Abstract
G-protein-coupled receptors are integral membrane proteins that transduce chemical signals from the extracellular matrix into the cell. Traditional drug design has considered ligand-receptor interactions only under normal conditions. However, studies on opioids indicate that such interactions are very different in diseased tissues. In such microenvironments, protons play an important role in structural and functional alterations of both ligands and receptors. The pertinent literature strongly suggests that future drug design should take these aspects into account in order to reduce adverse side effects while preserving desired effects of novel compounds.
Significance
Structure-based drug design has yielded novel opioid agonists that selectively activate receptors in injured, but not normal, tissues. These findings have important implications and may enable the development of improved GPCR drugs devoid of adverse side effects elicited in noninjured organs.
Introduction
G-protein-coupled receptors (GPCRs) are integral membrane proteins that transduce chemical signals from the extracellular matrix into the cell. They are the targets of over one third of all drugs available today (1,2). A prominent GPCR subgroup is opioid receptors. Opioid receptors are expressed throughout the nervous system and in many other organs (e.g., endocrine, immune, and intestinal systems). These receptors can be targeted by a large variety of endogenous (peptide) and exogenous (plant-derived or synthetic) opioid ligands (2,3,4,5). For centuries, opioid agonists have been the most efficient drugs for the treatment of severe pain. Conventional opioids (e.g., morphine, fentanyl) produce pain relief (analgesia) by the activation of mu-opioid receptors (MORs; the clinically most important opioid receptor type) and consequent modulation of downstream signaling molecules (G proteins, membrane ion channels) in central and peripheral sensory neurons (3,4,5,6,7). Unfortunately, this is accompanied by serious adverse effects such as addiction, dependence, cardio-respiratory arrest, locomotor disturbance, sedation, nausea, and/or constipation. These side effects are mediated by opioid receptors in the brain or intestine. In addition, tolerance (i.e., diminishing effects with repeated administration of opioid ligands) to all desired and undesired actions (e.g., analgesia, respiratory depression, nausea, sedation) can develop (4,5,6,8).
In view of these detrimental effects and the opioid crisis resulting therefrom, extensive research efforts are ongoing to find ways of eliminating adverse actions while preserving analgesia. A particularly promising—and clinically proven—approach is the selective (exclusive) activation of opioid receptors on peripheral sensory neurons innervating injured tissue (i.e., the source of pain generation) while leaving out opioid receptors in healthy environments (e.g., brain, intestinal wall) (4,6,9,10,11,12,13,14,15).
Injured tissue (resulting from, e.g., surgery, trauma, arthritis, colitis, cancer, infection, or lack of oxygen delivery) is accompanied by inflammation. This diseased milieu is made up of increased amounts of various cells (e.g., neutrophils, lymphocytes, macrophages) and mediators (e.g., cytokines, chemokines, extravasated proteins, prostaglandins, cyclooxygenase, oxygen radicals, lactate, protons) and is characterized by acidosis (pH < 7.35) (16,17,18) (Table 1). Importantly, protons are not only capable of directly activating so-called “proton-sensing” GPCRs (19,20) but can also influence interactions between different GPCRs and their cognate ligands (21,22). The present article will focus on the latter. In summary, current evidence suggests that future drug design should depart from analyzing the structure and function of GPCRs only under normal conditions. More attention should be paid to differences between healthy and diseased microenvironments (e.g., hypoxia, ionic strength).
Table 1.
pH values in injured/inflamed tissues measured in vivo/ex vivo
| Species, tissue | Lowest pH | Reference | 
|---|---|---|
| Infectious agents | ||
| Human, abscess | 5.4 | (23) | 
| Guinea pig, intraperitoneal bacterial inoculation | 5.6 | (24) | 
| Mouse, subcutaneous/intraperitoneal bacterial inoculation | 5.8 | (24) | 
| Rat, air pouch granuloma induced by carrageenan, dextran, staph. aureus | 6.87 | (25) | 
| Human, abdominal abscess | 6.0 | (26) | 
| Human, peritoneal fluid, abdominal infection | 5.9 | (27) | 
| Rat, Freund’s adjuvant paw inflammation | 6.8 | (28) | 
| Rat, paw incision | 7.02 | (28) | 
| Rat, Freund’s adjuvant paw inflammation | 6.82 | (29) | 
| Rat, Freund’s adjuvant paw inflammation | 6.84 | (30) | 
| Noninfectious agents | ||
| Dog, turpentine-induced pleural exudate | 6.6 | (31) | 
| Dog, turpentine-induced pleural exudate | 6.5 | (32) | 
| Dog, turpentine-induced pleural exudate | 6.0 | (33) | 
| Rabbit, diabetic skin wounds | 6.9 | (34) | 
| Rabbit, brain, wounds, ischemia | 5.0 | (35) | 
| Rat, ischemic heart, intracellular | 5.7 | (36) | 
| Rat, seminiferous tubules and epididymis | 6.57 ± 0.08 | (37) | 
| Human, exercised muscle, intracellular pH | 6.1 | (38) | 
| Human, exercised muscle, calculated intracellular pH | 6.31 ± 0.09 | (39) | 
| Rat, carrageenan inflammation, aspirated | 6.94 | (40) | 
| Human, exercised muscle, intracutaneous pH | 6.67 | (41) | 
| Human, atherosclerotic plaque | 7.15 ± 0.01 | (42) | 
| Rabbit, aorta | 7.40 ± 0.43 | (42) | 
| Human, umbilical artery | 7.24 ± 0.1 | (42) | 
| Rat, plantar/gastrocnemius incision | 6.54 ± 0.12 | (43) | 
| Human, muscle | 4.5 | (44) | 
| Minipig, intervertebral disc degeneration | 5.7 | (45) | 
| Rat, chronic sciatic nerve constriction | 6.91 | (46) | 
| Rat, intraperitoneal acetic acid injection | 4.52 (5 min), 6.97 (15 min) | (46) | 
| Mouse, colitis | 6.71 ± 0.09 | (47) | 
| Mouse, colitis | 6.36 ± 0.05 | (48) | 
| Cancer | ||
| Rat, implanted tumors | 6.82 | (49) | 
| Human, malignant tumors, inflamed tissues | 5.44 | (50) | 
| Human, malignant tumor | 5.7 | (51) | 
| Human, astrocytoma | 5.85 | (52) | 
| Human, melanoma | 6.4 | (53) | 
| Mouse, mammary carcinoma | 5.8 | (54) | 
| Nude mouse, implanted tumors | 6.65 | (55) | 
| Mouse, implanted tumor | 6.66 | (56) | 
| Rat, mouse, implanted tumors | 6.3 | (57) | 
| Mouse, implanted tumors | 6.0 | (58) | 
| Mouse, implanted subcutaneous lymphoma | 6.0 | (59) | 
| Arthritis | ||
| Human, osteoarthritis, joint injury, synovial fluid | 6.5 | (60) | 
| Human, rheumatoid arthritis synovial fluid | 7.08 | (61) | 
| Human, rheumatoid arthritis synovial fluid | 6.0 | (62) | 
| Human, rheumatoid arthritis synovial fluid | 6.4 | (63) | 
| Human, rheumatoid arthritis | 6.84 | (64) | 
| Human, arthritis | 6.60 | (65) | 
| Human, arthritis, synovial fluid | 6.2 | (66) | 
| Human, rheumatoid-/osteoarthritis synovial fluid | 6.85 | (67) | 
| Human, rheumatoid arthritis synovial fluid | 7.03 | (68) | 
| Horse, staph. aureus-induced arthritis, synovial fluid | 6.2 | (69) | 
| Rat, BSA-induced arthritis | 5.66 | (70) | 
Protonation state of ligands
Several previous studies have demonstrated that the function of different GPCRs is pH dependent (19,21,71,72,73,74,75,76,77,78). In MORs, it was shown that ionic interactions between a protonable tertiary amine in the ligand and the amino acid residue D1473.32 are particularly important for effective binding and receptor activation (71,79). Within the binding pocket, the protonated amine forms a salt bridge with the negatively charged carboxylate of D1473.32 (80,81,82,83). The protonation state of the tertiary amine is dependent on the electron density at the nitrogen atom, which can be influenced by neighboring electronegative moieties such as halogen atoms or other groups (e.g., cyclopropanyl) (84,85,86,87) (Fig. 1). Corresponding observations were made with different GPCRs and ligands (88,89). In addition, the protonation state of H2976.52 plays an important role (71,82,90,91). It appears that ligands containing a hydroxyl group (e.g., the peptide DAMGO or the morphinan derivative naloxone) can modulate MOR signaling by forming hydrogen bonds with protonated H2976.52 (92). Besides orthosteric binding, possible allosteric interactions with the receptor following pH-dependent absorption of the ligand into the cell membrane also have to be considered (93).
Figure 1.
Chemical structures of fentanyl, N-{1-[2-(2,6-difluorphenyl)ethyl]piperidine-4-yl} N-phenylpropionamide (FF6), (±)-N-[1-(2-fluoro-2-phenylethyl)piperidine-4-yl]-N-phenyl propionamide (FF3), and (±)-N-(3-fluoro-1-phenethylpiperidine-4-yl)-N-phenyl propionamide (NFEPP). The blue circle highlights the tertiary nitrogen atom subjected to pH-dependent protonation in whose vicinity electrons may be withdrawn to reduce the pKa value. The respective isomers of FF3 and NFEPP are shown. Sites of fluorination are indicated as F. Because FF3 and FF6 elicited some adverse side effects, they were not pursued further (adapted from (114)).
Protonation state of receptors
Not only ligands but also receptors undergo changes in inflamed microenvironments. There are substantial in silico and in vitro data suggesting that both the structure and function of opioid receptors differ between healthy and inflamed/acidic conditions: 1) computational modeling studies of MORs indicate that the pH-dependent protonation state of functionally important aspartic acid and histidine residues impacts ligand binding rates and hydrogen bond networks that extend throughout the receptor, thereby influencing the intracellular G-protein-coupling dynamics (91,94). 2) Further (in vitro) studies have suggested that H2976.52 is critically involved in MOR ligand binding (71,80,81,95,96), its protonation enables hydrogen bonds with specific ligands, and MOR binding and signaling induced by these ligands are pH sensitive (92). In other GPCRs, H6.52 was also shown to be important, although its protonation state was not explicitly examined (88,97,98). 3) Additional amino acid residues whose protonation could change at low pH include D2.50, E3.28, and E6.30 (83,99,100). 4) It appears that such pH-dependent changes affect not only the structure/conformation of the receptor itself but also downstream mechanisms of G-protein coupling, arrestin binding, and membrane ion currents (7,101).
A novel prototype ligand
A novel prototype developed along these lines is (±)-N-(3-fluoro-1-phenethylpiperidine-4-yl)-N-phenylpropionamide (NFEPP) (28) (Fig. 1). At the outset, it was hypothesized that an agonist designed to selectively activate MOR at low pH will not elicit side effects typically mediated by central or intestinal MORs exposed to physiological microenvironments. In computational simulations, structural data on the MOR and its ligand fentanyl were integrated. Because fentanyl’s acid dissociation constant (pKa) is above 7.4 (Table 2), it is protonated and can activate MORs in both normal (pH 7.4) and inflamed (pH 5–7) milieus. It was then hypothesized that a ligand with a pKa between 6 and 7 should be protonated and able to activate MORs exclusively in inflamed tissue. Computational modeling studies simulated the replacement of hydrogens by fluorine in the vicinity of the tertiary amine of the ligand. The fluorine atom was chosen because it is electronegative and decreases pKa and because it would not significantly change the overall size or geometry (and hence the pharmacokinetics) of the derivative compared to the parent compound (fentanyl) (84). In addition, the binding energies of protonated and deprotonated ligands were calculated. The strong interaction between protonated fentanyl and D1473.32 in MORs was lost without protonation. The most promising candidate regarding both quantum-chemically estimated pKa values and binding energies was identified. Accordingly, NFEPP was synthesized (Fig. 1). The chemical alteration increased the overall positive charge of its amine (due to the electron-withdrawing effect that fluorine has on neighboring atoms), decreased its pKa to 6.8, and restricted its protonation and binding/activation of MORs to environments with low pH (i.e., high proton concentrations as in injured/inflamed tissue) (28,84,87,94,101).
Table 2.
pKa values of common opioid ligands
Meanwhile, this novel concept has been widely appreciated by other groups (3,4,6,10,82,83,102,103,104,105,106,107,108,109,110,111,112,113) and has been extended to other ligands (e.g., further derivatives of fentanyl or morphine) and algorithms (84,85,86,89,114) (Fig. 1).
By now, multiple in vitro and in vivo investigations performed in our own and external independent laboratories have confirmed that NFEPP indeed blocked sensory neuron excitability and produced efficient analgesia in many rat and mouse models of painful conditions such as arthritis, cancer, nerve injury (neuropathy), bowel inflammation (colitis), abdominal wall inflammation (peritonitis), surgery, and wounds. Initial translational studies confirmed such findings in human sensory neurons (48). Importantly, analgesic doses of NFEPP did not elicit any of the typical adverse effects of conventional opioids (cardio-respiratory arrest, addiction potential, constipation, sedation, locomotor disturbance, tolerance) (28,30,46,47,48,84,114,115,116).
Implications for drug design
The standard drug development model begins with target selection (e.g., a specific receptor expressed in a disease-relevant tissue) and moves onto high-throughput screens, generation of druggable lead compounds, validation of these compounds in preclinical disease models, and toxicology/safety assessments (117). Drug design strategies employ a range of medicinal chemistry methods to study structure-activity relationships. Both pharmacodynamics (e.g., potency, affinity, efficacy, selectivity) and pharmacokinetics (absorption, distribution, metabolism, excretion, toxicity) are important parameters. Molecular docking, structure-based virtual screening, and molecular dynamics are among the most frequently used strategies in the analysis of molecular recognition events such as binding energetics, molecular interactions, and conformational changes (118).
However, to date, the above described differences between diseased and normal tissues have not been considered. Indeed, these recent findings indicate that traditional drug design is limited by its focus on ligand binding to ubiquitously expressed CPCRs under normal (physiological) conditions (pH 7.4) (80,81,96,119,120,121,122,123,124). Instead, future concepts should take into account that specific microenvironments (e.g., low pH) in different compartments of the body determine the respective ligand-receptor interactions and the resulting desired or undesired effects (94,111,125). In particular, a multi-objective optimal affinity approach was proposed that aims at maximizing the binding affinity to the receptor in injured tissues (at low pH) while minimizing it in healthy environments (at normal pH) (86). In the future, such approaches may include artificial intelligence methods. Opioids are a prominent example: because all available conventional opioid ligands have pKa values above 7.4 (physiological pH) (84) (Table 2), these compounds are protonated at both low and normal pH values. Thus, they can nonselectively bind and activate opioid receptors in both injured/inflamed and healthy (brain, intestinal wall) tissues and simultaneously produce pain inhibition and the above described detrimental side effects. The pH discrepancy between healthy and inflamed tissue provides an opportunity for the design of novel ligands that bind selectively under low pH conditions. Thereby, detrimental side effects mediated by opioid receptors in healthy tissues (brain, intestinal wall) can be avoided. In addition, studies indicate that the pKa of optimal ligands should be close to the local pH of a particular disease state (84,114), calculated pKA values often differ from measured ones (84,126), future opioid drugs should not contain protonable hydroxyl groups (92), pH-sensitive compounds exhibit their maximum efficacy at peak inflammation (48), and local protons (pH) play a more important role than other components of inflammation (e.g., oxygen radicals) (101).
pH-sensitive ligands in comparison to conventional drugs
As outlined above, pH-dependent GPCR ligands can selectively activate disease-relevant receptors (pathological) while keeping nonrelevant (physiological) receptors (in healthy tissues) untouched. In the case of opioids, the cited in vivo studies have already demonstrated that this concept can give rise to effective analgesics without adverse actions. There is no doubt that novel compounds will only be attractive if they are able to produce desired effects (analgesia) comparable in potency to other available drugs. While the cited animal studies have already demonstrated this, only clinical studies in human patients will finally answer this quest. In the pain medication market, novel pH-dependent opioids will have to compete with ion channel blockers, nonsteroidal anti-inflammatory drugs (NSAIDs), or G-protein-biased agonists (110). So far, studies suggest that pH-dependent opioids will have the following advantages: 1) most painful syndromes (e.g., surgery, arthritis, neuropathy, colitis, cancer) are driven by peripheral sensory neurons (18,127,128). Opioid receptors in these neurons are upregulated, and they can synergistically modulate multiple signaling components (cAMP, Ca2+, K+, Na+, ASIC, HCN, and TRP channels) that decrease electrical excitability (4,5,6,8). This implies a much wider range of efficacy than selective blockers of individual ion channels (e.g., Nav1.7, Nav1.8, TRPV1). In addition, these ion channels are ubiquitously expressed in many organs (e.g., heart, brain, cerebellum, spinal cord, sympathetic nerves) (particularly under pathological conditions), and therefore, such drugs are bound to produce adverse side effects (e.g., arrhythmia, confusion, ataxia, temperature dysregulation) (129,130,131). 2) Less severe pain states are commonly treated with NSAIDs, but over-the-counter availability and self-medication have also led to frequent abuse and toxicity. All available NSAIDs are associated with detrimental side effects such as bleeding, gastrointestinal ulcer, heart attack, or stroke (132,133). 3) G-protein-biased opioid agonists have been shown to elicit respiratory depression, addiction potential, and constipation (134,135). 4) pH-dependent opioids do not elicit typical adverse effects of conventional opioids.
Conclusion and outlook
In conclusion, the above described studies strongly suggest distinct ligand-GPCR interactions in healthy versus injured/inflamed milieus. Apparently, local proton concentrations play a decisive role. The structure and function of not only ligands but also receptors clearly differ between those conditions. As demonstrated by, e.g., hydrogen-bonding networks, the geometric configurations (conformations) of opioid receptors localized on peripheral sensory neurons innervating injured/inflamed tissues are not identical to the (normal) ones described so far. Indeed, it appears that peripheral opioid receptors are not the same as those in the central nervous system (e.g., brain). For opioid ligands, the protonation state of tertiary amines is crucial. pH-dependent ligands might constitute a key advance in the fight against the deteriorating opioid epidemic.
Apart from GPCRs that are directly activated by protons (19,20), the pH-dependent activation of MORs by cognate ligands is the first example in the GPCR family. Future drug design should take these aspects into account and depart from traditional concepts based solely on ligand-GPCR interactions within normal (pH 7.4) microenvironments. Such approaches could also include artificial intelligence methods. Given the high degree of homology between GPCRs, these considerations should also apply to other signaling pathways (e.g., from receptor to nucleus or from endosomes), other diseases involving GPCRs (e.g., cancer, arthritis, high blood pressure, addiction, depression, Alzheimer, Parkinson, virus-induced inflammation, atherosclerosis), or even nonhuman GPCRs in deranged environments (e.g., in animals or plants exposed to ocean acidification).
Author contributions
Writing and editing, C.S.
Acknowledgments
This work was supported by the US National Science Foundation (2113030), Bundesministerium für Bildung und Forschung (01GQ2109A), Deutsche Forschungsgemeinschaft (STE 477/19-1, STE 477/21-1, STE 477/22-1, FOR 5177, and EXC 2046 AA1-15), and Canadian Institutes of Health Research (497750).
Declaration of interests
C.S. is listed as an inventor on US patent 9,133,120 B2 (“Fentanyl derivatives as pH-dependent opioid receptor agonists”) and is co-founder of pHarm Therapeutics, Inc.
Editor: Diane Lidke.
References
- 1.Hauser A.S., Attwood M.M., et al. Gloriam D.E. Trends in GPCR drug discovery: new agents, targets and indications. Nat. Rev. Drug Discov. 2017;16:829–842. doi: 10.1038/nrd.2017.178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Yang D., Zhou Q., et al. Wang M.W. G protein-coupled receptors: structure- and function-based drug discovery. Signal Transduct. Targeted Ther. 2021;6:7. doi: 10.1038/s41392-020-00435-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Manglik A. Molecular Basis of Opioid Action: From Structures to New Leads. Biol. Psychiatr. 2020;87:6–14. doi: 10.1016/j.biopsych.2019.08.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Che T., Roth B.L. Structural Insights Accelerate the Discovery of Opioid Alternatives. Annu. Rev. Biochem. 2021;90:739–761. doi: 10.1146/annurev-biochem-061620-044044. [DOI] [PubMed] [Google Scholar]
- 5.Stein C. Opioid Receptors. Annu. Rev. Med. 2016;67:433–451. doi: 10.1146/annurev-med-062613-093100. [DOI] [PubMed] [Google Scholar]
- 6.Busserolles J., Lolignier S., et al. Eschalier A. Replacement of current opioid drugs focusing on MOR-related strategies. Pharmacol. Ther. 2020;210 doi: 10.1016/j.pharmthera.2020.107519. [DOI] [PubMed] [Google Scholar]
- 7.Celik M.O., Seitz V., et al. Stein C. Modulation of G-protein activation, calcium currents and opioid receptor phosphorylation by the pH-dependent antinociceptive agonist NFEPP. Front. Mol. Neurosci. 2023;16 doi: 10.3389/fnmol.2023.1171855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Stein C. New concepts in opioid analgesia. Expet Opin. Invest. Drugs. 2018;27:765–775. doi: 10.1080/13543784.2018.1516204. [DOI] [PubMed] [Google Scholar]
- 9.Machelska H., Celik M.O. Advances in Achieving Opioid Analgesia Without Side Effects. Front. Pharmacol. 2018;9:1388. doi: 10.3389/fphar.2018.01388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Jeske N.A. Dynamic Opioid Receptor Regulation in the Periphery. Mol. Pharmacol. 2019;95:463–467. doi: 10.1124/mol.118.114637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Wei J., Lei G.H., et al. Yang T.B. Single-dose intra-articular bupivacaine versus morphine after arthroscopic knee surgery: a meta-analysis of randomized-controlled studies. Clin. J. Pain. 2014;30:630–638. doi: 10.1097/AJP.0000000000000012. [DOI] [PubMed] [Google Scholar]
- 12.Sawynok J., Liu J. Contributions of peripheral, spinal, and supraspinal actions to analgesia. Eur. J. Pharmacol. 2014;734:114–121. doi: 10.1016/j.ejphar.2014.04.006. [DOI] [PubMed] [Google Scholar]
- 13.Zeng C., Gao S.G., et al. Lei G.H. Single-dose intra-articular morphine after arthroscopic knee surgery: a meta-analysis of randomized placebo-controlled studies. Arthroscopy. 2013;29:1450–1458.e2. doi: 10.1016/j.arthro.2013.04.005. [DOI] [PubMed] [Google Scholar]
- 14.Liu Y., Wu J.S., et al. Liang X.H. Multiple Treatment Meta-Analysis of Intra-Articular Injection for Temporomandibular Osteoarthritis. J. Oral Maxillofac. Surg. 2020;78:373.e1–373.e18. doi: 10.1016/j.joms.2019.10.016. [DOI] [PubMed] [Google Scholar]
- 15.Jagla C.A., Martus P., Stein C. Peripheral opioid receptor blockade increases postoperative morphine demands - a randomized, double-blind, placebo-controlled trial. Pain. 2014;155:2056–2062. doi: 10.1016/j.pain.2014.07.011. [DOI] [PubMed] [Google Scholar]
- 16.Kominsky D.J., Campbell E.L., Colgan S.P. Metabolic shifts in immunity and inflammation. J. Immunol. 2010;184:4062–4068. doi: 10.4049/jimmunol.0903002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Erra Diaz F., Dantas E., Geffner J. Unravelling the Interplay between Extracellular Acidosis and Immune Cells. Mediat. Inflamm. 2018;2018 doi: 10.1155/2018/1218297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yoneda T., Hiasa M., et al. White F. Contribution of acidic extracellular microenvironment of cancer-colonized bone to bone pain. Biochim. Biophys. Acta. 2015;1848:2677–2684. doi: 10.1016/j.bbamem.2015.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ludwig M.G., Vanek M., et al. Seuwen K. Proton-sensing G-protein-coupled receptors. Nature. 2003;425:93–98. doi: 10.1038/nature01905. [DOI] [PubMed] [Google Scholar]
- 20.Sisignano M., Fischer M.J.M., Geisslinger G. Proton-Sensing GPCRs in Health and Disease. Cells. 2021;10 doi: 10.3390/cells10082050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zhang X.C., Sun K., et al. Cao C. GPCR activation: protonation and membrane potential. Protein Cell. 2013;4:747–760. doi: 10.1007/s13238-013-3073-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Zhang X.C., Cao C., et al. Zhao Y. Proton transfer-mediated GPCR activation. Protein Cell. 2015;6:12–17. doi: 10.1007/s13238-014-0106-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Häbler C. Über den K- und Ca-Gehalt von Eiter und Exsudaten und seine Beziehungen zum Entzündungsschmerz. Klin. Wochenschr. 1929;34:1569–1572. [Google Scholar]
- 24.Koldajew B., Altschuler M. Zur Frage über die aktive Reaktion der Exsudate bei chronischen und akuten Entzündungen. Z. Immunitätsforsch. 1930;69:18–24. [Google Scholar]
- 25.Punnia-Moorthy A. Evaluation of pH changes in inflammation of the subcutaneous air pouch lining in the rat, induced by carrageenan, dextran and Staphylococcus aureus. J. Oral Pathol. 1987;16:36–44. doi: 10.1111/j.1600-0714.1987.tb00674.x. http://www.ncbi.nlm.nih.gov/pubmed/2435877 [DOI] [PubMed] [Google Scholar]
- 26.Simmen H.P., Blaser J. Analysis of pH and pO2 in abscesses, peritoneal fluid, and drainage fluid in the presence or absence of bacterial infection during and after abdominal surgery. Am. J. Surg. 1993;166:24–27. doi: 10.1016/s0002-9610(05)80576-8. http://www.ncbi.nlm.nih.gov/pubmed/8328625 [DOI] [PubMed] [Google Scholar]
- 27.Simmen H.P., Battaglia H., et al. Blaser J. Analysis of pH, pO2 and pCO2 in drainage fluid allows for rapid detection of infectious complications during the follow-up period after abdominal surgery. Infection. 1994;22:386–389. doi: 10.1007/BF01715494. [DOI] [PubMed] [Google Scholar]
- 28.Spahn V., Del Vecchio G., et al. Stein C. A nontoxic pain killer designed by modeling of pathological receptor conformations. Science. 2017;355:966–969. doi: 10.1126/science.aai8636. [DOI] [PubMed] [Google Scholar]
- 29.Gonzalez-Rodriguez S., Quadir M.A., et al. Stein C. Polyglycerol-opioid conjugate produces analgesia devoid of side effects. Elife. 2017;6:e27081. doi: 10.7554/eLife.27081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Celik M., Negrete R., et al. Stein C. Amelioration of injury-induced tissue acidosis by a nonsteroidal analgesic attenuates antinociceptive effects of the pH-dependent opioid agonist NFEPP. Sci. Rep. 2022;12 doi: 10.1038/s41598-022-19568-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Menkin V. Studies on Inflammation: X. The Cytological Picture of an Inflammatory Exudate in Relation to its Hydrogen Ion Concentration. Am. J. Pathol. 1934;10:193–210. http://www.ncbi.nlm.nih.gov/pubmed/19970136 [PMC free article] [PubMed] [Google Scholar]
- 32.Menkin V., Warner C.R. Studies on Inflammation: XIII. Carbohydrate Metabolism, Local Acidosis, and the Cytological Picture in Inflammation. Am. J. Pathol. 1937;13:25–44.1. http://www.ncbi.nlm.nih.gov/pubmed/19970311 [PMC free article] [PubMed] [Google Scholar]
- 33.Menkin V. In: Biochemical Mechanisms in Inflammation. Thomas C.C., editor. 1956. The role of hydrogen ion concentration and the cytology of an exudate; pp. 66–103. [Google Scholar]
- 34.Hutchins G.M., Sheldon W.H. The pH of inflammatory exudates in acidotic diabetic rabbits. Proc. Soc. Exp. Biol. Med. 1972;140:623–627. doi: 10.3181/00379727-140-36517. http://www.ncbi.nlm.nih.gov/pubmed/5037600 [DOI] [PubMed] [Google Scholar]
- 35.Silver I.A. Measurement of pH and ionic composition of pericellular sites. Philos. Trans. R. Soc. Lond. B Biol. Sci. 1975;271:261–272. doi: 10.1098/rstb.1975.0050. http://www.ncbi.nlm.nih.gov/pubmed/239420 [DOI] [PubMed] [Google Scholar]
- 36.Jacobus W.E., Taylor G.J.t., et al. Nunnally R.L. Phosphorus nuclear magnetic resonance of perfused working rat hearts. Nature. 1977;265:756–758. doi: 10.1038/265756a0. http://www.ncbi.nlm.nih.gov/pubmed/16217 [DOI] [PubMed] [Google Scholar]
- 37.Levine N., Kelly H. Measurement of pH in the rat epididymis in vivo. J. Reprod. Fertil. 1978;52:333–335. doi: 10.1530/jrf.0.0520333. http://www.ncbi.nlm.nih.gov/pubmed/24738 [DOI] [PubMed] [Google Scholar]
- 38.Pan J.W., Hamm J.R., et al. Shulman R.G. Intracellular pH in human skeletal muscle by 1H NMR. Proc. Natl. Acad. Sci. USA. 1988;85:7836–7839. doi: 10.1073/pnas.85.21.7836. http://www.ncbi.nlm.nih.gov/pubmed/3186694 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Hood V.L., Schubert C., et al. Muller S. Effect of systemic pH on pHi and lactic acid generation in exhaustive forearm exercise. Am. J. Physiol. 1988;255:F479–F485. doi: 10.1152/ajprenal.1988.255.3.F479. http://www.ncbi.nlm.nih.gov/pubmed/3414804 [DOI] [PubMed] [Google Scholar]
- 40.Alfaro V., Rodenas J., et al. Carbonell T. Factors influencing the acid-base changes in the air-pouch exudate following carrageenan induced inflammation in rats. Inflamm. Res. 1996;45:405–411. doi: 10.1007/BF02252936. http://www.ncbi.nlm.nih.gov/pubmed/8872514 [DOI] [PubMed] [Google Scholar]
- 41.Issberner U., Reeh P.W., Steen K.H. Pain due to tissue acidosis: a mechanism for inflammatory and ischemic myalgia? Neurosci. Lett. 1996;208:191–194. doi: 10.1016/0304-3940(96)12576-3. http://www.ncbi.nlm.nih.gov/pubmed/8733302 [DOI] [PubMed] [Google Scholar]
- 42.Naghavi M., John R., et al. Casscells W. pH Heterogeneity of human and rabbit atherosclerotic plaques; a new insight into detection of vulnerable plaque. Atherosclerosis. 2002;164:27–35. doi: 10.1016/s0021-9150(02)00018-7. [DOI] [PubMed] [Google Scholar]
- 43.Woo Y.C., Park S.S., et al. Brennan T.J. Changes in tissue pH and temperature after incision indicate acidosis may contribute to postoperative pain. Anesthesiology. 2004;101:468–475. doi: 10.1097/00000542-200408000-00029. http://www.ncbi.nlm.nih.gov/pubmed/15277931 [DOI] [PubMed] [Google Scholar]
- 44.Shah J.P., Phillips T.M., et al. Gerber L.H. An in vivo microanalytical technique for measuring the local biochemical milieu of human skeletal muscle. J. Appl. Physiol. 2005;99:1977–1984. doi: 10.1152/japplphysiol.00419. [DOI] [PubMed] [Google Scholar]
- 45.Bez M., Zhou Z., et al. Gazit D. Molecular pain markers correlate with pH-sensitive MRI signal in a pig model of disc degeneration. Sci. Rep. 2018;8 doi: 10.1038/s41598-018-34582-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Rodriguez-Gaztelumendi A., Spahn V., et al. Stein C. Analgesic effects of a novel pH-dependent μ-opioid receptor agonist in models of neuropathic and abdominal pain. Pain. 2018;159:2277–2284. doi: 10.1097/j.pain.0000000000001328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Jimenez-Vargas N.N., Yu Y., et al. Vanner S.J. Agonist that activates the mu-opioid receptor in acidified microenvironments inhibits colitis pain without side effects. Gut. 2022;71:695–704. doi: 10.1136/gutjnl-2021-324070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Degro C.E., Jimenez-Vargas N.N., et al. Vanner S.J. Evolving acidic microenvironments during colitis provide selective analgesic targets for a pH-sensitive opioid. Pain. 2023;164:2501–2515. doi: 10.1097/j.pain.0000000000002956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Voegtlin C. The influence of the parenteral administration of certain sugars on the pH of malignant tumors. Natl. Inst. Health Bull. 1935;164:1–14. [Google Scholar]
- 50.Meyer K.A., Kammerling E.M., et al. Hoffman S.J. pH studies of malignant tissues in human beings. Cancer Res. 1948;8:513–518. http://www.ncbi.nlm.nih.gov/pubmed/18114935 [PubMed] [Google Scholar]
- 51.Revici E., Stoopen E., et al. Ravich R.A. The relation of pain to local physico-chemical changes. Bull. Inst. Appl. Biol. 1949;1:21–38. [Google Scholar]
- 52.Pampus F. [Hydrogen-Ion Concentration of Brain Tissue in Space-Occupying Intracranial Processes] Acta Neurochir. 1963;11:305–318. doi: 10.1007/BF01402010. http://www.ncbi.nlm.nih.gov/pubmed/14059253 [DOI] [PubMed] [Google Scholar]
- 53.Ashby B.S. pH studies in human malignant tumours. Lancet. 1966;2:312–315. doi: 10.1016/s0140-6736(66)92598-0. http://www.ncbi.nlm.nih.gov/pubmed/4161494 [DOI] [PubMed] [Google Scholar]
- 54.Vaupel P.W., Frinak S., Bicher H.I. Heterogeneous oxygen partial pressure and pH distribution in C3H mouse mammary adenocarcinoma. Cancer Res. 1981;41:2008–2013. http://www.ncbi.nlm.nih.gov/pubmed/7214369 [PubMed] [Google Scholar]
- 55.Newell K., Franchi A., et al. Tannock I. Studies with glycolysis-deficient cells suggest that production of lactic acid is not the only cause of tumor acidity. Proc. Natl. Acad. Sci. USA. 1993;90:1127–1131. doi: 10.1073/pnas.90.3.1127. http://www.ncbi.nlm.nih.gov/pubmed/8430084 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Gillies R.J., Liu Z., Bhujwalla Z. 31P-MRS measurements of extracellular pH of tumors using 3-aminopropylphosphonate. Am. J. Physiol. 1994;267:C195–C203. doi: 10.1152/ajpcell.1994.267.1.C195. http://www.ncbi.nlm.nih.gov/pubmed/8048479 [DOI] [PubMed] [Google Scholar]
- 57.Stubbs M., McSheehy P.M., Griffiths J.R. Causes and consequences of acidic pH in tumors: a magnetic resonance study. Adv. Enzym. Regul. 1999;39:13–30. doi: 10.1016/s0065-2571(98)00018-1. http://www.ncbi.nlm.nih.gov/pubmed/10470364 [DOI] [PubMed] [Google Scholar]
- 58.Ojugo A.S., McSheehy P.M., et al. Griffiths J.R. Measurement of the extracellular pH of solid tumours in mice by magnetic resonance spectroscopy: a comparison of exogenous (19)F and (31)P probes. NMR Biomed. 1999;12:495–504. doi: 10.1002/(sici)1099-1492(199912)12:8<495::aid-nbm594>3.0.co;2-k. http://www.ncbi.nlm.nih.gov/pubmed/10668042 [DOI] [PubMed] [Google Scholar]
- 59.Gallagher F.A., Kettunen M.I., et al. Brindle K.M. Magnetic resonance imaging of pH in vivo using hyperpolarized 13C-labelled bicarbonate. Nature. 2008;453:940–943. doi: 10.1038/nature07017. [DOI] [PubMed] [Google Scholar]
- 60.Jebens E.H., Monk-Jones M.E. On the viscosity and pH of synovial fluid and the pH of blood. J. Bone Joint Surg. Br. 1959;41-B:388–400. doi: 10.1302/0301-620X.41B2.388. http://www.ncbi.nlm.nih.gov/pubmed/13641329 [DOI] [PubMed] [Google Scholar]
- 61.Cummings N.A., Nordby G.L. Measurement of synovial fluid pH in normal and arthritic knees. Arthritis Rheum. 1966;9:47–56. doi: 10.1002/art.1780090106. http://www.ncbi.nlm.nih.gov/pubmed/4952418 [DOI] [PubMed] [Google Scholar]
- 62.Goldie I., Nachemson A. Synovial pH in rheumatoid knee-joints. I. The effect of synovectomy. Acta Orthop. Scand. 1969;40:634–641. doi: 10.3109/17453676908989529. http://www.ncbi.nlm.nih.gov/pubmed/5378127 [DOI] [PubMed] [Google Scholar]
- 63.Goldie I., Nachemson A. Synovial pH in rheumatoid knee joints. II. The effect of local corticosteroid treatment. Acta Orthop. Scand. 1970;41:354–362. doi: 10.3109/17453677008991521. http://www.ncbi.nlm.nih.gov/pubmed/5486188 [DOI] [PubMed] [Google Scholar]
- 64.Falchuk K.H., Goetzl E.J., Kulka J.P. Respiratory gases of synovial fluids. An approach to synovial tissue circulatory-metabolic imbalance in rheumatoid arthritis. Am. J. Med. 1970;49:223–231. doi: 10.1016/s0002-9343(70)80078-x. http://www.ncbi.nlm.nih.gov/pubmed/5452943 [DOI] [PubMed] [Google Scholar]
- 65.Treuhaft P.S., MCCarty D.J. Synovial fluid pH, lactate, oxygen and carbon dioxide partial pressure in various joint diseases. Arthritis Rheum. 1971;14:475–484. doi: 10.1002/art.1780140407. http://www.ncbi.nlm.nih.gov/pubmed/5564921 [DOI] [PubMed] [Google Scholar]
- 66.Ward T.T., Steigbigel R.T. Acidosis of synovial fluid correlates with synovial fluid leukocytosis. Am. J. Med. 1978;64:933–936. doi: 10.1016/0002-9343(78)90446-1. [DOI] [PubMed] [Google Scholar]
- 67.Farr M., Garvey K., et al. Bacon P.A. Significance of the hydrogen ion concentration in synovial fluid in rheumatoid arthritis. Clin. Exp. Rheumatol. 1985;3:99–104. http://www.ncbi.nlm.nih.gov/pubmed/4017318 [PubMed] [Google Scholar]
- 68.Geborek P., Saxne T., et al. Wollheim F.A. Synovial fluid acidosis correlates with radiological joint destruction in rheumatoid arthritis knee joints. J. Rheumatol. 1989;16:468–472. http://www.ncbi.nlm.nih.gov/pubmed/2746586 [PubMed] [Google Scholar]
- 69.Tulamo R.M., Bramlage L.R., Gabel A.A. Sequential clinical and synovial fluid changes associated with acute infectious arthritis in the horse. Equine Vet. J. 1989;21:325–331. doi: 10.1111/j.2042-3306.1989.tb02681.x. http://www.ncbi.nlm.nih.gov/pubmed/2776718 [DOI] [PubMed] [Google Scholar]
- 70.Andersson S.E., Lexmuller K., et al. Ekstrom G.M. Tissue and intracellular pH in normal periarticular soft tissue and during different phases of antigen induced arthritis in the rat. J. Rheumatol. 1999;26:2018–2024. http://www.ncbi.nlm.nih.gov/pubmed/10493685 [PubMed] [Google Scholar]
- 71.Surratt C.K., Johnson P.S., et al. Uhl G.R. -mu opiate receptor. Charged transmembrane domain amino acids are critical for agonist recognition and intrinsic activity. J. Biol. Chem. 1994;269:20548–20553. https://www.ncbi.nlm.nih.gov/pubmed/8051154 [PubMed] [Google Scholar]
- 72.Selley D.E., Breivogel C.S., Childers S.R. Modification of G protein-coupled functions by low-pH pretreatment of membranes from NG108-15 cells: increase in opioid agonist efficacy by decreased inactivation of G proteins. Mol. Pharmacol. 1993;44:731–741. https://www.ncbi.nlm.nih.gov/pubmed/8232223 [PubMed] [Google Scholar]
- 73.Vetter I., Kapitzke D., et al. Cabot P.J. The effects of pH on beta-endorphin and morphine inhibition of calcium transients in dorsal root ganglion neurons. J. Pain. 2006;7:488–499. doi: 10.1016/j.jpain.2006.01.456. [DOI] [PubMed] [Google Scholar]
- 74.Hofmann K.P., Scheerer P., et al. Ernst O.P. A G protein-coupled receptor at work: the rhodopsin model. Trends Biochem. Sci. 2009;34:540–552. doi: 10.1016/j.tibs.2009.07.005. [DOI] [PubMed] [Google Scholar]
- 75.Deupi X., Kobilka B.K. Energy landscapes as a tool to integrate GPCR structure, dynamics, and function. Physiology. 2010;25:293–303. doi: 10.1152/physiol.00002.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ghanouni P., Schambye H., et al. Kobilka B.K. The effect of pH on beta(2) adrenoceptor function. Evidence for protonation-dependent activation. J. Biol. Chem. 2000;275:3121–3127. doi: 10.1074/jbc.275.5.3121. http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=10652295 [DOI] [PubMed] [Google Scholar]
- 77.Ye L., Van Eps N., et al. Prosser R.S. Activation of the A2A adenosine G-protein-coupled receptor by conformational selection. Nature. 2016;533:265–268. doi: 10.1038/nature17668. [DOI] [PubMed] [Google Scholar]
- 78.Pert C.B., Snyder S.H. Properties of opiate-receptor binding in rat brain. Proc. Natl. Acad. Sci. USA. 1973;70:2243–2247. doi: 10.1073/pnas.70.8.2243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Li J.G., Chen C., et al. Liu-Chen L.Y. ASP147 in the third transmembrane helix of the rat mu opioid receptor forms ion-pairing with morphine and naltrexone. Life Sci. 1999;65:175–185. doi: 10.1016/s0024-3205(99)00234-9. [DOI] [PubMed] [Google Scholar]
- 80.Huang W., Manglik A., et al. Kobilka B.K. Structural insights into μ-opioid receptor activation. Nature. 2015;524:315–321. doi: 10.1038/nature14886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Manglik A., Lin H., et al. Shoichet B.K. Structure-based discovery of opioid analgesics with reduced side effects. Nature. 2016;537:185–190. doi: 10.1038/nature19112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Vo Q.N., Mahinthichaichan P., et al. Ellis C.R. How mu-opioid receptor recognizes fentanyl. Nat. Commun. 2021;12:984. doi: 10.1038/s41467-021-21262-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Lešnik S., Bertalan É., et al. Bondar A.N. Opioid Receptors and Protonation-Coupled Binding of Opioid Drugs. Int. J. Mol. Sci. 2021;22:13353. doi: 10.3390/ijms222413353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Spahn V., Del Vecchio G., et al. Stein C. Opioid receptor signaling, analgesic and side effects induced by a computationally designed pH-dependent agonist. Sci. Rep. 2018;8:8965. doi: 10.1038/s41598-018-27313-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Rosas R., Jr., Huang X.P., et al. Dockendorff C. β-Fluorofentanyls Are pH-Sensitive Mu Opioid Receptor Agonists. ACS Med. Chem. Lett. 2019;10:1353–1356. doi: 10.1021/acsmedchemlett.9b00335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Secker C., Fackeldey K., et al. Schutte C. Novel multi-objective affinity approach allows to identify pH-specific mu-opioid receptor agonists. J. Cheminf. 2023;15:85. doi: 10.1186/s13321-023-00746-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Lesnik S., Hodoscek M., et al. Bondar A.N. Potential Energy Function for Fentanyl-Based Opioid Pain Killers. J. Chem. Inf. Model. 2020;60:3566–3576. doi: 10.1021/acs.jcim.0c00185. [DOI] [PubMed] [Google Scholar]
- 88.Wu H., Wacker D., et al. Stevens R.C. Structure of the human kappa-opioid receptor in complex with JDTic. Nature. 2012;485:327–332. doi: 10.1038/nature10939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Augenstein M., Alexander N., Gartner M. Computational design and molecular modeling of morphine derivatives for preferential binding in inflamed tissue. Pharmacol. Res. Perspect. 2023;11 doi: 10.1002/prp2.1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Mahinthichaichan P., Vo Q.N., et al. Shen J. Kinetics and Mechanism of Fentanyl Dissociation from the mu-Opioid Receptor. JACS Au. 2021;1:2208–2215. doi: 10.1021/jacsau.1c00341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Lesnik S., Bren U., et al. Bondar A.N. Fentanyl and the Fluorinated Fentanyl Derivative NFEPP Elicit Distinct Hydrogen-Bond Dynamics of the Opioid Receptor. J. Chem. Inf. Model. 2023;63:4732–4748. doi: 10.1021/acs.jcim.3c00197. [DOI] [PubMed] [Google Scholar]
- 92.Meyer J., Del Vecchio G., et al. Stein C. Modulation of mu-opioid receptor activation by acidic pH is dependent on ligand structure and an ionizable amino acid residue. Br. J. Pharmacol. 2019;176:4510–4520. doi: 10.1111/bph.14810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Ray S., Fackeldey K., et al. Weber M. Coarse-grained MD simulations of opioid interactions with the mu-opioid receptor and the surrounding lipid membrane. Biophysica. 2023;3:263–275. doi: 10.3390/biophysica3020017. [DOI] [Google Scholar]
- 94.Ray S., Sunkara V., et al. Weber M. How to calculate pH-dependent binding rates for receptor–ligand systems based on thermodynamic simulations with different binding motifs. Mol. Simulat. 2020;46:1443–1452. doi: 10.1080/08927022.2020.1839660. [DOI] [Google Scholar]
- 95.Spivak C.E., Beglan C.L., et al. Surratt C.K. Naloxone activation of mu-opioid receptors mutated at a histidine residue lining the opioid binding cavity. Mol. Pharmacol. 1997;52:983–992. doi: 10.1124/mol.52.6.983. [DOI] [PubMed] [Google Scholar]
- 96.Koehl A., Hu H., et al. Kobilka B.K. Structure of the μ-opioid receptor-Gi protein complex. Nature. 2018;558:547–552. doi: 10.1038/s41586-018-0219-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Granier S., Manglik A., et al. Kobilka B.K. Structure of the delta-opioid receptor bound to naltrindole. Nature. 2012;485:400–404. doi: 10.1038/nature11111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Fenalti G., Giguere P.M., et al. Stevens R.C. Molecular control of delta-opioid receptor signalling. Nature. 2014;506:191–196. doi: 10.1038/nature12944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Lans I., Dalton J.A., Giraldo J. Selective Protonation of Acidic Residues Triggers Opsin Activation. J. Phys. Chem. B. 2015;119:9510–9519. doi: 10.1021/acs.jpcb.5b01908. [DOI] [PubMed] [Google Scholar]
- 100.Miao Y., Caliman A.D., McCammon J.A. Allosteric effects of sodium ion binding on activation of the m3 muscarinic g-protein-coupled receptor. Biophys. J. 2015;108:1796–1806. doi: 10.1016/j.bpj.2015.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Thies A., Sunkara V., et al. Winkelmann S. Modelling altered signalling of G-protein coupled receptors in inflamed environment to advance drug design. Sci. Rep. 2023;13:607. doi: 10.1038/s41598-023-27699-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Chan H.C.S., McCarthy D., et al. Yuan S. Designing Safer Analgesics via μ-Opioid Receptor Pathways. Trends Pharmacol. Sci. 2017;38:1016–1037. doi: 10.1016/j.tips.2017.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Camilleri M. Towards a new era with safer μ-opiate receptor analgesia. Gut. 2022;71:1–2. doi: 10.1136/gutjnl-2021-324618. [DOI] [PubMed] [Google Scholar]
- 104.Lipinski P.F.J., Kosson P., et al. Sadlej J. Fentanyl Family at the Mu-Opioid Receptor: Uniform Assessment of Binding and Computational Analysis. Molecules. 2019;24:740. doi: 10.3390/molecules24040740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Meanwell N.A. Fluorine and Fluorinated Motifs in the Design and Application of Bioisosteres for Drug Design. J. Med. Chem. 2018;61:5822–5880. doi: 10.1021/acs.jmedchem.7b01788. [DOI] [PubMed] [Google Scholar]
- 106.Seth R., Kuppalli S.S., et al. Gulati A. Recent Advances in Peripheral Opioid Receptor Therapeutics. Curr. Pain Headache Rep. 2021;25:46. doi: 10.1007/s11916-021-00951-6. [DOI] [PubMed] [Google Scholar]
- 107.Martínez V., Abalo R. Peripherally acting opioid analgesics and peripherally-induced analgesia. Behav. Pharmacol. 2020;31:136–158. doi: 10.1097/fbp.0000000000000558. [DOI] [PubMed] [Google Scholar]
- 108.Ehrlich A.T., Kieffer B.L., Darcq E. Current strategies toward safer mu opioid receptor drugs for pain management. Expert Opin. Ther. Targets. 2019;23:315–326. doi: 10.1080/14728222.2019.1586882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Raja S.N., Ringkamp M., et al. Campbell J.N. John J. Bonica Award Lecture: Peripheral neuronal hyperexcitability: the "low-hanging" target for safe therapeutic strategies in neuropathic pain. Pain. 2022;161:S14–S26. doi: 10.1097/j.pain.0000000000001838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Yekkirala A.S., Roberson D.P., et al. Woolf C.J. Breaking barriers to novel analgesic drug development. Nat. Rev. Drug Discov. 2017;16:810. doi: 10.1038/nrd.2017.202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Lohse M.J., Bock A., Zaccolo M. G Protein-Coupled Receptor Signaling: New Insights Define Cellular Nanodomains. Annu. Rev. Pharmacol. Toxicol. 2024;64:387–415. doi: 10.1146/annurev-pharmtox-040623-115054. [DOI] [PubMed] [Google Scholar]
- 112.Marti-Solano M. A multi-dimensional view of context-dependent G protein-coupled receptor function. Biochem. Soc. Trans. 2023;51:13–20. doi: 10.1042/BST20210650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Kudla L., Przewlocki R. Influence of G protein-biased agonists of mu-opioid receptor on addiction-related behaviors. Pharmacol. Rep. 2021;73:1033–1051. doi: 10.1007/s43440-021-00251-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Del Vecchio G., Labuz D., et al. Stein C. pK(a) of opioid ligands as a discriminating factor for side effects. Sci. Rep. 2019;9 doi: 10.1038/s41598-019-55886-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Massaly N., Temp J., et al. Stein C. Uncovering the analgesic effects of a pH-dependent mu-opioid receptor agonist using a model of nonevoked ongoing pain. Pain. 2020;161:2798–2804. doi: 10.1097/j.pain.0000000000001968. [DOI] [PubMed] [Google Scholar]
- 116.Baamonde A., Menéndez L., et al. Machelska H. A low pKa ligand inhibits cancer-associated pain in mice by activating peripheral mu-opioid receptors. Sci. Rep. 2020;10 doi: 10.1038/s41598-020-75509-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Woolf C.J. Overcoming obstacles to developing new analgesics. Nat. Med. 2010;16:1241–1247. doi: 10.1038/nm.2230. [DOI] [PubMed] [Google Scholar]
- 118.Ferreira L.G., Dos Santos R.N., et al. Andricopulo A.D. Molecular docking and structure-based drug design strategies. Molecules. 2015;20:13384–13421. doi: 10.3390/molecules200713384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Sounier R., Mas C., et al. Granier S. Propagation of conformational changes during mu-opioid receptor activation. Nature. 2015;524:375–378. doi: 10.1038/nature14680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Johnston J.M., Filizola M. Beyond standard molecular dynamics: investigating the molecular mechanisms of G protein-coupled receptors with enhanced molecular dynamics methods. Adv. Exp. Med. Biol. 2014;796:95–125. doi: 10.1007/978-94-007-7423-0_6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Cvijic M.E., Sum C.S., et al. Zhang L. GPCR profiling: from hits to leads and from genotype to phenotype. Drug Discov. Today Technol. 2015;18:30–37. doi: 10.1016/j.ddtec.2015.10.005. [DOI] [PubMed] [Google Scholar]
- 122.Marti-Solano M., Schmidt D., et al. Selent J. Drugging specific conformational states of GPCRs: challenges and opportunities for computational chemistry. Drug Discov. Today. 2016;21:625–631. doi: 10.1016/j.drudis.2016.01.009. [DOI] [PubMed] [Google Scholar]
- 123.Kumari P., Ghosh E., Shukla A.K. Emerging Approaches to GPCR Ligand Screening for Drug Discovery. Trends Mol. Med. 2015;21:687–701. doi: 10.1016/j.molmed.2015.09.002. [DOI] [PubMed] [Google Scholar]
- 124.Ciancetta A., Sabbadin D., et al. Moro S. Advances in Computational Techniques to Study GPCR-Ligand Recognition. Trends Pharmacol. Sci. 2015;36:878–890. doi: 10.1016/j.tips.2015.08.006. [DOI] [PubMed] [Google Scholar]
- 125.Donati L., Weber M. Assessing transition rates as functions of environmental variables. J. Chem. Phys. 2022;157 doi: 10.1063/5.0109555. [DOI] [PubMed] [Google Scholar]
- 126.Settimo L., Bellman K., Knegtel R.M. Comparison of the accuracy of experimental and predicted pKa values of basic and acidic compounds. Pharm. Res. (N. Y.) 2014;31:1082–1095. doi: 10.1007/s11095-013-1232-z. [DOI] [PubMed] [Google Scholar]
- 127.Baron R., Hans G., Dickenson A.H. Peripheral input and its importance for central sensitization. Ann. Neurol. 2013;74:630–636. doi: 10.1002/ana.24017. [DOI] [PubMed] [Google Scholar]
- 128.Berta T., Qadri Y., et al. Ji R.R. Targeting dorsal root ganglia and primary sensory neurons for the treatment of chronic pain. Expert Opin. Ther. Targets. 2017;21:695–703. doi: 10.1080/14728222.2017.1328057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Martinowich K., Das D., et al. Maher B.J. Evaluation of Na(v)1.8 as a therapeutic target for Pitt Hopkins Syndrome. Mol. Psychiatr. 2023;28:76–82. doi: 10.1038/s41380-022-01811-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Koivisto A.P., Belvisi M.G., et al. Szallasi A. Advances in TRP channel drug discovery: from target validation to clinical studies. Nat. Rev. Drug Discov. 2022;21:41–59. doi: 10.1038/s41573-021-00268-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Goodwin G., McMahon S.B. The physiological function of different voltage-gated sodium channels in pain. Nat. Rev. Neurosci. 2021;22:263–274. doi: 10.1038/s41583-021-00444-w. [DOI] [PubMed] [Google Scholar]
- 132.Bindu S., Mazumder S., Bandyopadhyay U. Non-steroidal anti-inflammatory drugs (NSAIDs) and organ damage: A current perspective. Biochem. Pharmacol. 2020;180 doi: 10.1016/j.bcp.2020.114147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Bally M., Dendukuri N., et al. Brophy J.M. Risk of acute myocardial infarction with NSAIDs in real world use: bayesian meta-analysis of individual patient data. BMJ. 2017;357 doi: 10.1136/bmj.j1909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Conibear A.E., Kelly E. A Biased View of mu-Opioid Receptors? Mol. Pharmacol. 2019;96:542–549. doi: 10.1124/mol.119.115956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Gillis A., Kliewer A., et al. Canals M. Critical Assessment of G Protein-Biased Agonism at the μ-Opioid Receptor. Trends Pharmacol. Sci. 2020;41:947–959. doi: 10.1016/j.tips.2020.09.009. [DOI] [PubMed] [Google Scholar]

