U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Probe Reports from the NIH Molecular Libraries Program [Internet]. Bethesda (MD): National Center for Biotechnology Information (US); 2010-.

Cover of Probe Reports from the NIH Molecular Libraries Program

Probe Reports from the NIH Molecular Libraries Program [Internet].

Show details

Characterization of an agonist probe for opioid receptor mu 1 (OPRM1)-opioid receptor delta 1 (OPRD1) heterodimerization

, , , , , , , , , , , , , and .

Author Information and Affiliations

Received: ; Last Update: April 5, 2013.

Opiates such as morphine are the choice analgesic in the treatment of chronic pain due to their potent and rapid action. Opioid receptors belong to the family of G protein-coupled receptors (GPCRs), one of the largest gene families in the mammalian genome. The OPRM1 gene encodes the mu opioid receptor, which is the primary site of action for morphine and other commonly used opioids such as heroin, fentanyl, and methadone. The long-term use of opiates is limited because of the development of tolerance and dependence, as well as respiratory suppression and constipation. Due to their clinical importance, various strategies have been considered for making opiates more effective while curbing liabilities such as addiction. One such strategy has been to use a combination of drugs to improve the effectiveness of morphine. In particular, delta opioid receptor (OPRD1) ligands have been useful in enhancing morphine’s potency, but the underlying molecular basis is not understood. It has been shown that modulation of receptor function by physical association between OPRM1 and OPRD1 is a potential mechanism; heteromerization of OPRM1 with OPRD1 leads to the modulation of receptor binding and signaling properties. It has further been shown that the selective activation of the OPRM1-OPRD1 heteromer by a combination of OPRM1 agonist with OPRD1 antagonist can be blocked by antibodies that selectively recognize the heteromer. Therefore, the identification of compounds that selectively activate OPRM1-OPRD1 heterodimerization may have potential in the treatment of pain and alleviate unwanted effects associated with opiate use. The Scripps Research Institute Molecular Screening Center (SRIMSC), part of the Molecular Libraries Probe Production Centers Network (MLPCN), reports here an agonist for OPRM1-OPRD1 heterdimerization, ML335, with an EC50 of 403 nM, and selectivities vs. OPRM1, OPRD1, and HTR5A of 37, 2.7, and >99, respectively.

Assigned Assay Grant #: R03NS053751

Screening Center Name & PI: The Scripps Research Institute Molecular Screening Center (SRIMSC), H Rosen

Chemistry Center Name & PI: SRIMSC, H Rosen

Assay Submitter & Institution: Lakshmi Devi, Mount Sinai School of Medicine

PubChem Summary Bioassay Identifier (AID): 504355

Probe Structure and Characteristics

ML335.

ML335

CID/ML#Target NameTarget EC50 (nM) [SID, AID]Anti-target Name(s)Anti-target EC50 (μM) [SID, AID]Fold SelectiveSecondary Assay(s) Name: [SID, AID]
CID 23723457/ML335OPRM1-OPRD1403 [SID 134220670, AID 651685]HTR5A>40 [SID 134220670, AID 651688]>99Activation Assay: [SID 134220670, AID 651685]
Selectivity Assay HTR5A: [SID 134220670, AID 651688]
Selectivity Assay OPRD1: [SID 134220670, AID 651686]
Selectivity Assay OPRM1: [SID 134220670, AID 651689]
Cytotoxicity Assay: [SID 134220670, AID 651904]
OPRD11.1 [SID 134220670, AID 651686]2.7
OPRM114.9 [SID 134220670, AID 651689]37

1. Recommendations for Scientific Use of the Probe

Opiates such as morphine are the choice analgesic in the treatment of chronic pain. However their long-term use is limited because of the development of tolerance and dependence. Due to its importance in therapy, different strategies have been considered for making opiates such as morphine more effective while curbing its liability to be abused. One such strategy has been to use a combination of drugs to improve the effectiveness of morphine. In particular, OPRD1 opioid receptor ligands have been useful in enhancing morphine’s potency. The underlying molecular basis for these observations is not understood. The modulation of receptor function by physical association between OPRM1 and OPRD1 opioid receptors has been postulated as a potential mechanism. In support of this hypothesis, OPRM1-OPRD1 interacting complexes have been shown to exist in live cells and native membranes and the occupancy of OPRD1 receptors (by antagonists) is sufficient to enhance OPRM1 opioid receptor binding and signaling activity. Furthermore, OPRD1 receptor antagonists enhance morphine-mediated intrathecal analgesia. Thus heterodimeric associations between OPRM1-OPRD1 opioid receptors can be used as a model for the development of novel combination therapies for the treatment of chronic pain and other pathologies.

Accumulating evidence indicates that the majority of GPCRs form constitutive dimers in the endoplasmic reticulum and that dimerization is necessary for GPCR trafficking to the cell surface. Association between receptors from two distinct GPCR subfamilies has also been shown to lead to changes in receptor pharmacology. A growing number of studies point towards a role for GPCR heterodimerization in modulating receptor pharmacology and suggest that heterodimers could represent a functional unit, distinct from their cognate protomers. In essence, the association of two receptor molecules would allow the allosteric modulation of one monomeric unit by the other. The resulting cooperativity could be negative, neutral or positive, often resulting in changes in observed receptor pharmacology. This would be an extension of the concept of allosteric modulation of GPCRs by small molecules. This probe will be useful for investigations into the biology of OPRM1-OPRD1 heterodimers.

2. Materials and Methods

Chemistry

Solvents for extraction were ACS grade. Solvents for reaction were reagent grade. Unless otherwise noted, chemistry reagents were from Alfa Aesar, Fisher and Aldrich; highest quality available. TLC: silica gel 60 F254 aluminum plates, (Whatman, type Al Sil G/UV, 250 μm layer); visualization by UV absorption. Flash column chromatography (CC) was performed on silica gel 60 (0.40–0.63 mm, 230–440 mesh, EM Science). Biotage Flash+ systems were used for medium-pressure column chromatography. NMR: 1H and 13C spectra were obtained on a Bruker DRX-600 MHz spectrometer. Chemical shifts are given as δ values (ppm) using trimethylsilane as the internal standard. Reactions were monitored by LC/MS Shimadzu 210A.

2.1. Assays

Probe Characterization Assays

Solubility in PBS: The solubility of compounds are tested in triplicate in phosphate buffered saline (PBS), pH 7.4. per well, 198 μl PBS is added to a Millipore Solvinert Hydrophilic PTFE 96 well filter plate: pore size: 0.45um (MSRLN0450). Test compounds are introduced from 10 mM DMSO stock solutions (2 μl). The final concentration of DMSO was 1 percent. Samples are allowed to incubate at 22ºC for 18 hours. In the morning the plate is centrifuged where the soluble portion passes through the filter and is collected in a capture plate. Clotrimazole is included as a control to assure the assay is working properly. The samples are analyzed by HPLC. Peak area is compared to a standard of known concentration. In cases when the concentration was too low for UV analysis or when the compound did not possess a good chromophore, LC-MS-MS analysis is used.

Solubility in Media: The solubility of compounds are tested in triplicate in complete media (Dulbecco’s Modified Eagle Media [DMEM] plus 10% FBS). Per well, 198 μl PBS is added to a Millipore Solvinert Hydrophilic PTFE 96 well filter plate: pore size: 0.45um (MSRLN0450). Test compounds are introduced from 10 mM DMSO stock solutions (2 μl). The final concentration of DMSO was 1 percent. Samples are allowed to incubate at 22ºC for 18 hours. In the morning the plate is centrifuged where the soluble portion passes through the filter and is collected in a capture plate. The samples are analyzed by HPLC (Agilent 1100 with diode-array detector). Peak area is compared to a standard of known concentration. In cases when the concentration was too low for UV analysis or when the compound did not possess a good chromophore, LC-MS-MS analysis is used.

Stability in PBS: Demonstration of stability in PBS was conducted by addition of 10 μM compound from a DMSO stock to PBS in HPLC autosampler vials. Samples are held in the HPLC autosampler at ambient temperature. At approximately 0, 1, 2, 4, 8, 24, and 48 hours the samples are injected on the HPLC. Peak area and retention time are compared between injections. Data are log transformed and represented as half-life. DMSO is added as a co-solvent as needed for solubility.

Determination of Glutathione reactivity: One μL of a 10 mM compound stock solution was added to 1 mL of a freshly prepared solution of 50 μM reduced glutathione (GSH). Final compound concentration is 10 μM unless limited by solubility. The solution was allowed to incubate at 37°C for 6 hours prior to being directly analyzed for glutathione adduct formation. LC-MS/MS analysis of GSH adducts was performed on an API 4000 Q-TrapTM mass spectrometer equipped with a Turboionspray source (Applied Biosystems, Foster City, CA). Two methodologies were utilized: a negative precursor ion (PI) scan of m/z 272, corresponding to GSH fragmenting at the thioether bond, and a neutral loss scan of −129 AMU to detect GSH adducts. This triggered positive ion enhanced resolution and enhanced product ion scans (4243).

Primary Ultrahigh-Throughput Screening (uHTS) Assays

Primary uHTS assay to identify agonists of OPRD1-OPRM1 heterodimerization (AID 504326, AID 504904, and AID 588407)

Assay Overview: The purpose of this assay is to identify compounds that activate heterodimer formation between the OPRM1 and OPRD1 opioid receptors, resulting in membrane recruitment of beta-arrestin. The assay monitors GPCR-beta-arrestin proximity using low affinity fragment complementation of beta-galactosidase (beta-gal). This assay employs U2OS cells which express OPRD1, OPRM1 fused to a beta-gal peptide fragment (enzyme donor), and beta-arrestin fused to the complementary beta-gal fragment (enzyme acceptor). Cells are incubated with test compound, followed by measurement of well luminescence. As designed, compounds that induce formation of OPRD1 homodimers or OPRM1-OPRD1 heterodimers will cause beta-arrestin recruitment, resulting in reconstitution of the beta-gal holoenzyme. The reconstituted holoenzyme can then catalyze the hydrolysis of a substrate which yields a chemiluminescent signal, resulting in increased well luminescence. Deltorphin B is used as the high control for agonists, and wells containing cells treated with DMSO are used as the low control. Compounds were tested in singlicate (AID 504326) or triplicate (AID 504904) at a final nominal concentration of 9.3 μM, or in triplicate using a 10-point, 1:3 dilution series, starting at a nominal test concentration of 93 μM (AID 588407).

Protocol Summary:The U2OS-OPRM1-OPRD1 cell line was routinely cultured in T-175 sq cm flasks at 37°C and 95% relative humidity (RH). The growth media consisted of a 1:1 mixture of Ham’s F-12 Nutrient Media (F-12) and Dulbecco’s Modified Eagle Media (DMEM) supplemented with 10% v/v heat-inactivated certified fetal bovine serum, 25 mM HEPES, 250 μg/mL geneticin, 250 μg/mL hygromycin B, 0.25 μg/mL puromycin and 1X antibiotic mix (penicillin, streptomycin, and neomycin). The day before the assay, 1000 cells in 3 μL of cell plating media were seeded into each well of 1536 well microtiter plates and allowed to incubate at 37°C, 5% CO2, and 95% RH for 23 hours. Next, 28 nL of test compound in DMSO, Deltorphin B (0.9 μM final concentration) in DMSO, or DMSO alone were dispensed to the appropriate wells. The plates were then incubated for 3 hours at 37°C, 5% CO2, and 95% RH. The assay was started by adding 2 μL of PathHunter™ reagent (prepared according to the manufacturer’s protocol); followed by 1 hour incubation at room temperature. Well luminescence was then read on the ViewLux plate reader. Assay Cutoff: Compounds that exhibited ≥18.04% activation (AIDs 504326 and 504904) or an EC50 of <10 μM (AID 588407) were considered active.

Counterscreen uHTS assay to identify agonists of HTR5A (AID 504692, AID 504905 and AID 588408)

Assay Overview: The purpose of this assay is to identify compounds that activate the human serotonin receptor HTR5A, resulting in membrane recruitment of beta-arrestin. The assay monitors GPCR-beta-arrestin proximity using low affinity fragment complementation of beta-gal. This assay employs U2OS cells which express HTR5A fused to a beta-gal peptide fragment (enzyme donor), and beta-arrestin fused to the complementary beta-gal fragment (enzyme acceptor). Cells are incubated with test compound, followed by measurement of well luminescence. As designed, compounds that activate HTR5A will cause beta-arrestin recruitment, resulting in reconstitution of the beta-gal holoenzyme. The reconstituted holoenzyme can then catalyze the hydrolysis of a substrate which yields a chemiluminescent signal, resulting in increased well luminescence. Serotonin (5HT) is used as the high control for agonists, and wells containing cells treated with DMSO are used as the low control. Compounds were tested in singlicate (AID 504692) or triplicate (AID 504905) at a final nominal concentration of 9.3 μM, or in triplicate using a 10-point, 1:3 dilution series, starting at a nominal test concentration of 93 μM (AID 588408).

Protocol Summary: The U2OS-HTR5A cell line was routinely cultured in T-175 sq cm flasks at 37°C and 95% RH. The growth media consisted of a 1:1 mixture of Ham’s F-12 Nutrient Media (F-12) and DMEM supplemented with 10% v/v heat-inactivated certified fetal bovine serum, 25 mM HEPES, 250 ug/mL geneticin, 250 ug/mL hygromycin B, and 1X antibiotic mix (penicillin, streptomycin, and neomycin). The day before the assay, 1000 cells in 3 μL of cell plating media were seeded into each well of 1536 well microtiter plates and allowed to incubate at 37°C, 5% CO2, and 95% RH for 23 hours. Next, 28 nL of test compound in DMSO, or DMSO alone were dispensed to the appropriate wells. The assay was performed as described above (AID 504326, AID 504904, and AID 588407). Assay Cutoff: Compounds that exhibited ≥18.04% activation (AID 504692 and AID 504905) or an EC50 of <10 μM (AID 588408) were considered active.

Counterscreen uHTS assay to identify agonists of OPRD1 (AID 588411)

Assay Overview: The purpose of this counterscreen assay is to identify compounds that are agonists of OPRD1 homodimerization, resulting in membrane recruitment of beta-arrestin. This assay employs U2OS cells which express OPRD1 fused to a beta-gal peptide fragment (enzyme donor), and beta-arrestin fused to the complementary beta-gal fragment (enzyme acceptor). Cells are incubated with test compound, followed by measurement of well luminescence. As designed, compounds that induce formation of OPRD1 homodimers will cause beta-arrestin recruitment, resulting in reconstitution of the beta-gal holoenzyme. The reconstituted holoenzyme can then catalyze the hydrolysis of a substrate which yields a chemiluminescent signal, resulting in increased well luminescence. Compounds were tested in triplicate using a 10-point, 1:3 dilution series, starting at a nominal test concentration of 93 μM. Assay Cutoff: Compounds that exhibited an EC50 of <10 μM were considered active.

Protocol Summary: The U2OS-OPRD1 cell line was routinely cultured as described above (AID 504692, AID 504905 and AID 588408). The day before the assay, 1000 cells in 3 μL of cell plating media were seeded into each well of 1536 well microtiter plates and allowed to incubate at 37°C, 5% CO2, and 95% RH for 23 hours. Next, 28 nL of test compound in DMSO, Deltorphin B (0.9 μM final concentration) in DMSO, or DMSO alone were dispensed to the appropriate wells. The assay was performed as described above (AID 504326, AID 504904, and AID 588407). Assay Cutoff: Compounds that exhibited an EC50 of <10 μM were considered active.

Counterscreen uHTS assay to identify agonists of OPRM1 (AID 588435)

Assay Overview: The purpose of this counterscreen assay is to identify compounds that are agonists of OPRM1 homodimerization, resulting in membrane recruitment of beta-arrestin. The assay monitors GPCR-beta-arrestin proximity using low affinity fragment complementation of beta-gal. This assay employs U2OS cells which express OPRM1 fused to a beta-gal peptide fragment (enzyme donor), and beta-arrestin fused to the complementary beta-gal fragment (enzyme acceptor). Cells are incubated with test compound, followed by measurement of well luminescence. As designed, compounds that induce formation of OPRM1 homodimers will cause beta-arrestin recruitment, resulting in reconstitution of the beta-gal holoenzyme. The reconstituted holoenzyme can then catalyze the hydrolysis of a substrate which yields a chemiluminescent signal, resulting in increased well luminescence. Compounds were tested in triplicate using a 10-point, 1:3 dilution series, starting at a nominal test concentration of 93 μM.

Protocol Summary: The U2OS-OPRM1 cell line was routinely cultured as described above (AIDs 504692, 504905 and 588408). The day before the assay, 1000 cells in 3 μL of cell plating media were seeded into each well of 1536 well microtiter plates and allowed to incubate at 37°C, 5% CO2, and 95% RH for 23 hours. Next, 28 nL of test compound in DMSO, DAMGO (9.2 μM final concentration) in DMSO, or DMSO alone were dispensed to the appropriate wells. The assay was performed as described above (AID 504326, AID 504904, and AID 588407). Assay Cutoff: Compounds that exhibited an EC50 of <10 μM were considered active.

Secondary Assays

Assay to identify activators of OPRM1 and OPRD1 heterodimer formation (AID 651685, AID 651762)

Assay Overview: The purpose of this assay is to test powder samples of compounds that activate heterodimer formation between the OPRM1 and OPRD1 opioid receptors, resulting in membrane recruitment of beta-arrestin. The assay monitors GPCR-beta-arrestin proximity using low affinity fragment complementation of beta-gal. This assay employs U2OS cells which express OPRD1, OPRM1 fused to a beta-gal peptide fragment (enzyme donor), and beta-arrestin fused to the complementary beta-gal fragment (enzyme acceptor). Cells are incubated with test compound, followed by measurement of well luminescence. As designed, compounds that induce formation of OPRD1 homodimers or OPRM1-OPRD1 heterodimers will cause beta-arrestin recruitment, resulting in reconstitution of the beta-gal holoenzyme. The reconstituted holoenzyme can then catalyze the hydrolysis of a substrate which yields a chemiluminescent signal, resulting in increased well luminescence. Deltorphin B is used as the high control for agonists, and wells containing cells treated with DMSO are used as the low control. Compounds are tested in triplicate in a 10-point dilution series starting at a nominal concentration of 40 μM.

Protocol Summary: The U2OS-OPRM1-OPRD1 cell line was routinely cultured as described above (AID 504326, AID 504904, and AID 588407). The day before the assay 1000 cells in 20 μL of cell plating media were seeded into each well of 384 well plates and allowed to incubate at 37°C, 5% CO2, and 95% RH for 23 hours. Next, 100 nL of test compound in DMSO, Deltorphin B (1 μM final concentration) in DMSO, or DMSO alone were dispensed to the appropriate wells. The assay was performed as described above (AID 504326, AID 504904, and AID 588407). Assay Cutoff: Compounds that exhibited an EC50 of <10 μM were considered active.

Counterscreen assay to identify activators of OPRM1 homodimer formation (AID 651689, AID 651769)

Assay Overview: The purpose of this counterscreen assay is to test powder samples of compounds that are agonists of OPRM1-OPRD1 heterodimerization for agonist activity of OPRM1 homodimerization, resulting in membrane recruitment of beta-arrestin. The assay monitors GPCR-beta-arrestin proximity using low affinity fragment complementation of beta-gal. This assay employs U2OS cells which express OPRM1 fused to a beta-gal peptide fragment (enzyme donor), and beta-arrestin fused to the complementary beta-gal fragment (enzyme acceptor). Cells are incubated with test compound, followed by measurement of well luminescence. As designed, compounds that induce formation of OPRM1 homodimers will cause beta-arrestin recruitment, resulting in reconstitution of the beta-gal holoenzyme. The reconstituted holoenzyme can then catalyze the hydrolysis of a substrate which yields a chemiluminescent signal, resulting in increased well luminescence. DAMGO is used as the high control for agonists, and wells containing cells treated with DMSO is used as the low control. Compounds are tested in triplicate in a 10-point dilution series starting at a nominal concentration of 40 μM.

Protocol Summary: The U2OS-OPRM1 cell line was routinely cultured as described above (AID 504326, AID 504904, and AID 588407). The day before the assay, 5000 cells in 20 μL of cell plating media were seeded into each well of 384 well plates and allowed to incubate at 37°C, 5% CO2, and 95% RH for 23 hours. Next, 100 nL of test compound in DMSO, DAMGO (10 μM final concentration) in DMSO, or DMSO alone were dispensed to the appropriate wells. The assay was performed as described above (AID 504326, AID 504904, and AID 588407). Assay Cutoff: Compounds that exhibited an EC50 of <10 μM were considered active.

Counterscreen assay to identify activators of OPRD1 homodimer formation (AID 651686, AID 651764)

Assay Overview: The purpose of this counterscreen assay is to test powder samples of compounds that are agonists of OPRM1-OPRD1 heterodimerization for agonist activity of OPRD1 homodimerization, resulting in membrane recruitment of beta-arrestin. This assay employs U2OS cells which express OPRD1 fused to a beta-gal peptide fragment (enzyme donor), and beta-arrestin fused to the complementary beta-gal fragment (enzyme acceptor). Cells are incubated with test compound, followed by measurement of well luminescence. As designed, compounds that induce formation of OPRD1 homodimers will cause beta-arrestin recruitment, resulting in reconstitution of the beta-gal holoenzyme. The reconstituted holoenzyme can then catalyze the hydrolysis of a substrate which yields a chemiluminescent signal, resulting in increased well luminescence. Deltorphin B is used as the high control for agonists, and wells containing cells treated with DMSO are used as the low control. Compounds are tested in triplicate in a 10-point dilution series starting at a nominal concentration of 40 μM.

Protocol Summary: The U2OS-OPRD1 (Delta) cell line was routinely cultured as described above (AID 504326, AID 504904, and AID 588407). The day before the assay, 5000 cells in 20 μL of cell plating media were seeded into each well of 384 well plates and allowed to incubate at 37°C, 5% CO2, and 95 % RH for 23 hours. Next, 100 nL of test compound in DMSO, Deltorphin B (1 μM final concentration) in DMSO, or DMSO alone were dispensed to the appropriate wells. The assay was performed as described above (AIDs 504326, 504904, and 588407). Assay Cutoff: Compounds that exhibited an EC50 of <10 μM were considered active.

Counterscreen assay to identify activators of HTR5A (AID 651688, AID 651765)

Assay Overview: The purpose of this counterscreen assay is to test powder samples of compounds that are agonists of OPRM1-OPRD1 heterodimerization for activation of the human serotonin receptor HTR5A, resulting in membrane recruitment of beta-arrestin. The assay monitors GPCR-beta-arrestin proximity using low affinity fragment complementation of beta-gal. This assay employs U2OS cells which express HTR5A fused to a beta-gal peptide fragment (enzyme donor), and beta-arrestin fused to the complementary beta-gal fragment (enzyme acceptor). Cells are incubated with test compound, followed by measurement of well luminescence. As designed, compounds that activate HTR5A will cause beta-arrestin recruitment, resulting in reconstitution of the beta-gal holoenzyme. The reconstituted holoenzyme can then catalyze the hydrolysis of a substrate which yields a chemiluminescent signal, resulting in increased well luminescence. Serotonin is used as the high control for agonists, and wells containing cells treated with DMSO is used as the low control. Compounds were tested in singlicate at a final nominal concentration of 9.3 μM. Compounds were tested in triplicate in a 10-point dilution series starting at a nominal concentration of 40 μM.

Protocol Summary: The U2OS-HTR5A cell line was routinely cultured as described above (AID 504326, AID 504904, and AID 588407). The day before the assay, 5000 cells in 20 μL of cell plating media were seeded into each well of 384 well plates and allowed to incubate at 37°C, 5% CO2, and 95% RH for 23 hours. Next, 100 nL of test compound in DMSO, serotonin (5-CT) (3 μM final concentration) in DMSO, or DMSO alone were dispensed to the appropriate wells. The assay was performed as described above (AID 504326, AID 504904, and AID 588407). Assay Cutoff: Compounds that exhibited an EC50 of <10 μM were considered active.

Cytotoxicity assay to identify cytotoxic compounds (AID 651904)

Assay Overview: The purpose of this assay is to determine cytotoxicity of powder compounds that activate heterodimer formation between the OPRM1 and OPRD1 opioid receptors. In this assay, U2OS cells are incubated with test compound, followed by determination of cell viability. The assay utilizes the CellTiter-Glo luminescent reagent to measure intracellular ATP in viable cells. Luciferase present in the reagent catalyzes the oxidation of beetle luciferin to oxyluciferin and light in the presence of cellular ATP. Well luminescence is directly proportional to ATP levels and cell viability. As designed, compounds that reduce cell viability will reduce ATP levels, luciferin oxidation and light production, resulting in decreased well luminescence. Compounds were tested in quadruplicate in a 13-point 1:3 dilution series starting at a nominal test concentration of 10 uM.

Protocol Summary: This assay was started by dispensing PathHunter™ OPRM1/OPRD1-β-Arrestin U2OS cells in McCoy’s 5A medium plus 10% FBS, penicillin 100 U/mL and streptomycin 100 μg/mL (20 μL, 4 × 1000 cells/well) into the wells of a 384-well plate. Thirteen 1:3 serial dilutions of compound (100 μM in growth media) were made. 5 μL of diluted compound or media were added to wells, giving final compound concentrations of 0–10 μM. The plate was incubated at 37°C in a humidified incubator for 24 hours, then equilibrated to room temperature for 30 minutes. 25 μL CellTitre-Glo reagent was added to each well, followed by incubation of the plate in the dark for 10 minutes. Well luminescence was measured on the Envision plate reader. Assay Cutoff: Compounds that exhibited a CC50 (defined as the concentration of compound that induces 50% cell death) of >10 μM were considered non-cytotoxic.

2.2. Probe Chemical Characterization

CID 23723457 SID 134220670 ML335.

CID 23723457
SID 134220670
ML335

The probe structure was verified by 1H and 13C NMR (see Section 2.3) and high resolution LC/MS (Figure 1). Purity was assessed to be greater than 95% by LC-MS and NMR.

Figure 1. LC/MS analysis for ML335.

Figure 1

LC/MS analysis for ML335.

Solubility (at room temperature) for ML335 in PBS and in DMEM plus 10% FBS media was determined to be 58.6 μM and 66.1 μM, respectively. ML335 has a half-life of >48 hours in PBS at room temperature (94.6% compound remaining at 48 hours) (Figure 2).

Figure 2. Stability of ML335 (CYM51010) in PBS.

Figure 2

Stability of ML335 (CYM51010) in PBS.

No Michael acceptor adducts were observed when a sample of the probe was incubated with 50 μM glutathione and analyzed by LC-MS.

The following compounds have been submitted to the SMR collection (Table 1).

Table 1. Compounds submitted to the SMR collection (12-17-2012).

Table 1

Compounds submitted to the SMR collection (12-17-2012).

2.3. Probe Preparation

Figure 3. Synthetic scheme for ML335.

Figure 3Synthetic scheme for ML335

To a stirred solution of LDA (2M) (4.22 mmol, 1.09 equiv.) in THF at −78°C was added dropwise a solution of 1 (3.88 mmol, 1 equiv.) in THF (8 mL) and the reaction was stirred for 1 hour at −78°C. A solution of 2 (4.03 mmol, 1.04 equiv.) in anhydrous THF (8 mL) was added over a period of 30 minutes. The reaction mixture was slowly warmed to room temperature and the reaction was stirred for an additional 8 hours. The mixture was poured onto ice-water and the product extracted with ethyl acetate (3X). The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The product was purified by flash column chromatography using hexanes/ethyl acetate. Product 3 was obtained as an oil in 35% yield (1.36 mmol, 490 mg). MS (EI) m/z: 362 (M+).

To a stirred solution of 3 (1.1 mmol, 1 equiv.) in CH2Cl2, TFA (22.1 mmol, 20 equiv.) was added at room temperature and the reaction mixture was stirred for 30 minutes. The mixture was concentrated under reduced pressure. The crude was diluted in water and slowly basified with NaOH (1M) (~pH 12). The product was extracted using ethyl acetate (5X) and the organic phase dried over anhydrous sodium sulfate and concentrated under reduced pressure. The product was used without further purification.

A mixture of the crude (0.057 mmol, 1 equiv.) chloride 4 (0.063 mmol, 1.1 equiv.) and sodium carbonate (0.063 mmol, 1.1 equiv.) in acetonitrile was stirred at room temperature overnight. The mixture was quenched with silica gel and the product purified by flash column chromatography using CH2Cl2/MeOH. The product was obtained as yellow oil in 60% yield (13.7 mg, 0.034mmol) and judged to be >95% pure by LC-MS (Figure 1).

1H NMR (600 MHz, CD3OD): δ 7.51 (d, J = 8.4 Hz, 2H), 7.26 (d, J = 8.4 Hz, 2H), 7.22 (d, J = 7.5 Hz, 2H), 7.15-7.10 (m, 3H), 4.15 (q, J = 7.12 Hz, 2H), 3.47 (s, 2H), 2.77-2.44 (m, 2H), 2.50-2.46 (m, 2H), 2.21-2.11 (m, 4H), 2.11 (s, 3H), 1.81-1.76 (m, 2H), 1.59-1.52 (m, 2H), 1.26 (t, J = 7.12 Hz, 3H); 13C NMR (150 MHz, CDCl3): δ 174.12, 169.68, 140.58 (2C), 140.16, 131.64, 128.52, 128.21, 126.24, 120.27, 61.47, 60.53, 50.04, 44.19, 41.91, 30.54, 30.31, 24.26, 14.27 MS (EI) m/z: 409 (M+H). HRMS (EI) for C25H33N2O3 (M+H): calcd 409.2486, found 409.2481.

3. Results

3.1. Dose Response Curves for Probe

Figure 4 shows the dose response curve for binding of ML335 to U2OS cells which express OPRD1 and OPRM1.

Figure 4. IC50 curve for probe ML335 determined in assay AID 651685.

Figure 4

IC50 curve for probe ML335 determined in assay AID 651685.

3.2. Cellular Activity

ML335 has been evaluated in a series of cell-based assays (the OPRM1-OPRD1 screen, the OPRM1, OPRD1, 5HTRA counterscreens, and the cytotoxicity assay) and has been shown to have activity in a cell-based system.

3.3. Profiling Assays

To date, ML335 has been tested in 438 bioassays deposited in PubChem, and has shown activity in 10 of those assays, four of which are for this project. The other six assays give a hit rate of 1.4%, indicating that this molecule is generally inactive across a broad range of cell-based and non–cell-based assays. Five of the six cross-reactive assays are for transient receptor potential cation channel (TRPC) 4 and C6. These assays are performed with HEK cells expressing both mu opioid and TRPC. It is likely the cross-reactivity is due to activity on the mu receptor. The remaining assay (AID 504834) is “Primary qHTS for delayed death inhibitors of the malarial parasite plastid, 96 hour incubation”. It is likely the probe is active (4.1 μM) in this assay, but the species and dosing are not relevant to the current project.

4. Discussion

4.1. Comparison to Existing Art and How the New Probe is an Improvement

There are currently no known potent and selective small molecule agonists of OPRM1-OPRD1 heterodimer formation.

5. References

1.
Schellekens GA, de Jong BA, van den Hoogen FH, van de Putte LB, van Venrooij WJ. Citrulline is an essential constituent of antigenic determinants recognized by rheumatoid arthritis-specific autoantibodies. J Clin Invest. 1998;101:273–281. [PMC free article: PMC508564] [PubMed: 9421490]
2.
Chen CC, Isomoto H, Narumi Y, Sato K, Oishi Y, Kobayashi T, Yanagihara K, Mizuta Y, Kohno S, Tsukamoto K. Haplotypes of PADI4 susceptible to rheumatoid arthritis are also associated with ulcerative colitis in the Japanese population. Clin Immunol. 2008;126:165–171. [PubMed: 17980669]
3.
Moscarello MA, Mastronardi FG, Wood DD. The role of citrullinated proteins suggests a novel mechanism in the pathogenesis of multiple sclerosis. Neurochem Res. 2007;32:251–256. [PMC free article: PMC1794624] [PubMed: 17031564]
4.
Ishigami A, Ohsawa T, Hiratsuka M, Taguchi H, Kobayashi S, Saito Y, Murayama S, Asaga H, Toda T, Kimura N, Maruyama N. Abnormal accumulation of citrullinated proteins catalyzed by peptidylarginine deiminase in hippocampal extracts from patients with Alzheimer’s disease. J Neurosci Res. 2005;80:120–128. [PubMed: 15704193]
5.
Anzilotti C, Pratesi F, Tommasi C, Migliorini P. Peptidylarginine deiminase 4 and citrullination in health and disease. Autoimmun Rev. 2010;9:158–160. [PubMed: 19540364]
6.
Chang X, Han J. Expression of peptidylarginine deiminase type 4 (PAD4) in various tumors. Mol Carcinog. 2006;45:183–196. [PubMed: 16355400]
7.
Chang X, Yamada R, Suzuki A, Sawada T, Yoshino S, Tokuhiro S, Yamamoto K. Localization of peptidylarginine deiminase 4 (PADI4) and citrullinated protein in synovial tissue of rheumatoid arthritis. Rheumatology (Oxford). 2005;44:40–50. [PubMed: 15466895]
8.
De Ceuleneer M, Van Steendam K, Dhaenens M, Deforce D. In vivo relevance of citrullinated proteins and the challenges in their detection. Proteomics. 2012;12:752–760. [PubMed: 22318877]
9.
Jones JE, Causey CP, Knuckley B, Slack-Noyes JL, Thompson PR. Protein arginine deiminase 4 (PAD4): Current understanding and future therapeutic potential. Curr Opin Drug Discov Devel. 2009;12:616–627. [PMC free article: PMC3771078] [PubMed: 19736621]
10.
Mohanan S, Cherrington BD, Horibata S, McElwee JL, Thompson PR, Coonrod SA. Potential role of peptidylarginine deiminase enzymes and protein citrullination in cancer pathogenesis. Biochem Res Int 2012. 2012:895343. [PMC free article: PMC3457611] [PubMed: 23019525]
11.
Vossenaar ER, Zendman AJ, van Venrooij WJ, Pruijn GJ. PAD, a growing family of citrullinating enzymes: genes, features and involvement in disease. Bioessays. 2003;25:1106–1118. [PubMed: 14579251]
12.
Wegner N, Lundberg K, Kinloch A, Fisher B, Malmstrom V, Feldmann M, Venables PJ. Autoimmunity to specific citrullinated proteins gives the first clues to the etiology of rheumatoid arthritis. Immunol Rev. 2010;233:34–54. [PubMed: 20192991]
13.
Auger I, Charpin C, Balandraud N, Martin M, Roudier J. Autoantibodies to PAD4 and BRAF in rheumatoid arthritis. Autoimmun Rev. 2012;11:801–803. [PubMed: 22349616]
14.
Knuckley B, Causey CP, Pellechia PJ, Cook PF, Thompson PR. Haloacetamidine-based inactivators of protein arginine deiminase 4 (PAD4): evidence that general acid catalysis promotes efficient inactivation. Chembiochem. 2010;11:161–165. [PMC free article: PMC3056394] [PubMed: 20014086]
15.
Luo Y, Arita K, Bhatia M, Knuckley B, Lee YH, Stallcup MR, Sato M, Thompson PR. Inhibitors and inactivators of protein arginine deiminase 4: functional and structural characterization. Biochemistry. 2006;45:11727–11736. [PMC free article: PMC1808342] [PubMed: 17002273]
16.
Luo Y, Knuckley B, Lee YH, Stallcup MR, Thompson PR. A fluoroacetamidine-based inactivator of protein arginine deiminase 4: design, synthesis, and in vitro and in vivo evaluation. J Am Chem Soc. 2006;128:1092–1093. [PMC free article: PMC1850713] [PubMed: 16433522]
17.
Li P, Yao H, Zhang Z, Li M, Luo Y, Thompson PR, Gilmour DS, Wang Y. Regulation of p53 target gene expression by peptidylarginine deiminase 4. Mol Cell Biol. 2008;28:4745–4758. [PMC free article: PMC2493360] [PubMed: 18505818]
18.
Yao H, Li P, Venters BJ, Zheng S, Thompson PR, Pugh BF, Wang Y. Histone Arg modifications and p53 regulate the expression of OKL38, a mediator of apoptosis. J Biol Chem. 2008;283:20060–20068. [PMC free article: PMC2459274] [PubMed: 18499678]
19.
Chumanevich AA, Causey CP, Knuckley BA, Jones JE, Poudyal D, Chumanevich AP, Davis T, Matesic LE, Thompson PR, Hofseth LJ. Suppression of colitis in mice by Cl-amidine: a novel peptidylarginine deiminase inhibitor. Am J Physiol Gastrointest Liver Physiol. 2011;300:G929–938. [PMC free article: PMC3119113] [PubMed: 21415415]
20.
McElwee JL, Mohanan S, Griffith OL, Breuer HC, Anguish LJ, Cherrington BD, Palmer AM, Howe LR, Subramanian V, Causey CP, Thompson PR, Gray JW, Coonrod SA. Identification of PADI2 as a potential breast cancer biomarker and therapeutic target. BMC cancer. 2012;12:500. [PMC free article: PMC3571905] [PubMed: 23110523]
21.
Wang Y, Li P, Wang S, Hu J, Chen XA, Wu J, Fisher M, Oshaben K, Zhao N, Gu Y, Wang D, Chen G. Anticancer peptidylarginine deiminase (PAD) inhibitors regulate the autophagy flux and the mammalian target of rapamycin complex 1 activity. J Biol Chem. 2012;287:25941–25953. [PMC free article: PMC3406678] [PubMed: 22605338]
22.
Kearney PL, Bhatia M, Jones NG, Luo Y, Glascock MC, Catchings KL, Yamada M, Thompson PR. Kinetic characterization of protein arginine deiminase 4: a transcriptional corepressor implicated in the onset and progression of rheumatoid arthritis. Biochemistry. 2005;44:10570–10582. [PubMed: 16060666]
23.
Imparl JM, Senshu T, Graves DJ. Studies of calcineurin-calmodulin interaction: probing the role of arginine residues using peptidylarginine deiminase. Arch Biochem Biophys. 1995;318:370–377. [PubMed: 7733665]
24.
Lamensa JW, Moscarello MA. Deimination of human myelin basic protein by a peptidylarginine deiminase from bovine brain. J Neurochem. 1993;61:987–996. [PubMed: 7689646]
25.
Tarcsa E, Marekov LN, Mei G, Melino G, Lee SC, Steinert PM. Protein unfolding by peptidylarginine deiminase. Substrate specificity and structural relationships of the natural substrates trichohyalin and filaggrin. J Biol Chem. 1996;271:30709–30716. [PubMed: 8940048]
26.
Chavanas S, Mechin MC, Takahara H, Kawada A, Nachat R, Serre G, Simon M. Comparative analysis of the mouse and human peptidylarginine deiminase gene clusters reveals highly conserved non-coding segments and a new human gene, PADI6. Gene. 2004;330:19–27. [PubMed: 15087120]
27.
Vossenaar ER, Nijenhuis S, Helsen MM, van der Heijden A, Senshu T, van den Berg WB, van Venrooij WJ, Joosten LA. Citrullination of synovial proteins in murine models of rheumatoid arthritis. Arthritis Rheum. 2003;48:2489–2500. [PubMed: 13130468]
28.
Ellsworth RE, Vertrees A, Love B, Hooke JA, Ellsworth DL, Shriver CD. Chromosomal alterations associated with the transition from in situ to invasive breast cancer. Ann Surg Oncol. 2008;15:2519–2525. [PubMed: 18622645]
29.
Chang X, Fang K. PADI4 and tumourigenesis. Cancer Cell International. 2010;10:7. [PMC free article: PMC2845578] [PubMed: 20222985]
30.
Cherrington BD, Morency E, Struble AM, Coonrod SA, Wakshlag JJ. Potential role for peptidylarginine deiminase 2 (PAD2) in citrullination of canine mammary epithelial cell histones. PLoS One. 2010;5:e11768. [PMC free article: PMC2909897] [PubMed: 20668670]
31.
Senshu T, Akiyama K, Nagata S, Watanabe K, Hikichi K. Peptidylarginine deiminase in rat pituitary: sex difference, estrous cycle-related changes, and estrogen dependence. Endocrinology. 1989;124:2666–2670. [PubMed: 2721440]
32.
Takahara H, Tsuchida M, Kusubata M, Akutsu K, Tagami S, Sugawara K. Peptidylarginine deiminase of the mouse. Distribution, properties, and immunocytochemical localization. J Biol Chem. 1989;264:13361–13368. [PubMed: 2753915]
33.
Darrah E, Rosen A, Giles JT, Andrade F. Peptidylarginine deiminase 2, 3 and 4 have distinct specificities against cellular substrates: novel insights into autoantigen selection in rheumatoid arthritis. Ann Rheum Dis. 2012;71:92–98. [PMC free article: PMC3302156] [PubMed: 21859690]
34.
Ishigami A, Asaga H, Ohsawa T, Akiyama K, Maruyama N. Peptidylarginine deiminase type I, type II, type III and type IV are expressed in rat epidermis. Biomed Res. 2001;22:63–65.
35.
Knuckley B, Causey CP, Jones JE, Bhatia M, Dreyton CJ, Osborne TC, Takahara H, Thompson PR. Substrate specificity and kinetic studies of PADs 1, 3, and 4 identify potent and selective inhibitors of protein arginine deiminase 3. Biochemistry. 2010;49:4852–4863. [PMC free article: PMC2884139] [PubMed: 20469888]
36.
Terakawa H, Takahara H, Sugawara K. Three types of mouse peptidylarginine deiminase: characterization and tissue distribution. J Biochem. 1991;110:661–666. [PubMed: 1778991]
37.
Watanabe K, Akiyama K, Hikichi K, Ohtsuka R, Okuyama A, Senshu T. Combined biochemical and immunochemical comparison of peptidylarginine deiminases present in various tissues. Biochim Biophys Acta. 1988;966:375–383. [PubMed: 3416014]
38.
Knuckley B, Bhatia M, Thompson PR. Protein arginine deiminase 4: evidence for a reverse protonation mechanism. Biochemistry. 2007;46:6578–6587. [PMC free article: PMC2212595] [PubMed: 17497940]
39.
Knuckley B, Luo Y, Thompson PR. Profiling Protein Arginine Deiminase 4 (PAD4): a novel screen to identify PAD4 inhibitors. Bioorg Med Chem. 2008;16:739–745. [PMC free article: PMC2267747] [PubMed: 17964793]
40.
Slack JL, Causey CP, Thompson PR. Protein arginine deiminase 4: a target for an epigenetic cancer therapy. Cell Mol Life Sci. 2011;68:709–720. [PMC free article: PMC3815436] [PubMed: 20706768]
41.
Luo Y, Knuckley B, Bhatia M, Pellechia PJ, Thompson PR. Activity-Based Protein Profiling Reagents for Protein Arginine Deiminase 4 (PAD4): Synthesis and in vitro Evaluation of a Fluorescently Labeled Probe. Journal of the American Chemical Society. 2006;128:14468–14469. [PMC free article: PMC2234596] [PubMed: 17090024]
42.
Knuckley B, Jones JE, Bachovchin DA, Slack J, Causey CP, Brown SJ, Rosen H, Cravatt BF, Thompson PR. A fluopol-ABPP HTS assay to identify PAD inhibitors. Chem Commun (Camb). 2010;46:7175–7177. [PMC free article: PMC2943038] [PubMed: 20740228]
43.
Bachovchin DA, Brown SJ, Rosen H, Cravatt BF. Identification of selective inhibitors of uncharacterized enzymes by high-throughput screening with fluorescent activity-based probes. Nature Biotechnology. 2009;27:387–394. [PMC free article: PMC2709489] [PubMed: 19329999]
44.
Jones JE, Slack JL, Fang P, Zhang X, Subramanian V, Causey CP, Coonrod SA, Guo M, Thompson PR. Synthesis and Screening of a Haloacetamidine Containing Library To Identify PAD4 Selective Inhibitors. ACS Chemical Biology. 2011;7:160–165. [PMC free article: PMC3262960] [PubMed: 22004374]
45.
Li X, He Y, Ruiz CH, Koenig M, Cameron MD, Vojkovsky T. Characterization of dasatinib and its structural analogs as CYP3A4 mechanism-based inactivators and the proposed bioactivation pathways. Drug Metab Dispos. 2009;37:1242–1250. [PMC free article: PMC3202349] [PubMed: 19282395]
46.
Leatherbarrow RJ. Grafit Ver 5.0. Staines, UK: Erathicus Software; 2004.
47.
Boger DL, Yasuda M, Mitscher LA, Drake SD, Kitos PA, Thompson SC. Streptonigrin and lavendamycin partial structures. Probes for the minimum, potent pharmacophore of streptonigrin, lavendamycin, and synthetic quinoline-5,8-diones. J Med Chem. 1987;30:1918–1928. [PubMed: 3656364]
48.
Corey EJ, Chaykovsky M. Dimethyloxosulfonium Methylide ((CH3)2SOCH2) and Dimethylsulfonium Methylide ((CH3)2SCH2). Formation and Application to Organic Synthesis. Journal of the American Chemical Society. 1965;87:1353–1364.
49.
Boger DL, Duff SR, Panek JS, Yasuda M. Inverse electron demand Diels-Alder reactions of heterocyclic azadienes. Studies on the total synthesis of lavendamycin: investigative studies on the preparation of the CDE .beta.-carboline ring system and AB quinoline-5,8-quinone ring system. The Journal of Organic Chemistry. 1985;50:5782–5789.
50.
Leung D, Hardouin C, Boger DL, Cravatt BF. Discovering potent and selective reversible inhibitors of enzymes in complex proteomes. Nat Biotechnol. 2003;21:687–691. [PubMed: 12740587]
51.
Weerapana E, Simon GM, Cravatt BF. Disparate proteome reactivity profiles of carbon electrophiles. Nature Chemical Biology. 2008;4:405–407. [PMC free article: PMC2440582] [PubMed: 18488014]
52.
Bachovchin DA, Zuhl AM, Speers AE, Wolfe MR, Weerapana E, Brown SJ, Rosen H, Cravatt BF. Discovery and Optimization of Sulfonyl Acrylonitriles as Selective, Covalent Inhibitors of Protein Phosphatase Methylesterase-1. Journal of Medicinal Chemistry. 2011;54:5229–5236. [PMC free article: PMC3144155] [PubMed: 21639134]
53.
Johnson DS, Weerapana E, Cravatt BF. Strategies for discovering and derisking covalent, irreversible enzyme inhibitors. Future Med Chem. 2010;2:949–964. [PMC free article: PMC2904065] [PubMed: 20640225]
54.
Kodadek T. Rethinking screening. Nat Chem Biol. 2010;6:162–165. [PubMed: 20154660]
55.
Potashman MH, Duggan ME. Covalent modifiers: an orthogonal approach to drug design. J Med Chem. 2009;52:1231–1246. [PubMed: 19203292]
56.
Singh J, Petter RC, Baillie TA, Whitty A. The resurgence of covalent drugs. Nat Rev Drug Discov. 2011;10:307–317. [PubMed: 21455239]
57.
Bachovchin DA, Mohr JT, Speers AE, Wang C, Berlin JM, Spicer TP, Fernandez-Vega V, Chase P, Hodder PS, Schurer SC, Nomura DK, Rosen H, Fu GC, Cravatt BF. Academic cross-fertilization by public screening yields a remarkable class of protein phosphatase methylesterase-1 inhibitors. Proc Natl Acad Sci U S A. 2011;108:6811–6816. [PMC free article: PMC3084096] [PubMed: 21398589]
58.
Vincent PW, Bridges AJ, Dykes DJ, Fry DW, Leopold WR, Patmore SJ, Roberts BJ, Rose S, Sherwood V, Zhou H, Elliott WL. Anticancer efficacy of the irreversible EGFr tyrosine kinase inhibitor PD 0169414 against human tumor xenografts. Cancer Chemother Pharmacol. 2000;45:231–238. [PubMed: 10663641]
59.
Robertson JG. Mechanistic Basis of Enzyme-Targeted Drugs. Biochemistry. 2005;44:5561–5571. [PubMed: 15823014]
60.
Dong S, Zhang Z, Takahara H. Estrogen-enhanced peptidylarginine deiminase type IV gene (PADI4) expression in MCF-7 cells is mediated by estrogen receptor-alpha-promoted transfactors activator protein-1, nuclear factor-Y, and Sp1. Mol Endocrinol. 2007;21:1617–1629. [PubMed: 17456793]
61.
Horibata S, Coonrod SA, Cherrington BD. Role for peptidylarginine deiminase enzymes in disease and female reproduction. J Reprod Dev. 2012;58:274–282. [PubMed: 22790870]
62.
Li P, Wang D, Yao H, Doret P, Hao G, Shen Q, Qiu H, Zhang X, Wang Y, Chen G. Coordination of PAD4 and HDAC2 in the regulation of p53-target gene expression. Oncogene. 2010;29:3153–3162. [PMC free article: PMC2913128] [PubMed: 20190809]
63.
Teo CY, Shave S, Chor AL, Salleh AB, Rahman MB, Walkinshaw MD, Tejo BA. Discovery of a new class of inhibitors for the protein arginine deiminase type 4 (PAD4) by structure-based virtual screening. BMC Bioinformatics. 2012;13 Suppl 17:S4. [PMC free article: PMC3521205] [PubMed: 23282142]
64.
Wei L, Wasilewski E, Chakka SK, Bello AM, Moscarello MA, Kotra LP. Novel Inhibitors of Protein Arginine Deiminase with Potential Activity in Multiple Sclerosis Animal Model. J. Med. Chem. 2013 Feb 19; [Epub ahead of print] [PubMed: 23421315]

Views

  • PubReader
  • Print View
  • Cite this Page

Related information

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...