-specific delivery of basolateral membrane proteins, the regulated tankyrase recruitment to this site is consistent with a role in polarized protein targeting in epithelial cells.


Bioorganic & Medicinal Chemistry | 2009

Design, synthesis and biological evaluation of novel stilbene-based antitumor agents

Daniele Simoni; Francesco Paolo Invidiata; Marco Eleopra; Paolo Marchetti; Riccardo Rondanin; Riccardo Baruchello; Giuseppina Grisolia; Ashutosh Tripathi; Glen E. Kellogg; David Durrant; Ray M. Lee

A series of novel stilbene derivatives has been synthesized and studied with the main goal to investigate SAR of the amino compound 1a, as well as to improve its water solubility, a potentially negative aspect of the molecule that could be a serious obstacle for a pre-clinical development. We have obtained derivatives with good cytotoxic activity, in particular, the derivatives 5c and 6b could represent two novel leads for further investigation. Compound 8b, a morpholino-carbamate derivative, prodrug of 1a, has a very good solubility in water, and is active in suppressing growth of tumor cells at a concentration of 5000 nM, which is a concentration 100 times higher than the parent stilbene 1a.


American Journal of Hematology | 2008

Stilbene derivatives that are colchicine site microtubule inhibitors have antileukemic activity and minimal systemic toxicity

Thai M. Cao; David Durrant; Ashutosh Tripathi; Jihua Liu; Schickwann Tsai; Glen E. Kellogg; Daniele Simoni; Ray M. Lee

Stilbenes are a group of natural compounds with many biological activities. Two highly potent stilbenes, cis‐3,4′,5‐trimethoxy‐3′‐aminostilbene (stilbene 5c) and cis‐3,4′,5‐trimethoxy‐3′‐hydroxystilbene (stilbene 6c) induce G2/M cell‐cycle arrest and leukemic cell death in nanomolarity range without affecting normal bone marrow progenitor cells. The mechanism of stilbenes is mediated by interfering with microtubule polymerization through the colchicine‐binding site. Docking of the stilbenes into tubulin structure confirms that stilbenes fit into the colchicine‐binding pocket. Animal studies show that stilbenes are well tolerated in mice and are capable of inducing more than 50% leukemic cell death by a single dose injection. A 5‐day treatment with low‐dose stilbenes suppresses tumor growth in mice with established tumor xenografts. No major organ damage was detected by histological section. Our results indicate that stilbene 5c is a microtubule‐interfering agent and can be potentially useful in leukemic therapy. Am. J. Hematol., 2008.


Investigational New Drugs | 2009

Development of water soluble derivatives of cis-3, 4′, 5-trimethoxy-3′-aminostilbene for optimization and use in cancer therapy

David Durrant; Joanna Richards; Ashutosh Tripathi; Glen E. Kellogg; Paolo Marchetti; Marco Eleopra; Giuseppina Grisolia; Daniele Simoni; Ray M. Lee

SummaryColchicine site tubulin inhibitors are currently developed as vascular disrupting agents (VDAs). However, they were found to have cardiotoxicity in clinical trials. To overcome the problem, we developed a stilbene derivative, cis-3, 4′, 5-trimethoxy-3′-aminostilbene (stilbene 5c), which is highly potent and has no bone marrow and cardiac toxicity in mice. Here we attempt to optimize stilbene 5c using computer-based drug design and synthesize derivatives with benzimidazole or indole group. Biological evaluation showed that they are weaker than stilbene 5c without better water solubility. Alternative approach was thus adopted to make prodrugs of stilbene 5c. A water-soluble prodrug PD7 was synthesized by addition of a morpholino group with carbamate linkage to the amino group of stilbene 5c. In vitro studies show that PD7 induces mitotic arrest and disrupts microtubule similar to stilbene 5c. The cell signaling events in Cdc2, p53, Akt, and aurora kinase are similar in cells treated with stilbene 5c, CA4 or PD7, suggesting that they share the same mechanism. Although PD7 is less effective than stilbene 5c in vitro, the biological activity of PD7 as a single agent is similar to that of stilbene 5c. Combination of PD7 with VEGF inhibitor bevacizumab significantly enhances the therapeutic efficacy of PD7 in mouse xenograft model. These data suggest that PD7 could be a good candidate for further pre-clinical and clinical development as a new VDA for cancer therapy.


Journal of Enzyme Inhibition and Medicinal Chemistry | 2009

Hydropathic analysis and biological evaluation of stilbene derivatives as colchicine site microtubule inhibitors with anti-leukemic activity

Ashutosh Tripathi; David Durrant; Ray M. Lee; Riccardo Baruchello; Romeo Romagnoli; Daniele Simoni; Glen E. Kellogg

The crucial role of the microtubule in cell division has identified tubulin as a target for the development of therapeutics for cancer; in particular, tubulin is a target for antineoplastic agents that act by interfering with the dynamic stability of microtubules. A molecular modeling study was carried out to accurately represent the complex structure and the binding mode of a new class of stilbene-based tubulin inhibitors that bind at the αβ-tubulin colchicine site. Computational docking along with HINT (Hydropathic INTeractions) score analysis fitted these inhibitors into the colchicine site and revealed detailed structure–activity information useful for inhibitor design. Quantitative analysis of the results was in good agreement with the in vitro antiproliferative activity of these derivatives (ranging from 3 nM to 100 μM) such that calculated and measured free energies of binding correlate with an r2 of 0.89 (standard error ± 0.85 kcal mol−1). This correlation suggests that the activity of unknown compounds may be predicted.


Cancer Research | 2012

Abstract 3957: Cell death and growth arrest pathways mediating the actions of stilbene 5C in HCT-116 colon cancer cells

Moureq R. Alotaibi; Matthew J. Beckman; Xu Di; Ray M. Lee; David A. Gewirtz

The stilbene derivative, cis-3, 4′, 5-trimethoxy-3′-aminostilbene (stilbene 5c), is a potentially potent antitumor agent that acts via binding to the colchicine-binding pocket in microtubules. Earlier studies have shown that stilbene 5c induces cell death in ovarian cancer cells and leukemic cells (Lee RM et al, 2008; Lee RM et al, 2008). The present study was designed to investigate the effectiveness of this microtubule poison against the HCT-116 human colon cancer cell line and its mechanisms of action. Time course studies demonstrated that stilbene 5c produces a biphasic decrease in cell viability. The capacity of the cells to proliferate was not restored upon removal of the drug after 6 days of exposure. Consistent with the results of the time course studies, β-galactosidase staining indicated that treatment with stilbene 5c also promotes senescence. In addition to senescence, stilbene 5c-treated HCT-116 cells displayed formation of autophagic vesicles by acridine orange staining, which was supported by fluorescence-activated cell sorting (FACS). Further evidence of autophagy was derived from treatment of HCT116 cells carrying an RFP-LC3 construct with stilbene 5c, in which LC3 puncta formation increased in a time-dependent manner. DAPI staining, TUNEL, and Annexin 5 staining indicated that apoptosis is also occurring in stilbene 5c-treated HCT-116 cells. Cell cycle analysis demonstrated growth arrest at both G1 and G2/M, and an increase in the subG1 population at days 3 and 5, which correspond to senescence and apoptosis respectively. Interestingly, DAPI and Hoechst staining revealed morphological changes in the cell nuclei (binucleated and micronucleated cells), which suggest that mitotic catastrophe may also serve as a mode of cell death after treatment with stilbene 5c. Consistent with previous studies in other experimental cancer models, this work indicates that stilbene 5c could potentially be effective against colon cancer through the promotion of multiple modes of cell death. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3957. doi:1538-7445.AM2012-3957


Biochemical Journal | 2007

Phospholipid scramblase-3 regulates cardiolipin de novo biosynthesis and its resynthesis in growing HeLa cells.

Quyen Van; Jihua Liu; Biao Lu; Kenneth R. Feingold; Yuguang Shi; Ray M. Lee; Grant M. Hatch

Collaboration


Dive into the Ray M. Lee's collaboration.

Top Co-Authors

Avatar

David Durrant

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ashutosh Tripathi

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Glen E. Kellogg

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David A. Gewirtz

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joanna Richards

Virginia Commonwealth University

View shared research outputs
Researchain Logo
Decentralizing Knowledge