Network


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

Hotspot


Dive into the research topics where Derek D. Norman is active.

Publication


Featured researches published by Derek D. Norman.


Bioconjugate Chemistry | 2013

Gold Nanorods Carrying Paclitaxel for Photothermal-Chemotherapy of Cancer

Fei Ren; Saheel Bhana; Derek D. Norman; Jermaine Johnson; Lijing Xu; Daniel L. Baker; Xiaohua Huang

Nanotechnology-based photothermal therapy has emerged as a promising treatment for cancer during the past decade. However, heterogeneous laser heating and limited light penetration can lead to incomplete tumor cell eradication. Here, we developed a method to overcome these limitations by combining chemotherapy with photothermal therapy using paclitaxel-loaded gold nanorods. Paclitaxel was loaded to gold nanorods with high density (2.0 × 10(4) paclitaxel per gold nanorod) via nonspecific adsorption, followed by stabilization with poly(ethylene glycol) linked with 11-mercaptoundecanoic acid. Paclitaxel was entrapped in the hydrophobic pocket of the polymeric monolayer on the surface of gold nanorods, which allows direct cellular delivery of the hydrophobic drugs via the lipophilic plasma membrane. Highly efficient drug release was demonstrated in a cell membrane mimicking two-phase solution. Combined photothermal therapy and chemotherapy with the paclitaxel-loaded gold nanorods was shown to be highly effective in killing head and neck cancer cells and lung cancer cells, superior to photothermal therapy or chemotherapy alone due to a synergistic effect. The paclitaxel-gold nanorod enabled photothermal chemotherapy has the potential of preventing tumor reoccurrence and metastasis and may have an important impact on the treatment of head and neck cancer and other malignancies in the clinic.


Blood | 2014

Interaction of platelet-derived autotaxin with tumor integrin αVβ3 controls metastasis of breast cancer cells to bone.

Raphael Leblanc; Sue Chin Lee; Marion David; Jean Claude Bordet; Derek D. Norman; Renukadevi Patil; Duane D. Miller; Debashish Sahay; Johnny Ribeiro; Philippe Clézardin; Gabor Tigyi; Olivier Peyruchaud

Autotaxin (ATX), through its lysophospholipase D activity controls physiological levels of lysophosphatidic acid (LPA) in blood. ATX is overexpressed in multiple types of cancers, and together with LPA generated during platelet activation promotes skeletal metastasis of breast cancer. However, the pathophysiological sequelae of regulated interactions between circulating LPA, ATX, and platelets remain undefined in cancer. In this study, we show that ATX is stored in α-granules of resting human platelets and released upon tumor cell-induced platelet aggregation, leading to the production of LPA. Our in vitro and in vivo experiments using human breast cancer cells that do not express ATX (MDA-MB-231 and MDA-B02) demonstrate that nontumoral ATX controls the early stage of bone colonization by tumor cells. Moreover, expression of a dominant negative integrin αvβ3-Δ744 or treatment with the anti-human αvβ3 monoclonal antibody LM609, completely abolished binding of ATX to tumor cells, demonstrating the requirement of a fully active integrin αvβ3 in this process. The present results establish a new mechanism for platelet contribution to LPA-dependent metastasis of breast cancer cells, and demonstrate the therapeutic potential of disrupting the binding of nontumor-derived ATX with the tumor cells for the prevention of metastasis.


Journal of Lipid Research | 2011

The phospholipase A1 activity of lysophospholipase A-I links platelet activation to LPA production during blood coagulation

Alyssa L. Bolen; Anjaparavanda P. Naren; Sunitha Yarlagadda; Sarka Beranova-Giorgianni; Li Chen; Derek D. Norman; Daniel L. Baker; Meng M. Rowland; Michael D. Best; Takamitsu Sano; Károly Liliom; Yasuyuki Igarashi; Gabor Tigyi

Platelet activation initiates an upsurge in polyunsaturated (18:2 and 20:4) lysophosphatidic acid (LPA) production. The biochemical pathway(s) responsible for LPA production during blood clotting are not yet fully understood. Here we describe the purification of a phospholipase A1 (PLA1) from thrombin-activated human platelets using sequential chromatographic steps followed by fluorophosphonate (FP)-biotin affinity labeling and proteomics characterization that identified acyl-protein thioesterase 1 (APT1), also known as lysophospholipase A-I (LYPLA-I; accession code O75608) as a novel PLA1. Addition of this recombinant PLA1 significantly increased the production of sn-2-esterified polyunsaturated LPCs and the corresponding LPAs in plasma. We examined the regioisomeric preference of lysophospholipase D/autotaxin (ATX), which is the subsequent step in LPA production. To prevent acyl migration, ether-linked regioisomers of oleyl-sn-glycero-3-phosphocholine (lyso-PAF) were synthesized. ATX preferred the sn-1 to the sn-2 regioisomer of lyso-PAF. We propose the following LPA production pathway in blood: 1) Activated platelets release PLA1; 2) PLA1 generates a pool of sn-2 lysophospholipids; 3) These newly generated sn-2 lysophospholipids undergo acyl migration to yield sn-1 lysophospholipids, which are the preferred substrates of ATX; and 4) ATX cleaves the sn-1 lysophospholipids to generate sn-1 LPA species containing predominantly 18:2 and 20:4 fatty acids.


Molecular Cancer Research | 2015

Autotaxin and LPA1 and LPA5 receptors exert disparate functions in tumor cells versus the host tissue microenvironment in melanoma invasion and metastasis.

Sue Chin Lee; Yuko Fujiwara; Jianxiong Liu; Junming Yue; Yoshibumi Shimizu; Derek D. Norman; Yaohong Wang; Ryoko Tsukahara; Erzsebet Szabo; Renukadevi Patil; Souvik Banerjee; Duane D. Miller; Louisa Balazs; Manik C. Ghosh; Christopher M. Waters; Tamas Oravecz; Gabor Tigyi

Autotaxin (ENPP2/ATX) and lysophosphatidic acid (LPA) receptors represent two key players in regulating cancer progression. The present study sought to understand the mechanistic role of LPA G protein–coupled receptors (GPCR), not only in the tumor cells but also in stromal cells of the tumor microenvironment. B16F10 melanoma cells predominantly express LPA5 and LPA2 receptors but lack LPA1. LPA dose dependently inhibited invasion of cells across a Matrigel layer. RNAi-mediated knockdown of LPA5 relieved the inhibitory effect of LPA on invasion without affecting basal invasion. This suggests that LPA5 exerts an anti-invasive action in melanoma cells in response to LPA. In addition, both siRNA-mediated knockdown and pharmacologic inhibition of LPA2 reduced the basal rate invasion. Unexpectedly, when probing the role of this GPCR in host tissues, it was found that the incidence of melanoma-derived lung metastasis was greatly reduced in LPA5 knockout (KO) mice compared with wild-type (WT) mice. LPA1-KO but not LPA2-KO mice also showed diminished melanoma-derived lung metastasis, suggesting that host LPA1 and LPA5 receptors play critical roles in the seeding of metastasis. The decrease in tumor cell residence in the lungs of LPA1-KO and LPA5-KO animals was apparent 24 hours after injection. However, KO of LPA1, LPA2, or LPA5 did not affect the subcutaneous growth of melanoma tumors. Implications: These findings suggest that tumor and stromal LPA receptors, in particular LPA1 and LPA5, play different roles in invasion and the seeding of metastasis. Mol Cancer Res; 13(1); 174–85. ©2014 AACR.


Molecular Pharmacology | 2013

Hits of a High-Throughput Screen Identify the Hydrophobic Pocket of Autotaxin/Lysophospholipase D As an Inhibitory Surface

James I. Fells; Sue Chin Lee; Yuko Fujiwara; Derek D. Norman; Keng Gat Lim; Ryoko Tsukahara; Jianxiong Liu; Renukadevi Patil; Duane D. Miller; R. Jason Kirby; Sandra Nelson; William Seibel; Ruben Papoian; Daniel L. Baker; Robert Bittman; Gabor Tigyi

Autotaxin (ATX), a lysophospholipase D, plays an important role in cancer invasion, metastasis, tumor progression, tumorigenesis, neuropathic pain, fibrotic diseases, cholestatic pruritus, lymphocyte homing, and thrombotic diseases by producing the lipid mediator lysophosphatidic acid (LPA). A high-throughput screen of ATX inhibition using the lysophosphatidylcholine-like substrate fluorogenic substrate 3 (FS-3) and ∼10,000 compounds from the University of Cincinnati Drug Discovery Center identified several small-molecule inhibitors with IC50 vales ranging from nanomolar to low micromolar. The pharmacology of the three most potent compounds: 918013 (1; 2,4-dichloro-N-(3-fluorophenyl)-5-(4-morpholinylsulfonyl) benzamide), 931126 (2; 4-oxo-4-{2-[(5-phenoxy-1H-indol-2-yl)carbonyl]hydrazino}-N-(4-phenylbutan-2-yl)butanamide), and 966791 (3; N-(2,6-dimethylphenyl)-2-[N-(2-furylmethyl)(4-(1,2,3,4-tetraazolyl)phenyl)carbonylamino]-2-(4-hydroxy-3-methoxyphenyl) acetamide), were further characterized in enzyme, cellular, and whole animal models. Compounds 1 and 2 were competitive inhibitors of ATX-mediated hydrolysis of the lysophospholipase substrate FS-3. In contrast, compound 3 was a competitive inhibitor of both FS-3 and the phosphodiesterase substrate p-nitrophenyl thymidine 5′-monophosphate. Computational docking and mutagenesis suggested that compounds 1 and 2 target the hydrophobic pocket, thereby blocking access to the active site of ATX. The potencies of compounds 1–3 were comparable to each other in each of the assays. All of these compounds significantly reduced invasion of A2058 human melanoma cells in vitro and the colonization of lung metastases by B16-F10 murine melanoma cells in C57BL/6 mice. The compounds had no agonist or antagonist effects on select LPA or sphingosine 1-phosphate receptors, nor did they inhibit nucleotide pyrophosphatase/phosphodiesterase (NPP) enzymes NPP6 and NPP7. These results identify the molecular surface of the hydrophobic pocket of ATX as a target-binding site for inhibitors of enzymatic activity.


Experimental Parasitology | 2008

Dermacentor variabilis: Regulation of fibroblast migration by tick salivary gland extract and saliva

Carolyn D. Kramer; Zachary Nahmias; Derek D. Norman; Tara A. Mulvihill; Lewis B. Coons; Judith A. Cole

We examined the effects of tick SGx and saliva on basal- and platelet-derived growth factor (PDGF)-stimulated cell migration and extracellular signal-regulated kinase (ERK) signaling in fibroblasts. Repair of injured monolayers was delayed by SGx pretreatment and was not associated with reductions in cell number. In migration assays, SGx suppressed both basal- and PDGF-stimulated fibroblast movement. Furthermore, SGx and saliva reduced PDGF-stimulated ERK activity. Thus, the delayed repair of monolayer injuries resulted from SGx inhibiting fibroblast migratory responses to chemotactic signals. SGx also suppressed injury- and growth factor-induced ERK activation in renal epithelial OK cells. Our data suggest that maintenance of the tick feeding lesion results, in part, from suppressing ERK signaling and fibroblast migration, events playing integral roles in the wound healing response. The effects of SGx on cells not involved in wound healing suggest that a constituent(s) in tick saliva has global effects on the ERK signaling pathway.


Journal of Biological Chemistry | 2014

Structural determinants of the transient receptor potential 1 (TRPV1) channel activation by phospholipid analogs.

Sara L. Morales-Lázaro; Barbara Serrano-Flores; Itzel Llorente; Enrique Hernández-García; Ricardo González-Ramírez; Souvik Banerjee; Duane D. Miller; Veeresh Gududuru; James I. Fells; Derek D. Norman; Gabor Tigyi; Diana Escalante-Alcalde; Tamara Rosenbaum

Background: The TRPV1 ion channel can be regulated by negatively charged lipids. Results: TRPV1 shows specificity for LPA analogs containing monounsaturated hydrocarbon chains with a negatively charged phosphate, cyclic phosphate, and thiophosphate headgroup. Conclusion: TRPV1 activation is highly restricted to natural lipids with oleyl or oleoyl side chains. Significance: Production of endogenous C18:1 lysophospholipids can selectively trigger activation of TRPV1 and nociceptive neuronal responses. The transient receptor potential vanilloid 1 (TRPV1) ion channel is a polymodal protein that responds to various stimuli, including capsaicin (the pungent compound found in chili peppers), extracellular acid, and basic intracellular pH, temperatures close to 42 °C, and several lipids. Lysophosphatidic acid (LPA), an endogenous lipid widely associated with neuropathic pain, is an agonist of the TRPV1 channel found in primary afferent nociceptors and is activated by other noxious stimuli. Agonists or antagonists of lipid and other chemical natures are known to possess specific structural requirements for producing functional effects on their targets. To better understand how LPA and other lipid analogs might interact and affect the function of TRPV1, we set out to determine the structural features of these lipids that result in the activation of TRPV1. By changing the acyl chain length, saturation, and headgroup of these LPA analogs, we established strict requirements for activation of TRPV1. Among the natural LPA analogs, we found that only LPA 18:1, alkylglycerophosphate 18:1, and cyclic phosphatidic acid 18:1, all with a monounsaturated C18 hydrocarbon chain activate TRPV1, whereas polyunsaturated and saturated analogs do not. Thus, TRPV1 shows a more restricted ligand specificity compared with LPA G-protein-coupled receptors. We synthesized fatty alcohol phosphates and thiophosphates and found that many of them with a single double bond in position Δ9, 10, or 11 and Δ9 cyclopropyl group can activate TRPV1 with efficacy similar to capsaicin. Finally, we developed a pharmacophore and proposed a mechanistic model for how these lipids could induce a conformational change that activates TRPV1.


Cellular Signalling | 2015

The autotaxin–LPA2 GPCR axis is modulated by γ-irradiation and facilitates DNA damage repair

Andrea Balogh; Yoshibumi Shimizu; Sue Chin Lee; Derek D. Norman; Ruchika Gangwar; Mitul Bavaria; Chang Suk Moon; Pradeep K. Shukla; Radakrishna Rao; Ramesh M. Ray; Anjaparavanda P. Naren; Souvik Banerje; Duane D. Miller; Louisa Balazs; Louis M. Pelus; Gabor Tigyi

In this study we characterized the effects of radiation injury on the expression and function of the autotaxin (ATX)-LPA2 GPCR axis. In IEC-6 crypt cells and jejunum enteroids quantitative RT-PCR showed a time- and dose-dependent upregulation of lpa2 in response to γ-irradiation that was abolished by mutation of the NF-κB site in the lpa2 promoter or by inhibition of ATM/ATR kinases with CGK-733, suggesting that lpa2 is a DNA damage response gene upregulated by ATM via NF-κB. The resolution kinetics of the DNA damage marker γ-H2AX in LPA-treated IEC-6 cells exposed to γ-irradiation was accelerated compared to vehicle, whereas pharmacological inhibition of LPA2 delayed the resolution of γ-H2AX. In LPA2-reconstituted MEF cells lacking LPA1&3 the levels of γ-H2AX decreased rapidly, whereas in Vector MEF were high and remained sustained. Inhibition of ERK1&2 or PI3K/AKT signaling axis by pertussis toxin or the C311A/C314A/L351A mutation in the C-terminus of LPA2 abrogated the effect of LPA on DNA repair. LPA2 transcripts in Lin(-)Sca-1(+)c-Kit(+) enriched for bone marrow stem cells were 27- and 5-fold higher than in common myeloid or lymphoid progenitors, respectively. Furthermore, after irradiation higher residual γ-H2AX levels were detected in the bone marrow or jejunum of irradiated LPA2-KO mice compared to WT mice. We found that γ-irradiation increases plasma ATX activity and LPA level that is in part due to the previously established radiation-induced upregulation of TNFα. These findings identify ATX and LPA2 as radiation-regulated genes that appear to play a physiological role in DNA repair.


FEBS Journal | 2014

Targeting the hydrophobic pocket of autotaxin with virtual screening of inhibitors identifies a common aromatic sulfonamide structural motif.

James I. Fells; Sue Chin Lee; Derek D. Norman; Ryoko Tsukahara; Jason Kirby; Sandra Nelson; William Seibel; Ruben Papoian; Renukadevi Patil; Duane D. Miller; Truc Chi T. Pham; Daniel L. Baker; Robert Bittman; Gabor Tigyi

Modulation of autotaxin (ATX), the lysophospholipase D enzyme that produces lysophosphatidic acid, with small‐molecule inhibitors is a promising strategy for blocking the ATX–lysophosphatidic acid signaling axis. Although discovery campaigns have been successful in identifying ATX inhibitors, many of the reported inhibitors target the catalytic cleft of ATX. A recent study provided evidence for an additional inhibitory surface in the hydrophobic binding pocket of ATX, confirming prior studies that relied on enzyme kinetics and differential inhibition of substrates varying in size. Multiple hits from previous high‐throughput screening for ATX inhibitors were obtained with aromatic sulfonamide derivatives interacting with the hydrophobic pocket. Here, we describe the development of a ligand‐based strategy and its application in virtual screening, which yielded novel high‐potency inhibitors that target the hydrophobic pocket of ATX. Characterization of the structure–activity relationship of these new inhibitors forms the foundation of a new pharmacophore model of the hydrophobic pocket of ATX.


Bioorganic & Medicinal Chemistry | 2013

Autotaxin inhibition: development and application of computational tools to identify site-selective lead compounds.

Derek D. Norman; Ayolah Ibezim; Whitney E. Scott; Stanley White; Daniel L. Baker

Autotaxin (ATX) catalyzes the conversion of lysophosphatidyl choline (LPC) to lysophosphatidic acid (LPA). Both ATX and LPA have been linked to pathophysiologies ranging from cancer to neuropathic pain. Inhibition of LPA production by ATX is therefore of therapeutic interest. Here we report the application of previously-developed, subsite-targeted pharmacophore models in a screening workflow that involves either docking or binary QSAR as secondary filters to identify ATX inhibitors from previously unreported structural types, four of which have sub-micromolar inhibition constants. Cell-based assays demonstrate that ATX inhibition and cytotoxicity structure-activity-relationships (SAR) exhibit selectivity cliffs, characterized by structurally similar compounds exhibiting similar biological activities with respect to ATX inhibition but very different biological activities with respect to cytotoxicity. Thus, general cytotoxicity should not be used as an early filter to eliminate candidate ATX inhibitor scaffolds from further SAR studies. Assays using two substrates of vastly different sizes demonstrate that the tools developed to identify compounds binding outside the central core of the active site did identify compounds acting at an allosteric site. In contrast, tools developed to identify active-site directed compounds did not identify active-site directed compounds. The stronger volume overlap imposed when selecting screening candidates expected to bind outside the active site is likely responsible for the stronger match between intended and actual target site.

Collaboration


Dive into the Derek D. Norman's collaboration.

Top Co-Authors

Avatar

Gabor Tigyi

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Duane D. Miller

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Sue Chin Lee

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Andrea Balogh

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James I. Fells

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Erzsebet Szabo

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Louisa Balazs

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Renukadevi Patil

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Ryoko Tsukahara

University of Tennessee Health Science Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge