140 results on '"Majumdar, Susruta"'
Search Results
102. Generation of novel radiolabeled opiates through site-selective iodination
- Author
-
Majumdar, Susruta, Burgman, Maxim, Haselton, Nathan, Grinnell, Steven, Ocampo, Julia, Pasternak, Anna Rose, and Pasternak, Gavril W
- Published
- 2011
- Full Text
- View/download PDF
103. ChemInform Abstract: Three-Component Coupling Approach for the Synthesis of Diverse Heterocycles Utilizing Reactive Nitrilium Trapping.
- Author
-
Varadi, Andras, primary, Palmer, Travis C., additional, Notis, Paula R., additional, Redel-Traub, Gabriel N., additional, Afonin, Daniel, additional, Subrath, Joan J., additional, Pasternak, Gavril W., additional, Hu, Chunhua, additional, Sharma, Indrajeet, additional, and Majumdar, Susruta, additional
- Published
- 2014
- Full Text
- View/download PDF
104. Pharmacologic Characterization in the Rat of a Potent Analgesic Lacking Respiratory Depression, IBNtxA
- Author
-
Grinnell, Steven G., primary, Majumdar, Susruta, additional, Narayan, Ankita, additional, Le Rouzic, Valerie, additional, Ansonoff, Michael, additional, Pintar, John E., additional, and Pasternak, Gavril W., additional
- Published
- 2014
- Full Text
- View/download PDF
105. Endomorphin analogs with analgesic actions mediated through exon 11 variants of the mu opioid receptor (MOR‐1) (659.10)
- Author
-
Marrone, Gina, primary, Grinnell, Stephen, additional, Pan, Ying‐Xian, additional, Pintar, John, additional, Majumdar, Susruta, additional, and Pasternak, Gavril, additional
- Published
- 2014
- Full Text
- View/download PDF
106. Buprenorphine analgesia requires exon 11‐associated mu opioid receptor splice variants (659.11)
- Author
-
Grinnell, Steven, primary, Majumdar, Susruta, additional, Ansonoff, Michael, additional, Pintar, John, additional, Pan, Ying‐Xian, additional, and Pasternak, Gavril, additional
- Published
- 2014
- Full Text
- View/download PDF
107. Three-Component Coupling Approach for the Synthesis of Diverse Heterocycles Utilizing Reactive Nitrilium Trapping
- Author
-
Váradi, András, primary, Palmer, Travis C., additional, Notis, Paula R., additional, Redel-Traub, Gabriel N., additional, Afonin, Daniel, additional, Subrath, Joan J., additional, Pasternak, Gavril W., additional, Hu, Chunhua, additional, Sharma, Indrajeet, additional, and Majumdar, Susruta, additional
- Published
- 2014
- Full Text
- View/download PDF
108. Cross‐species and Painless: Targeting truncated mu opioid receptor splice variants in the rat
- Author
-
Grinnell, Steven, primary, Majumdar, Susruta, additional, and Pasternak, Gavril W, additional
- Published
- 2013
- Full Text
- View/download PDF
109. Synthetic and Receptor Signaling Explorations of the Mitragyna Alkaloids: Mitragynine as an Atypical Molecular Framework for Opioid Receptor Modulators.
- Author
-
Kruegel, Andrew C., Gassaway, Madalee M., Kapoor, Abhijeet, Váradi, András, Majumdar, Susruta, Filizola, Marta, Javitch, Jonathan A., and Sames, Dalibor
- Published
- 2016
- Full Text
- View/download PDF
110. Isocyanide-Based Multicomponent Reactions for the Synthesis of Heterocycles.
- Author
-
Váradi, András, Palmer, Travis C., Dardashti, Rebecca Notis, and Majumdar, Susruta
- Subjects
ISOCYANIDES ,HETEROCYCLIC compounds ,NITRILIUM salts ,CHEMICAL synthesis ,DRUG design - Abstract
Multicomponent reactions (MCRs) are extremely popular owing to their facile execution, high atom-efficiency and the high diversity of products. MCRs can be used to access various heterocycles and highly functionalized scaffolds, and thus have been invaluable tools in total synthesis, drug discovery and bioconjugation. Traditional isocyanide-based MCRs utilize an external nucleophile attacking the reactive nitrilium ion, the key intermediate formed in the reaction of the imine and the isocyanide. However, when reactants with multiple nucleophilic groups (bisfunctional reactants) are used in the MCR, the nitrilium intermediate can be trapped by an intramolecular nucleophilic attack to form various heterocycles. The implications of nitrilium trapping along with widely applied conventional isocyanide-based MCRs in drug design are discussed in this review. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
111. Synthesis and Evaluation of Aryl-Naloxamide Opiate Analgesics Targeting Truncated Exon 11-Associated μ Opioid Receptor (MOR-1) Splice Variants
- Author
-
Majumdar, Susruta, primary, Subrath, Joan, additional, Le Rouzic, Valerie, additional, Polikar, Lisa, additional, Burgman, Maxim, additional, Nagakura, Kuni, additional, Ocampo, Julie, additional, Haselton, Nathan, additional, Pasternak, Anna R., additional, Grinnell, Steven, additional, Pan, Ying-Xian, additional, and Pasternak, Gavril W., additional
- Published
- 2012
- Full Text
- View/download PDF
112. Exploration of novel radioiodine‐labeling techniques for opioid peptides
- Author
-
Pickett, Julie E., primary, Majumdar, Susruta, additional, Burgman, Maxim, additional, and Pasternak, Gavril W., additional
- Published
- 2010
- Full Text
- View/download PDF
113. Soft Alkyl Ether Prodrugs of a Model Phenolic Drug: The Effect of Incorporation of Ethyleneoxy Groups on Transdermal Delivery
- Author
-
Thomas, Joshua, primary, Majumdar, Susruta, additional, and Sloan, Kenneth, additional
- Published
- 2009
- Full Text
- View/download PDF
114. N-Alkyl- N-alkyloxycarbonylaminomethyl (NANAOCAM) prodrugs of carboxylic acid containing drugs
- Author
-
Majumdar, Susruta and Sloan, Kenneth B.
- Published
- 2007
- Full Text
- View/download PDF
115. An efficient 1,3-allylic carbonyl transposition of chalcones
- Author
-
Khurana, Jitender M., primary, Dawra, Kiran, additional, and Majumdar, Susruta, additional
- Published
- 2008
- Full Text
- View/download PDF
116. α-(1H-Imidazol-1-yl)alkyl (IMIDA) carboxylic acid esters as prodrugs of carboxylic acid containing drugs
- Author
-
Majumdar, Susruta, primary, Spaeth, Maren Mueller, additional, Sivendran, Sashikala, additional, Juntunen, Juha, additional, Thomas, Joshua D., additional, and Sloan, K.B., additional
- Published
- 2007
- Full Text
- View/download PDF
117. Synthesis of soft alkyl phenolic ether prodrugs using Mitsunobu chemistry
- Author
-
Majumdar, Susruta, primary, Juntunen, Juha, additional, Sivendran, Sashi, additional, Bharti, Neelam, additional, and Sloan, K.B., additional
- Published
- 2006
- Full Text
- View/download PDF
118. Practical Synthesis of N‐Alkyl‐N‐alkyloxycarbonylaminomethyl Prodrug Derivatives of Acetaminophen, Theophylline, and 6‐Mercaptopurine
- Author
-
Majumdar, Susruta, primary and Sloan, Kenneth B., additional
- Published
- 2006
- Full Text
- View/download PDF
119. Synthesis, hydrolyses and dermal delivery of N-alkyl-N-alkyloxycarbonylaminomethyl (NANAOCAM) derivatives of phenol, imide and thiol containing drugs
- Author
-
Majumdar, Susruta, primary and Sloan, Kenneth B., additional
- Published
- 2006
- Full Text
- View/download PDF
120. Three-Component Coupling Approach for the Synthesisof Diverse Heterocycles Utilizing Reactive Nitrilium Trapping.
- Author
-
Váradi, András, Palmer, Travis C., Notis, Paula R., Redel-Traub, Gabriel N., Afonin, Daniel, Subrath, Joan J., Pasternak, Gavril W., Hu, Chunhua, Sharma, Indrajeet, and Majumdar, Susruta
- Published
- 2014
- Full Text
- View/download PDF
121. Synthesis and Evaluationof Aryl-Naloxamide OpiateAnalgesics Targeting Truncated Exon 11-Associated μ Opioid Receptor(MOR-1) Splice Variants.
- Author
-
Majumdar, Susruta, Subrath, Joan, Le Rouzic, Valerie, Polikar, Lisa, Burgman, Maxim, Nagakura, Kuni, Ocampo, Julie, Haselton, Nathan, Pasternak, Anna R., Grinnell, Steven, Pan, Ying-Xian, and Pasternak, Gavril W.
- Subjects
- *
NARCOTICS , *TARGETED drug delivery , *AMIDE synthesis , *OPIOID receptors , *DRUG synergism , *DRUG side effects - Abstract
3-Iodobenzoylnaltrexamide 1(IBNtxA) isa potent analgesicacting through a novel receptor target that lack many side-effectsof traditional opiates composed, in part, of exon 11-associated truncatedsix transmembrane domain MOR-1 (6TM/E11) splice variants. To betterunderstand the SAR of this drug target, a number of 4,5-epoxymorphinananalogues were synthesized. Results show the importance of a free3-phenolic group, a phenyl ring at the 6 position, an iodine at the3â²or 4â² position of the phenyl ring, and an N-allyl or c-propylmethyl group to maintainhigh 6TM/E11 affinity and activity. 3-Iodobenzoylnaloxamide 15(IBNalA) with a N-allyl group displayedlower δ opioid receptor affinity than its naltrexamine analogue,was 10-fold more potent an analgesic than morphine, elicited no respiratorydepression or physical dependence, and only limited inhibition ofgastrointestinal transit. Thus, the aryl-naloxamide scaffold can generatea potent analgesic acting through the 6TM/E11 sites with advantageousside-effect profile and greater selectivity. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
122. IUPHAR themed review: Opioid efficacy, bias, and selectivity.
- Author
-
Ramos-Gonzalez, Nokomis, Paul, Barnali, and Majumdar, Susruta
- Subjects
- *
OPIOID receptors , *OPIOIDS , *DRUG receptors , *OPIOID analgesics , *DRUG efficacy , *G proteins , *RESPIRATORY insufficiency , *CHRONIC pain - Abstract
Drugs acting at the opioid receptor family are clinically used to treat chronic and acute pain, though they represent the second line of treatment behind GABA analogs, antidepressants and SSRI's. Within the opioid family mu and kappa opioid receptor are commonly targeted. However, activation of the mu opioid receptor has side effects of constipation, tolerance, dependence, euphoria, and respiratory depression; activation of the kappa opioid receptor leads to dysphoria and sedation. The side effects of mu opioid receptor activation have led to mu receptor drugs being widely abused with great overdose risk. For these reasons, newer safer opioid analgesics are in high demand. For many years a focus within the opioid field was finding drugs that activated the G protein pathway at mu opioid receptor, without activating the β-arrestin pathway, known as biased agonism. Recent advances have shown that this may not be the way forward to develop safer analgesics at mu opioid receptor, though there is still some promise at the kappa opioid receptor. Here we discuss recent novel approaches to develop safer opioid drugs including efficacy vs bias and fine-tuning receptor activation by targeting sub-pockets in the orthosteric site, we explore recent works on the structural basis of bias, and we put forward the suggestion that Gα subtype selectivity may be an exciting new area of interest. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
123. Synthesis and Characterization of Azido Aryl Analogs of IBNtxA for Radio-Photoaffinity Labeling Opioid Receptors in Cell Lines and in Mouse Brain.
- Author
-
Grinnell, Steven G., Uprety, Rajendra, Varadi, Andras, Subrath, Joan, Hunkele, Amanda, Pan, Ying Xian, Pasternak, Gavril W., and Majumdar, Susruta
- Subjects
- *
OPIOID receptors , *CELL receptors , *CELL lines , *RNA splicing , *MICE , *ALKYNE derivatives , *PHOTOAFFINITY labeling - Abstract
Mu opioid receptors (MOR-1) mediate the biological actions of clinically used opioids such as morphine, oxycodone, and fentanyl. The mu opioid receptor gene, OPRM1, undergoes extensive alternative splicing, generating multiple splice variants. One type of splice variants are truncated variants containing only six transmembrane domains (6TM) that mediate the analgesic action of novel opioid drugs such as 3′-iodobenzoylnaltrexamide (IBNtxA). Previously, we have shown that IBNtxA is a potent analgesic effective in a spectrum of pain models but lacks many side-effects associated with traditional opiates. In order to investigate the targets labeled by IBNtxA, we synthesized two arylazido analogs of IBNtxA that allow photolabeling of mouse mu opioid receptors (mMOR-1) in transfected cell lines and mMOR-1 protein complexes that may comprise the 6TM sites in mouse brain. We demonstrate that both allyl and alkyne arylazido derivatives of IBNtxA efficiently radio-photolabeled mMOR-1 in cell lines and MOR-1 protein complexes expressed either exogenously or endogenously, as well as found in mouse brain. In future, design and application of such radio-photolabeling ligands with a conjugated handle will provide useful tools for further isolating or purifying MOR-1 to investigate site specific ligand–protein contacts and its signaling complexes. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
124. Kratom Alkaloids, Natural and Semi-Synthetic, Show Less Physical Dependence and Ameliorate Opioid Withdrawal.
- Author
-
Wilson, Lisa L., Chakraborty, Soumen, Eans, Shainnel O., Cirino, Thomas J., Stacy, Heather M., Simons, Chloe A., Uprety, Rajendra, Majumdar, Susruta, and McLaughlin, Jay P.
- Subjects
- *
KRATOM , *ALKALOIDS , *LABORATORY mice , *OPIOID analgesics , *ISOQUINOLINE alkaloids , *OPIOIDS , *NALOXONE - Abstract
Chronic administration of opioids produces physical dependence and opioid-induced hyperalgesia. Users claim the Thai traditional tea "kratom" and component alkaloid mitragynine ameliorate opioid withdrawal without increased sensitivity to pain. Testing these claims, we assessed the combined kratom alkaloid extract (KAE) and two individual alkaloids, mitragynine (MG) and the analog mitragynine pseudoindoxyl (MP), evaluating their ability to produce physical dependence and induce hyperalgesia after chronic administration, and as treatments for withdrawal in morphine-dependent subjects. C57BL/6J mice (n = 10/drug) were administered repeated saline, or graded, escalating doses of morphine (intraperitoneal; i.p.), kratom alkaloid extract (orally, p.o.), mitragynine (p.o.), or MP (subcutaneously, s.c.) for 5 days. Mice treated chronically with morphine, KAE, or mitragynine demonstrated significant drug-induced hyperalgesia by day 5 in a 48 °C warm-water tail-withdrawal test. Mice were then administered naloxone (10 mg/kg, s.c.) and tested for opioid withdrawal signs. Kratom alkaloid extract and the two individual alkaloids demonstrated significantly fewer naloxone-precipitated withdrawal signs than morphine-treated mice. Additional C57BL/6J mice made physically dependent on morphine were then used to test the therapeutic potential of combined KAE, mitragynine, or MP given twice daily over the next 3 days at either a fixed dose or in graded, tapering descending doses. When administered naloxone, mice treated with KAE, mitragynine, or MP under either regimen demonstrated significantly fewer signs of precipitated withdrawal than control mice that continued to receive morphine. In conclusion, while retaining some liabilities, kratom, mitragynine, and mitragynine pseudoindoxyl produced significantly less physical dependence and ameliorated precipitated withdrawal in morphine-dependent animals, suggesting some clinical value. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
125. Synthesis of spiro-2,6-dioxopiperazine and spiro-2,6-dioxopyrazine scaffolds using amino acids in a three-component reaction to generate potential Sigma-1 (σ1) receptor selective ligands.
- Author
-
Uprety, Rajendra, Váradi, András, Allaoa, Abdullah, Redel-Traub, Gabriel N., Palmer, Travis C., Feinberg, Evan N., Ferris, Alex C., Pande, Vijay S., Pasternak, Gavril W., and Majumdar, Susruta
- Subjects
- *
PYRAZINES , *AMINO acids , *CHEMICAL reactions , *LIGANDS (Biochemistry) , *ISOCYANIDES - Abstract
Abstract A library-friendly approach to generate new scaffolds is decisive for the development of molecular probes, drug like molecules and preclinical entities. Here, we present the design and synthesis of novel heterocycles with spiro-2,6-dioxopiperazine and spiro-2,6-pyrazine scaffolds through a three-component reaction using various amino acids, ketones, and isocyanides. Screening of select compounds over fifty CNS receptors including G-protein coupled receptors (GPCRs), ion channels, transporters, and enzymes through the NIMH psychoactive drug screening program indicated that a novel spiro-2,6-dioxopyrazine scaffold, UVM147 , displays high binding affinity at sigma-1 (σ 1) receptor in the nanomolar range. In addition, molecular docking of UVM147 at the human σ 1 receptor have shown that it resides in the same binding site that was occupied by the ligand 4-IBP used to obtain a crystal structure of the human sigma-1 (σ 1) receptor. Graphical abstract Image 1 Highlights • Novel spiro-2,6-dioxopiperazine and spiro-2,6-pyrazine heterocycles were synthesized. • The structure of methyl spiro-2,6-dioxopyrazine UVM147 was determined by X-ray crystallographic data analysis. • The methyl spiro-2,6-pyrazine UVM147 (K i , 8.6 nM) is a potent ligand at sigma-1 (σ 1) receptor. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
126. Mediation of opioid analgesia by a truncated 6-transmembrane GPCR.
- Author
-
Zhigang Lu, Jin Xu, Rossi, Grace C., Majumdar, Susruta, Pasternak, Gavril W., and Ying-Xian Pan
- Subjects
- *
OPIOID receptors , *G protein coupled receptors , *MORPHINE , *METHADONE hydrochloride , *ANALGESICS , *LENTIVIRUSES , *LABORATORY mice - Abstract
The generation of potent opioid analgesics that lack the side effects of traditional opioids may be possible by targeting truncated splice variants of the μ-opioid receptor. μ-Opioids act through GPCRs that are generated from the Oprml gene, which undergoes extensive alternative splicing. The most abundant set of Oprml variants encode classical full-length 7 transmembrane domain (7TM) μ-opioid receptors that mediate the actions of the traditional μ-opioid drugs morphine and methadone. In contrast, 3-iodobenzoyl-6β-naltrexamide (IBNtxA) is a potent analgesic against thermal, inflammatory, and neuropathic pain that acts independently of 7TM μ-opioid receptors but has no activity in mice lacking a set of 6TM truncated μ-opioid receptor splice variants. Unlike traditional opioids, IBNtxA does not depress respiration or result in physical dependence or reward behavior, suggesting it acts through an alternative μ-opioid receptor target. Here we demonstrated that a truncated STM splice variant, mMOR-IG, can rescue IBNtxA analgesia in a μ-opioid receptor-deficient mouse that lacks all Oprml splice variants, ablating μ-opioid activity in these animals. Intrathecal administration of lentivirus containing the STM variant mMOR-ID restored IBNtxA, but not morphine, analgesia in Oprm7-deficient animals. Together, these results confirm that a truncated 6TM GPCR is both necessary and sufficient for IBNtxA analgesia. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
127. Oxa-Iboga alkaloids lack cardiac risk and disrupt opioid use in animal models.
- Author
-
Havel V, Kruegel AC, Bechand B, McIntosh S, Stallings L, Hodges A, Wulf MG, Nelson M, Hunkele A, Ansonoff M, Pintar JE, Hwu C, Ople RS, Abi-Gerges N, Zaidi SA, Katritch V, Yang M, Javitch JA, Majumdar S, Hemby SE, and Sames D
- Subjects
- Animals, Humans, Male, Rats, Opioid-Related Disorders drug therapy, Analgesics, Opioid adverse effects, Analgesics, Opioid pharmacology, Rats, Sprague-Dawley, Disease Models, Animal, Receptors, Opioid, kappa metabolism, Receptors, Opioid, kappa agonists, Receptors, Opioid, kappa genetics, Alkaloids pharmacology, Hyperalgesia chemically induced, Hyperalgesia drug therapy, Ibogaine analogs & derivatives, Ibogaine pharmacology, Ibogaine therapeutic use, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism
- Abstract
Ibogaine and its main metabolite noribogaine provide important molecular prototypes for markedly different treatment of substance use disorders and co-morbid mental health illnesses. However, these compounds present a cardiac safety risk and a highly complex molecular mechanism. We introduce a class of iboga alkaloids - termed oxa-iboga - defined as benzofuran-containing iboga analogs and created via structural editing of the iboga skeleton. The oxa-iboga compounds lack the proarrhythmic adverse effects of ibogaine and noribogaine in primary human cardiomyocytes and show superior efficacy in animal models of opioid use disorder in male rats. They act as potent kappa opioid receptor agonists in vitro and in vivo, but exhibit atypical behavioral features compared to standard kappa opioid agonists. Oxa-noribogaine induces long-lasting suppression of morphine, heroin, and fentanyl intake after a single dose or a short treatment regimen, reversal of persistent opioid-induced hyperalgesia, and suppression of opioid drug seeking in rodent relapse models. As such, oxa-iboga compounds represent mechanistically distinct iboga analogs with therapeutic potential., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
128. Signaling Modulation Mediated by Ligand Water Interactions with the Sodium Site at μOR.
- Author
-
Ople RS, Ramos-Gonzalez N, Li Q, Sobecks BL, Aydin D, Powers AS, Faouzi A, Polacco BJ, Bernhard SM, Appourchaux K, Sribhashyam S, Eans SO, Tsai BA, Dror RO, Varga BR, Wang H, Hüttenhain R, McLaughlin JP, and Majumdar S
- Abstract
The mu opioid receptor (μOR) is a target for clinically used analgesics. However, adverse effects, such as respiratory depression and physical dependence, necessitate the development of alternative treatments. Recently we reported a novel strategy to design functionally selective opioids by targeting the sodium binding allosteric site in μOR with a supraspinally active analgesic named C6guano . Presently, to improve systemic activity of this ligand, we used structure-based design, identifying a new ligand named RO76 where the flexible alkyl linker and polar guanidine guano group is swapped with a benzyl alcohol, and the sodium site is targeted indirectly through waters. A cryoEM structure of RO76 bound to the μOR-G
i complex confirmed that RO76 interacts with the sodium site residues through a water molecule, unlike C6guano which engages the sodium site directly. Signaling assays coupled with APEX based proximity labeling show binding in the sodium pocket modulates receptor efficacy and trafficking. In mice, RO76 was systemically active in tail withdrawal assays and showed reduced liabilities compared to those of morphine. In summary, we show that targeting water molecules in the sodium binding pocket may be an avenue to modulate signaling properties of opioids, and which may potentially be extended to other G-protein coupled receptors where this site is conserved., Competing Interests: The authors declare the following competing financial interest(s): S.M. is a cofounder of Sparian Biosciences. R.O.D. holds equity in Septerna, Inc. Other authors declare no competing financial interest., (© 2024 The Authors. Published by American Chemical Society.)- Published
- 2024
- Full Text
- View/download PDF
129. Structure-Guided Design of Partial Agonists at an Opioid Receptor.
- Author
-
Che T, Varga B, Bernhard SM, El Daibani A, Zaidi S, Lam J, Aguilar J, Appourchaux K, Nazarova A, Kouvelis A, Eans S, Margolis E, Fay J, Pradhan A, Katritch V, McLaughlin J, and Majumdar S
- Abstract
The persistence of chronic pain and continuing overdose deaths from pain-relieving opioids targeting μ opioid receptor (μOR) have fueled the need for reliable long-term analgesics which use different targets and mechanisms. The δ opioid receptor (δOR) is a potential alternative target for non-addictive analgesics to alleviate chronic pain, made more attractive by its lack of respiratory depression associated with μOR agonists. However, early δOR full agonists were found to induce seizures, precluding clinical use. Partial δOR agonists may offer more controlled activation of the receptor compared to full agonists, but the development of such ligands has been hindered by uncertainty over the molecular mechanism mediating partial agonism. Using a structure-based approach, we explored the engagement of the sodium binding pocket in δOR and developed a bitopic ligand, C6-Quino, predicted to be a selective δOR partial agonist. Functional studies of C6-Quino revealed that it displayed δOR partial agonist activity at both G-protein and arrestin pathways. Its interaction with the sodium pocket was confirmed and analyzed using a single particle cryo-EM. Additionally, C6-Quino demonstrated favorable chemical and physiological properties like oral activity, and analgesic activity in multiple chronic pain models. Notably, μOR-related hyperlocomotion and respiratory depression, and δOR-related convulsions, were not observed at analgesic doses of C6-Quino. This fundamentally new approach to designing δOR ligands provides a blueprint for the development of partial agonists as safe analgesics and acts as a generic method to optimize signaling profiles of other Class A GPCRs., Competing Interests: Competing financial interests The authors declare no competing financial interests.
- Published
- 2024
- Full Text
- View/download PDF
130. A µ-opioid receptor modulator that works cooperatively with naloxone.
- Author
-
O'Brien ES, Rangari VA, El Daibani A, Eans SO, Hammond HR, White E, Wang H, Shiimura Y, Krishna Kumar K, Jiang Q, Appourchaux K, Huang W, Zhang C, Kennedy BJ, Mathiesen JM, Che T, McLaughlin JP, Majumdar S, and Kobilka BK
- Subjects
- Animals, Humans, Male, Mice, Allosteric Regulation drug effects, Binding Sites drug effects, Cryoelectron Microscopy, Fentanyl antagonists & inhibitors, Fentanyl pharmacology, Kinetics, Ligands, Models, Molecular, Morphine antagonists & inhibitors, Morphine pharmacology, Narcotic Antagonists administration & dosage, Narcotic Antagonists chemistry, Narcotic Antagonists metabolism, Narcotic Antagonists pharmacology, Opiate Overdose drug therapy, Protein Conformation drug effects, Protein Stability drug effects, Sf9 Cells, Signal Transduction drug effects, Mice, Inbred C57BL, Analgesics, Opioid antagonists & inhibitors, Analgesics, Opioid pharmacology, Drug Evaluation, Preclinical, Naloxone administration & dosage, Naloxone chemistry, Naloxone metabolism, Naloxone pharmacology, Receptors, Opioid, mu agonists, Receptors, Opioid, mu antagonists & inhibitors, Receptors, Opioid, mu chemistry, Receptors, Opioid, mu metabolism, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology
- Abstract
The µ-opioid receptor (µOR) is a well-established target for analgesia
1 , yet conventional opioid receptor agonists cause serious adverse effects, notably addiction and respiratory depression. These factors have contributed to the current opioid overdose epidemic driven by fentanyl2 , a highly potent synthetic opioid. µOR negative allosteric modulators (NAMs) may serve as useful tools in preventing opioid overdose deaths, but promising chemical scaffolds remain elusive. Here we screened a large DNA-encoded chemical library against inactive µOR, counter-screening with active, G-protein and agonist-bound receptor to 'steer' hits towards conformationally selective modulators. We discovered a NAM compound with high and selective enrichment to inactive µOR that enhances the affinity of the key opioid overdose reversal molecule, naloxone. The NAM works cooperatively with naloxone to potently block opioid agonist signalling. Using cryogenic electron microscopy, we demonstrate that the NAM accomplishes this effect by binding a site on the extracellular vestibule in direct contact with naloxone while stabilizing a distinct inactive conformation of the extracellular portions of the second and seventh transmembrane helices. The NAM alters orthosteric ligand kinetics in therapeutically desirable ways and works cooperatively with low doses of naloxone to effectively inhibit various morphine-induced and fentanyl-induced behavioural effects in vivo while minimizing withdrawal behaviours. Our results provide detailed structural insights into the mechanism of negative allosteric modulation of the µOR and demonstrate how this can be exploited in vivo., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
131. Design and structural validation of peptide-drug conjugate ligands of the kappa-opioid receptor.
- Author
-
Muratspahić E, Deibler K, Han J, Tomašević N, Jadhav KB, Olivé-Marti AL, Hochrainer N, Hellinger R, Koehbach J, Fay JF, Rahman MH, Hegazy L, Craven TW, Varga BR, Bhardwaj G, Appourchaux K, Majumdar S, Muttenthaler M, Hosseinzadeh P, Craik DJ, Spetea M, Che T, Baker D, and Gruber CW
- Subjects
- Male, Mice, Animals, Ligands, Receptors, Opioid, mu metabolism, Peptides, Cyclic chemistry, Receptors, Opioid, kappa metabolism, Analgesics, Opioid chemistry
- Abstract
Despite the increasing number of GPCR structures and recent advances in peptide design, the development of efficient technologies allowing rational design of high-affinity peptide ligands for single GPCRs remains an unmet challenge. Here, we develop a computational approach for designing conjugates of lariat-shaped macrocyclized peptides and a small molecule opioid ligand. We demonstrate its feasibility by discovering chemical scaffolds for the kappa-opioid receptor (KOR) with desired pharmacological activities. The designed De Novo Cyclic Peptide (DNCP)-β-naloxamine (NalA) exhibit in vitro potent mixed KOR agonism/mu-opioid receptor (MOR) antagonism, nanomolar binding affinity, selectivity, and efficacy bias at KOR. Proof-of-concept in vivo efficacy studies demonstrate that DNCP-β-NalA(1) induces a potent KOR-mediated antinociception in male mice. The high-resolution cryo-EM structure (2.6 Å) of the DNCP-β-NalA-KOR-Gi1 complex and molecular dynamics simulations are harnessed to validate the computational design model. This reveals a network of residues in ECL2/3 and TM6/7 controlling the intrinsic efficacy of KOR. In general, our computational de novo platform overcomes extensive lead optimization encountered in ultra-large library docking and virtual small molecule screening campaigns and offers innovation for GPCR ligand discovery. This may drive the development of next-generation therapeutics for medical applications such as pain conditions., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
132. Ligand and G-protein selectivity in the κ-opioid receptor.
- Author
-
Han J, Zhang J, Nazarova AL, Bernhard SM, Krumm BE, Zhao L, Lam JH, Rangari VA, Majumdar S, Nichols DE, Katritch V, Yuan P, Fay JF, and Che T
- Subjects
- Analgesics, Opioid metabolism, Analgesics, Opioid pharmacology, Signal Transduction, Substrate Specificity, Allosteric Regulation drug effects, Hallucinogens metabolism, Hallucinogens pharmacology, Cryoelectron Microscopy, Ligands, Receptors, Opioid, kappa chemistry, Receptors, Opioid, kappa metabolism, Receptors, Opioid, kappa ultrastructure, Heterotrimeric GTP-Binding Proteins chemistry, Heterotrimeric GTP-Binding Proteins metabolism, Heterotrimeric GTP-Binding Proteins ultrastructure
- Abstract
The κ-opioid receptor (KOR) represents a highly desirable therapeutic target for treating not only pain but also addiction and affective disorders
1 . However, the development of KOR analgesics has been hindered by the associated hallucinogenic side effects2 . The initiation of KOR signalling requires the Gi/o -family proteins including the conventional (Gi1 , Gi2 , Gi3 , GoA and GoB ) and nonconventional (Gz and Gg ) subtypes. How hallucinogens exert their actions through KOR and how KOR determines G-protein subtype selectivity are not well understood. Here we determined the active-state structures of KOR in a complex with multiple G-protein heterotrimers-Gi1 , GoA , Gz and Gg -using cryo-electron microscopy. The KOR-G-protein complexes are bound to hallucinogenic salvinorins or highly selective KOR agonists. Comparisons of these structures reveal molecular determinants critical for KOR-G-protein interactions as well as key elements governing Gi/o -family subtype selectivity and KOR ligand selectivity. Furthermore, the four G-protein subtypes display an intrinsically different binding affinity and allosteric activity on agonist binding at KOR. These results provide insights into the actions of opioids and G-protein-coupling specificity at KOR and establish a foundation to examine the therapeutic potential of pathway-selective agonists of KOR., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
133. Insights into distinct signaling profiles of the µOR activated by diverse agonists.
- Author
-
Qu Q, Huang W, Aydin D, Paggi JM, Seven AB, Wang H, Chakraborty S, Che T, DiBerto JF, Robertson MJ, Inoue A, Suomivuori CM, Roth BL, Majumdar S, Dror RO, Kobilka BK, and Skiniotis G
- Subjects
- Animals, beta-Arrestins metabolism, GTP-Binding Proteins metabolism, Binding Sites, Signal Transduction, Analgesics, Opioid chemistry, Analgesics, Opioid pharmacology
- Abstract
Drugs targeting the μ-opioid receptor (μOR) are the most effective analgesics available but are also associated with fatal respiratory depression through a pathway that remains unclear. Here we investigated the mechanistic basis of action of lofentanil (LFT) and mitragynine pseudoindoxyl (MP), two μOR agonists with different safety profiles. LFT, one of the most lethal opioids, and MP, a kratom plant derivative with reduced respiratory depression in animal studies, exhibited markedly different efficacy profiles for G protein subtype activation and β-arrestin recruitment. Cryo-EM structures of μOR-Gi1 complex with MP (2.5 Å) and LFT (3.2 Å) revealed that the two ligands engage distinct subpockets, and molecular dynamics simulations showed additional differences in the binding site that promote distinct active-state conformations on the intracellular side of the receptor where G proteins and β-arrestins bind. These observations highlight how drugs engaging different parts of the μOR orthosteric pocket can lead to distinct signaling outcomes., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
134. Opioid signaling and design of analgesics.
- Author
-
Paul B, Sribhashyam S, and Majumdar S
- Subjects
- Humans, Analgesics pharmacology, Signal Transduction, Receptors, Opioid, Analgesics, Opioid adverse effects, Receptors, Opioid, mu agonists, Receptors, Opioid, mu metabolism
- Abstract
Clinical treatment of acute to severe pain relies on the use of opioids. While their potency is significant, there are considerable side effects that can negatively affect patients. Their rise in usage has correlated with the current opioid epidemic in the United States, which has led to more than 70,000 deaths per year (Volkow and Blanco, 2021). Opioid-related drug development aims to make target compounds that show strong potency but with diminished side effects. Research into pharmaceuticals that could act as potential alternatives to current pains medications has relied on mechanistic insights of opioid receptors, a class of G-protein coupled receptors (GPCRs), and biased agonism, a common phenomenon among pharmaceutical compounds where downstream effects can be altered at the same receptor via different agonists. Opioids function typically by binding to an active site on the extracellular portion of opioid receptors. Once activated, the opioid receptor initiates a G-protein signaling pathway and/or the β-arrestin2 pathway. The proposed concept for the development of safe analgesics around mu and kappa opioid receptor subtypes has focused on not recruiting β-arrestin2 (biased agonism) and/or having low efficacy at the receptor (partial agonism). By altering chemical motifs on a common scaffold, chemists can take advantage of biased agonism as well as create compounds with low intrinsic efficacy for the desired treatments. This review will focus on ligands with bias profile, signaling aspects of the receptor and probe into the structural basis of receptor that leads to bias and/or partial agonism., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
135. Structure-based design of bitopic ligands for the µ-opioid receptor.
- Author
-
Faouzi A, Wang H, Zaidi SA, DiBerto JF, Che T, Qu Q, Robertson MJ, Madasu MK, El Daibani A, Varga BR, Zhang T, Ruiz C, Liu S, Xu J, Appourchaux K, Slocum ST, Eans SO, Cameron MD, Al-Hasani R, Pan YX, Roth BL, McLaughlin JP, Skiniotis G, Katritch V, Kobilka BK, and Majumdar S
- Subjects
- Animals, Mice, Analgesics, Opioid chemistry, Analgesics, Opioid metabolism, Arrestins metabolism, Cryoelectron Microscopy, Ligands, Binding Sites, Nociception, Fentanyl analogs & derivatives, Fentanyl chemistry, Fentanyl metabolism, Morphinans chemistry, Morphinans metabolism, Receptors, Opioid, mu agonists, Receptors, Opioid, mu chemistry, Receptors, Opioid, mu metabolism, Receptors, Opioid, mu ultrastructure, Drug Design
- Abstract
Mu-opioid receptor (µOR) agonists such as fentanyl have long been used for pain management, but are considered a major public health concern owing to their adverse side effects, including lethal overdose
1 . Here, in an effort to design safer therapeutic agents, we report an approach targeting a conserved sodium ion-binding site2 found in µOR3 and many other class A G-protein-coupled receptors with bitopic fentanyl derivatives that are functionalized via a linker with a positively charged guanidino group. Cryo-electron microscopy structures of the most potent bitopic ligands in complex with µOR highlight the key interactions between the guanidine of the ligands and the key Asp2.50 residue in the Na+ site. Two bitopics (C5 and C6 guano) maintain nanomolar potency and high efficacy at Gi subtypes and show strongly reduced arrestin recruitment-one (C6 guano) also shows the lowest Gz efficacy among the panel of µOR agonists, including partial and biased morphinan and fentanyl analogues. In mice, C6 guano displayed µOR-dependent antinociception with attenuated adverse effects, supporting the µOR sodium ion-binding site as a potential target for the design of safer analgesics. In general, our study suggests that bitopic ligands that engage the sodium ion-binding pocket in class A G-protein-coupled receptors can be designed to control their efficacy and functional selectivity profiles for Gi , Go and Gz subtypes and arrestins, thus modulating their in vivo pharmacology., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
136. Site selective C-H functionalization of Mitragyna alkaloids reveals a molecular switch for tuning opioid receptor signaling efficacy.
- Author
-
Bhowmik S, Galeta J, Havel V, Nelson M, Faouzi A, Bechand B, Ansonoff M, Fiala T, Hunkele A, Kruegel AC, Pintar JE, Majumdar S, Javitch JA, and Sames D
- Subjects
- Analgesics chemistry, Analgesics pharmacology, Animals, Ethylene Glycol chemistry, Humans, Mice, Knockout, Models, Chemical, Molecular Structure, Plant Extracts chemistry, Protein Binding, Receptors, Opioid, mu genetics, Receptors, Opioid, mu metabolism, Secologanin Tryptamine Alkaloids chemistry, Mice, Mitragyna chemistry, Plant Extracts pharmacology, Receptors, Opioid, mu agonists, Secologanin Tryptamine Alkaloids pharmacology
- Abstract
Mitragynine (MG) is the most abundant alkaloid component of the psychoactive plant material "kratom", which according to numerous anecdotal reports shows efficacy in self-medication for pain syndromes, depression, anxiety, and substance use disorders. We have developed a synthetic method for selective functionalization of the unexplored C11 position of the MG scaffold (C6 position in indole numbering) via the use of an indole-ethylene glycol adduct and subsequent iridium-catalyzed borylation. Through this work we discover that C11 represents a key locant for fine-tuning opioid receptor signaling efficacy. 7-Hydroxymitragynine (7OH), the parent compound with low efficacy on par with buprenorphine, is transformed to an even lower efficacy agonist by introducing a fluorine substituent in this position (11-F-7OH), as demonstrated in vitro at both mouse and human mu opioid receptors (mMOR/hMOR) and in vivo in mouse analgesia tests. Low efficacy opioid agonists are of high interest as candidates for generating safer opioid medications with mitigated adverse effects.
- Published
- 2021
- Full Text
- View/download PDF
137. Canvass: A Crowd-Sourced, Natural-Product Screening Library for Exploring Biological Space.
- Author
-
Kearney SE, Zahoránszky-Kőhalmi G, Brimacombe KR, Henderson MJ, Lynch C, Zhao T, Wan KK, Itkin Z, Dillon C, Shen M, Cheff DM, Lee TD, Bougie D, Cheng K, Coussens NP, Dorjsuren D, Eastman RT, Huang R, Iannotti MJ, Karavadhi S, Klumpp-Thomas C, Roth JS, Sakamuru S, Sun W, Titus SA, Yasgar A, Zhang YQ, Zhao J, Andrade RB, Brown MK, Burns NZ, Cha JK, Mevers EE, Clardy J, Clement JA, Crooks PA, Cuny GD, Ganor J, Moreno J, Morrill LA, Picazo E, Susick RB, Garg NK, Goess BC, Grossman RB, Hughes CC, Johnston JN, Joullie MM, Kinghorn AD, Kingston DGI, Krische MJ, Kwon O, Maimone TJ, Majumdar S, Maloney KN, Mohamed E, Murphy BT, Nagorny P, Olson DE, Overman LE, Brown LE, Snyder JK, Porco JA Jr, Rivas F, Ross SA, Sarpong R, Sharma I, Shaw JT, Xu Z, Shen B, Shi W, Stephenson CRJ, Verano AL, Tan DS, Tang Y, Taylor RE, Thomson RJ, Vosburg DA, Wu J, Wuest WM, Zakarian A, Zhang Y, Ren T, Zuo Z, Inglese J, Michael S, Simeonov A, Zheng W, Shinn P, Jadhav A, Boxer MB, Hall MD, Xia M, Guha R, and Rohde JM
- Abstract
Natural products and their derivatives continue to be wellsprings of nascent therapeutic potential. However, many laboratories have limited resources for biological evaluation, leaving their previously isolated or synthesized compounds largely or completely untested. To address this issue, the Canvass library of natural products was assembled, in collaboration with academic and industry researchers, for quantitative high-throughput screening (qHTS) across a diverse set of cell-based and biochemical assays. Characterization of the library in terms of physicochemical properties, structural diversity, and similarity to compounds in publicly available libraries indicates that the Canvass library contains many structural elements in common with approved drugs. The assay data generated were analyzed using a variety of quality control metrics, and the resultant assay profiles were explored using statistical methods, such as clustering and compound promiscuity analyses. Individual compounds were then sorted by structural class and activity profiles. Differential behavior based on these classifications, as well as noteworthy activities, are outlined herein. One such highlight is the activity of (-)-2( S )-cathafoline, which was found to stabilize calcium levels in the endoplasmic reticulum. The workflow described here illustrates a pilot effort to broadly survey the biological potential of natural products by utilizing the power of automation and high-throughput screening., Competing Interests: The authors declare no competing financial interest.
- Published
- 2018
- Full Text
- View/download PDF
138. Fentanyl-related designer drugs W-18 and W-15 lack appreciable opioid activity in vitro and in vivo.
- Author
-
Huang XP, Che T, Mangano TJ, Le Rouzic V, Pan YX, Majumdar S, Cameron MD, Baumann MH, Pasternak GW, and Roth BL
- Subjects
- Analgesics, Opioid, Animals, Drug Evaluation, Preclinical, HEK293 Cells, Humans, Illicit Drugs, Mice, Receptor, Cannabinoid, CB1 drug effects, Receptor, Cannabinoid, CB2 drug effects, Receptors, Opioid drug effects, Receptors, Serotonin drug effects, Designer Drugs chemistry, Designer Drugs pharmacology, Fentanyl chemistry, Fentanyl pharmacology
- Abstract
W-18 (4-chloro-N-[1-[2-(4-nitrophenyl)ethyl]-2-piperidinylidene]-benzenesulfonamide) and W-15 (4-chloro-N-[1-(2-phenylethyl)-2-piperidinylidene]-benzenesulfonamide) represent two emerging drugs of abuse chemically related to the potent opioid agonist fentanyl (N-(1-(2-phenylethyl)-4-piperidinyl)-N-phenylpropanamide). Here, we describe the comprehensive pharmacological profiles of W-18 and W-15, as examination of their structural features predicted that they might lack opioid activity. We found W-18 and W-15 to be without detectible activity at μ, δ, κ, and nociception opioid receptors in a variety of assays. We also tested W-18 and W-15 for activity as allosteric modulators at opioid receptors and found them devoid of significant positive or negative allosteric modulatory activity. Comprehensive profiling at essentially all the druggable GPCRs in the human genome using the PRESTO-Tango platform revealed no significant activity. Weak activity at the sigma receptors and the peripheral benzodiazepine receptor was found for W-18 (Ki = 271 nM). W-18 showed no activity in either the radiant heat tail-flick or the writhing assays and also did not induce classical opioid behaviors. W-18 is extensively metabolized, but its metabolites also lack opioid activity. Thus, although W-18 and W-15 have been suggested to be potent opioid agonists, our results reveal no significant activity at these or other known targets for psychoactive drugs.
- Published
- 2017
- Full Text
- View/download PDF
139. Radioligand Binding Assay for an Exon 11-Associated Mu Opioid Receptor Target.
- Author
-
Marrone GF, Majumdar S, and Pasternak GW
- Subjects
- Animals, Binding, Competitive, Brain metabolism, Ligands, Mice, Naltrexone analogs & derivatives, Naltrexone metabolism, Protein Binding, Exons genetics, Iodine Radioisotopes metabolism, Receptors, Opioid, mu genetics, Receptors, Opioid, mu metabolism
- Abstract
Receptor binding provides a valuable approach for characterization of drugs and their receptors. There are three major families of opioid receptors: mu, delta, and kappa. Highly selective radioligands are available for all three classes of traditional receptors. Of the three, the mu receptor undergoes extensive alternative splicing, generating a number of traditional mu receptor subtypes as well as a nontraditional, truncated set of variants associated with exon 11. These exon 11-associated truncated variants are not readily labeled with current radioligands. Here we describe the synthesis of a radioiodinated ligand suitable for carrying out binding studies for this target.
- Published
- 2015
- Full Text
- View/download PDF
140. Fit of fluxes of sunscreens and other compounds from propylene glycol:water (30:70) through human skin and silicone membrane to the Roberts-Sloan equation: the effect of polar vehicle (or water) solubility.
- Author
-
Sloan KB, Devarajan-Ketha H, Synovec J, and Majumdar S
- Subjects
- Humans, Propylene Glycol chemistry, Propylene Glycol pharmacokinetics, Silicones, Skin metabolism, Solubility, Sunscreening Agents pharmacokinetics, Water chemistry
- Abstract
It would be useful to develop a surrogate for animal skin, which could be use to predict flux through human skin. The fluxes (and physicochemical properties) of sunscreens and other compounds from propylene glycol (PG):water (AQ), 30:70, through human skin have previously been reported. We measured the fluxes of several of those sunscreens and other compounds from PG:AQ, 30:70, through silicone membrane and fit both sets of data to the Roberts-Sloan (RS) equation to determine any similarities. For both sets of data, the fluxes were directly dependent on their solubilities in a lipid solvent [octanol (OCT), in this case] and in a polar solvent (PG:AQ, 30:70, or AQ in this case) and inversely on their molecular weights. The fit of the experimental (EXP) fluxes through human skin in vivo to RS was excellent: r² = 0.92 if the vehicle (VEH) PG:AQ, 30:70 was the polar solvent (RS¹) or r² = 0.97 if water was the polar solvent (RS²). The fit of the EXP fluxes through silicone membrane to RS was good: r² = 0.80 if the VEH PG:AQ, 30:70, was the polar solvent (RS¹) or r² = 0.81 if water was the polar solvent (RS²). The correlations between their EXP fluxes through human skin in vivo and their EXP fluxes through silicone membrane were good (r² = 0.85). In addition, the correlation between EXP fluxes from PG:AQ, 30:70, through human skin in vivo and their fluxes calculated from the coefficients of the fit of solubilities, molecular weights and fluxes from water through silicone membranes from a previous n = 22 database to RS was even better (r² = 0.94). These results suggest that flux through human skin can be calculated from flux through a silicone membrane.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.