1
|
Hagerty S, Pustovyy O, Globa L, Vodyanoy V, Singletary M. Mediation of mammalian olfactory response by presence of odor-evoked potassium current. FRONTIERS IN ALLERGY 2024; 5:1478529. [PMID: 39479387 PMCID: PMC11521970 DOI: 10.3389/falgy.2024.1478529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
It is well understood that odorants interact with specialized G-protein coupled receptors embedded in the ciliary membrane of olfactory sensory neurons (OSN) which initiates a voltage-generating intracellular cascade of signal transduction events that can be recorded at the epithelial level as an electroolfactogram (EOG). While the depolarizing excitatory pathway in vertebrates involving cyclic adenosine monophosphate (cAMP)-induced Na+/Ca2+ influx and calcium-induced Cl- efflux is well established, there is evidence of potassium-associated inhibitory currents that correspond with cellular activation. While several Ca2+-dependent feedback mechanisms contribute to cellular deactivation which have been commonly attributed to these inhibitory currents, the frequently observed positive ionic conductance prior to excitatory depolarization have led many to suggest an additional earlier inhibitory mechanism at the receptor level that may be independent of downstream calcium influx. Due to conflicting conclusions, the role and mechanism behind Ca2+-independent inhibitory currents in olfactory cells is not fully understood. We investigated the functional and temporal involvement of potassium channels in odor transduction by comparing electroolfactogram (EOG) recordings in rat olfactory epithelia following ion channel inhibition and targeted activation of downstream components with or without potassium-blocking. Several K+-channel blocking agents (4-Aminopyridine, charybdotoxin, & iberiotoxin) demonstrated a diminished pre-action potential positive current that corresponded with reduced excitatory response to odor stimulation that was recovered when blockers were removed. We further assessed EOG responses in the absence of odor or with odor response enhancing zinc nanoparticles. Chemically eliciting membrane excitation in the absence of odor stimulation with a phosphodiesterase inhibitor, 3-isobutyl-1-methylxanthine (IBMX), in combination with K+-channel inhibition, further indicated potassium channel activation precedes excitatory events and is independent of cAMP-induced calcium influx. These results support previous findings of odor-activated inhibitory potassium currents that may play a functional role in subsequent G-protein activity.
Collapse
Affiliation(s)
- Samantha Hagerty
- Department of Anatomy, Physiology and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, United States
| | - Oleg Pustovyy
- Department of Anatomy, Physiology and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, United States
| | - Ludmila Globa
- Department of Anatomy, Physiology and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, United States
| | - Vitaly Vodyanoy
- Department of Anatomy, Physiology and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, United States
| | - Melissa Singletary
- Department of Anatomy, Physiology and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, United States
- Canine Performance Sciences Program, Auburn University College of Veterinary Medicine, Auburn, AL, United States
| |
Collapse
|
2
|
Oh S, Lee J, Choi HJ, Kim S, Sapuru V, Kim M, Hite RK. Discovery of Selective Inhibitors for the Lysosomal Parkinson's Disease Channel TMEM175. J Am Chem Soc 2024; 146:23230-23239. [PMID: 39116214 PMCID: PMC11513884 DOI: 10.1021/jacs.4c05623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
TMEM175 is a lysosomal potassium and proton channel that is associated with the development of Parkinson's disease. Advances in understanding the physiological roles of TMEM175 have been hampered by the absence of selective inhibitors, and studies involving genetic perturbations have yielded conflicting results. Here, we report the discovery and characterization of the first reported TMEM175-selective inhibitors, 2-phenylpyridin-4-ylamine (2-PPA), and AP-6. Cryo-EM structures of human TMEM175 bound by 2-PPA and AP-6 reveal that they act as pore blockers, binding at distinct sites in the pore and occluding the ion permeation pathway. Acute inhibition of TMEM175 by 2-PPA or AP-6 increases the level of lysosomal macromolecule catabolism, thereby accelerating macropinocytosis and other digestive processes. These inhibitors may serve as valuable tools to study the roles of TMEM175 in regulating lysosomal function and provide useful templates for future therapeutic development in Parkinson's disease.
Collapse
Affiliation(s)
- SeCheol Oh
- Structural Biology Program, Memorial Sloan Kettering Cancer Center; New York, New York 10065, USA
| | - Jooyeon Lee
- Department of Chemistry, Chungbuk National University, Republic of Korea
| | - Ho Jeong Choi
- Department of Chemistry, Chungbuk National University, Republic of Korea
| | - Songwon Kim
- Structural Biology Program, Memorial Sloan Kettering Cancer Center; New York, New York 10065, USA
| | - Vinay Sapuru
- Structural Biology Program, Memorial Sloan Kettering Cancer Center; New York, New York 10065, USA
- Physiology, Biophysics, and Systems Biology (PBSB) Program, Weill Cornell Graduate School of Biomedical Sciences, 1300 York Avenue, New York, NY, 10065, USA
| | - Min Kim
- Department of Chemistry, Chungbuk National University, Republic of Korea
| | - Richard K. Hite
- Structural Biology Program, Memorial Sloan Kettering Cancer Center; New York, New York 10065, USA
| |
Collapse
|
3
|
Sun Y, Rodríguez-Rangel S, Zhang LL, Sánchez-Rodríguez JE, Brugarolas P. Chemical and biophysical characterization of novel potassium channel blocker 3-fluoro-5-methylpyridin-4-amine. Sci Rep 2024; 14:11105. [PMID: 38750155 PMCID: PMC11096398 DOI: 10.1038/s41598-024-61465-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 05/06/2024] [Indexed: 05/18/2024] Open
Abstract
4-aminopyridine (4AP) is a potassium (K+) channel blocker used clinically to improve walking in people with multiple sclerosis (MS). 4AP binds to exposed K+ channels in demyelinated axons, reducing the leakage of intracellular K+ and enhancing impulse conduction. Multiple derivatives of 4AP capable of blocking K+ channels have been reported including three radiolabeled with positron emitting isotopes for imaging demyelinated lesions using positron emission tomography (PET). However, there remains a demand for novel molecules with suitable physicochemical properties and binding affinity that can potentially be radiolabeled and used as PET radiotracers. In this study, we introduce 3-fluoro-5-methylpyridin-4-amine (5Me3F4AP) as a novel trisubstituted K+ channel blocker with potential application in PET. 5Me3F4AP has comparable potency to 4AP and the PET tracer 3-fluoro-4-aminopyridine (3F4AP). Compared to 3F4AP, 5Me3F4AP exhibits comparable basicity (pKa = 7.46 ± 0.01 vs. 7.37 ± 0.07, P-value = 0.08), greater lipophilicity (logD = 0.664 ± 0.005 vs. 0.414 ± 0.002, P-value < 0.0001) and higher permeability to an artificial brain membrane (Pe = 88.1 ± 18.3 vs. 31.1 ± 2.9 nm/s, P-value = 0.03). 5Me3F4AP is also more stable towards oxidation in vitro by the cytochrome P450 enzyme CYP2E1 (IC50 = 36.2 ± 2.5 vs. 15.4 ± 5.1, P-value = 0.0003); the enzyme responsible for the metabolism of 4AP and 3F4AP. Taken together, 5Me3F4AP has promising properties as a candidate for PET imaging warranting additional investigation.
Collapse
Affiliation(s)
- Yang Sun
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | | | - Lauren L Zhang
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | | | - Pedro Brugarolas
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
4
|
Zhou YP, Wilks MQ, Dhaynaut M, Guehl NJ, Vesper DR, Moon SH, Rice PA, El Fakhri G, Normandin MD, Brugarolas P. Radiosynthesis automation, non-human primate biodistribution and dosimetry of K + channel tracer [ 11C]3MeO4AP. EJNMMI Res 2024; 14:43. [PMID: 38683467 PMCID: PMC11058135 DOI: 10.1186/s13550-024-01092-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/04/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND 4-Aminopyridine (4AP) is a medication for the symptomatic treatment of multiple sclerosis. Several 4AP-based PET tracers have been developed for imaging demyelination. In preclinical studies, [11C]3MeO4AP has shown promise due to its high brain permeability, high metabolic stability, high plasma availability, and high in vivo binding affinity. To prepare for the translation to human studies, we developed a cGMP-compatible automated radiosynthesis protocol and evaluated the whole-body biodistribution and radiation dosimetry of [11C]3MeO4AP in non-human primates (NHPs). METHODS Automated radiosynthesis was carried out using a GE TRACERlab FX-C Pro synthesis module. One male and one female adult rhesus macaques were used in the study. A high-resolution CT from cranial vertex to knee was acquired. PET data were collected using a dynamic acquisition protocol with four bed positions and 13 passes over a total scan time of ~ 150 min. Based on the CT and PET images, volumes of interest (VOIs) were manually drawn for selected organs. Non-decay corrected time-activity curves (TACs) were extracted for each VOI. Radiation dosimetry and effective dose were calculated from the integrated TACs using OLINDA software. RESULTS Fully automated radiosynthesis of [11C]3MeO4AP was achieved with 7.3 ± 1.2% (n = 4) of non-decay corrected radiochemical yield within 38 min of synthesis and purification time. [11C]3MeO4AP distributed quickly throughout the body and into the brain. The organs with highest dose were the kidneys. The average effective dose of [11C]3MeO4AP was 4.0 ± 0.6 μSv/MBq. No significant changes in vital signs were observed during the scan. CONCLUSION A cGMP-compatible automated radiosynthesis of [11C]3MeO4AP was developed. The whole-body biodistribution and radiation dosimetry of [11C]3MeO4AP was successfully evaluated in NHPs. [11C]3MeO4AP shows lower average effective dose than [18F]3F4AP and similar average effective dose as other carbon-11 tracers.
Collapse
Affiliation(s)
- Yu-Peng Zhou
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Bulfinch 051, Boston, MA, 02114, USA
| | - Moses Q Wilks
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Bulfinch 051, Boston, MA, 02114, USA
| | - Maeva Dhaynaut
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Bulfinch 051, Boston, MA, 02114, USA
| | - Nicolas J Guehl
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Bulfinch 051, Boston, MA, 02114, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Danielle R Vesper
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Bulfinch 051, Boston, MA, 02114, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Sung-Hyun Moon
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Bulfinch 051, Boston, MA, 02114, USA
| | - Peter A Rice
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Bulfinch 051, Boston, MA, 02114, USA
| | - Georges El Fakhri
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Bulfinch 051, Boston, MA, 02114, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Marc D Normandin
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Bulfinch 051, Boston, MA, 02114, USA.
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, 06520, USA.
| | - Pedro Brugarolas
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Bulfinch 051, Boston, MA, 02114, USA.
| |
Collapse
|
5
|
Rodríguez SG, Crosby P, Hansen LL, Grünewald E, Beale AD, Spangler RK, Rabbitts BM, Partch CL, Stangherlin A, O’Neill JS, van Ooijen G. Potassium rhythms couple the circadian clock to the cell cycle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.02.587153. [PMID: 38617352 PMCID: PMC11014554 DOI: 10.1101/2024.04.02.587153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Circadian (~24 h) rhythms are a fundamental feature of life, and their disruption increases the risk of infectious diseases, metabolic disorders, and cancer1-6. Circadian rhythms couple to the cell cycle across eukaryotes7,8 but the underlying mechanism is unknown. We previously identified an evolutionarily conserved circadian oscillation in intracellular potassium concentration, [K+]i9,10. As critical events in the cell cycle are regulated by intracellular potassium11,12, an enticing hypothesis is that circadian rhythms in [K+]i form the basis of this coupling. We used a minimal model cell, the alga Ostreococcus tauri, to uncover the role of potassium in linking these two cycles. We found direct reciprocal feedback between [K+]i and circadian gene expression. Inhibition of proliferation by manipulating potassium rhythms was dependent on the phase of the circadian cycle. Furthermore, we observed a total inhibition of cell proliferation when circadian gene expression is inhibited. Strikingly, under these conditions a sudden enforced gradient of extracellular potassium was sufficient to induce a round of cell division. Finally, we provide evidence that interactions between potassium and circadian rhythms also influence proliferation in mammalian cells. These results establish circadian regulation of intracellular potassium levels as a primary factor coupling the cell- and circadian cycles across diverse organisms.
Collapse
Affiliation(s)
- Sergio Gil Rodríguez
- School of Biological Sciences, University of Edinburgh, Max Born Crescent EH9 3BF Edinburgh, United Kingdom
| | - Priya Crosby
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, 95064, USA
| | - Louise L. Hansen
- School of Biological Sciences, University of Edinburgh, Max Born Crescent EH9 3BF Edinburgh, United Kingdom
| | - Ellen Grünewald
- School of Biological Sciences, University of Edinburgh, Max Born Crescent EH9 3BF Edinburgh, United Kingdom
| | - Andrew D. Beale
- UKRI MRC Laboratory of Molecular Biology, Francis Crick Ave, Cambridge, CB2 0QH, United Kingdom
| | - Rebecca K. Spangler
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, 95064, USA
| | - Beverley M. Rabbitts
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, 95064, USA
| | - Carrie L. Partch
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, 95064, USA
| | - Alessandra Stangherlin
- Faculty of Medicine and University Hospital Cologne, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Institute for Mitochondrial Diseases and Ageing, University of Cologne, Joseph-Stelzmann-Str, 50931, Cologne, Germany
| | - John S. O’Neill
- UKRI MRC Laboratory of Molecular Biology, Francis Crick Ave, Cambridge, CB2 0QH, United Kingdom
| | - Gerben van Ooijen
- School of Biological Sciences, University of Edinburgh, Max Born Crescent EH9 3BF Edinburgh, United Kingdom
| |
Collapse
|
6
|
McClintic WT, Chandler ZD, Karchalla LM, Ondeck CA, O'Brien SW, Campbell CJ, Jacobson AR, McNutt PM. Aminopyridines Restore Ventilation and Reverse Respiratory Acidosis at Late Stages of Botulism in Mice. J Pharmacol Exp Ther 2024; 388:637-646. [PMID: 37977816 PMCID: PMC10801772 DOI: 10.1124/jpet.123.001773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 11/19/2023] Open
Abstract
Botulinum neurotoxin (BoNT) is a potent protein toxin that causes muscle paralysis and death by asphyxiation. Treatments for symptomatic botulism are intubation and supportive care until respiratory function recovers. Aminopyridines have recently emerged as potential treatments for botulism. The clinically approved drug 3,4-diaminopyridine (3,4-DAP) rapidly reverses toxic signs of botulism and has antidotal effects when continuously administered in rodent models of lethal botulism. Although the therapeutic effects of 3,4-DAP likely result from the reversal of diaphragm paralysis, the corresponding effects on respiratory physiology are not understood. Here, we combined unrestrained whole-body plethysmography (UWBP) with arterial blood gas measurements to study the effects of 3,4-DAP, and other aminopyridines, on ventilation and respiration at terminal stages of botulism in mice. Treatment with clinically relevant doses of 3,4-DAP restored ventilation in a dose-dependent manner, producing significant improvements in ventilatory parameters within 10 minutes. Concomitant with improved ventilation, 3,4-DAP treatment reversed botulism-induced respiratory acidosis, restoring blood levels of CO2, pH, and lactate to normal physiologic levels. Having established that 3,4-DAP-mediated improvements in ventilation were directly correlated with improved respiration, we used UWBP to quantitatively evaluate nine additional aminopyridines in BoNT/A-intoxicated mice. Multiple aminopyridines were identified with comparable or enhanced therapeutic efficacies compared with 3,4-DAP, including aminopyridines that selectively improved tidal volume versus respiratory rate and vice versa. In addition to contributing to a growing body of evidence supporting the use of aminopyridines to treat clinical botulism, these data lay the groundwork for the development of aminopyridine derivatives with improved pharmacological properties. SIGNIFICANCE STATEMENT: There is a critical need for fast-acting treatments to reverse respiratory paralysis in patients with botulism. This study used unrestrained, whole-body plethysmography and arterial blood gas analysis to show that aminopyridines rapidly restore ventilation and respiration and reverse respiratory acidosis when administered to mice at terminal stages of botulism. In addition to supporting the use of aminopyridines as first-line treatments for botulism symptoms, these data are expected to contribute to the development of new aminopyridine derivatives with improved pharmacological properties.
Collapse
Affiliation(s)
- William T McClintic
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina
| | - Zachary D Chandler
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina
| | - Lalitha M Karchalla
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina
| | - Celinia A Ondeck
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina
| | - Sean W O'Brien
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina
| | - Charity J Campbell
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina
| | - Alan R Jacobson
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina
| | - Patrick M McNutt
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina
| |
Collapse
|
7
|
Geunes EP, Meinhardt JM, Wu EJ, Knowles RR. Photocatalytic Anti-Markovnikov Hydroamination of Alkenes with Primary Heteroaryl Amines. J Am Chem Soc 2023; 145:21738-21744. [PMID: 37787499 PMCID: PMC10589911 DOI: 10.1021/jacs.3c08428] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
We report a light-driven method for the intermolecular anti-Markovnikov hydroamination of alkenes with primary heteroaryl amines. In this protocol, electron transfer between an amine substrate and an excited-state iridium photocatalyst affords an aminium radical cation (ARC) intermediate that undergoes C-N bond formation with a nucleophilic alkene. Integral to reaction success is the electronic character of the amine, wherein increasingly electron-deficient heteroaryl amines generate increasingly reactive ARCs. Counteranion-dependent reactivity is observed, and iridium triflate photocatalysts are employed in place of conventional iridium hexafluorophosphate complexes. This method exhibits broad functional group tolerance across 55 examples of N-alkylated products derived from pharmaceutically relevant heteroaryl amines.
Collapse
Affiliation(s)
- Eric P Geunes
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - Jonathan M Meinhardt
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - Emily J Wu
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - Robert R Knowles
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
8
|
Sun Y, Rodríguez-Rangel S, Zhang LL, Sánchez-Rodríguez JE, Brugarolas P. Chemical and biophysical characterization of novel potassium channel blocker 3-fluoro-5-methylpyridin-4-amine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.08.550404. [PMID: 37609160 PMCID: PMC10441322 DOI: 10.1101/2023.08.08.550404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
4-aminopyridine (4AP) is a potassium (K+) channel blocker used clinically to improve walking in people with multiple sclerosis (MS). 4AP binds to exposed K+ channels in demyelinated axons, reducing the leakage of intracellular K+ and enhancing impulse conduction. Multiple derivatives of 4AP capable of blocking K+ channels have been reported including three radiolabeled with positron emitting isotopes for imaging demyelinated lesions using positron emission tomography (PET). Here, we describe 3-fluoro-5-methylpyridin-4-amine (5Me3F4AP), a novel K+ channel blocker with potential application in PET. 5Me3F4AP has comparable potency to 4AP and the PET tracer 3-fluoro-4-aminopyridine (3F4AP). Compared to 3F4AP, 5Me3F4AP is more lipophilic (logD = 0.664 ± 0.005 vs. 0.414 ± 0.002) and slightly more basic (pKa = 7.46 ± 0.01 vs. 7.37 ± 0.07). In addition, 5Me3F4AP appears to be more permeable to an artificial brain membrane and more stable towards oxidation by the cytochrome P450 enzyme family 2 subfamily E member 1 (CYP2E1), responsible for the metabolism of 4AP and 3F4AP. Taken together, 5Me3F4AP has promising properties for PET imaging warranting additional investigation.
Collapse
Affiliation(s)
- Yang Sun
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Lauren L. Zhang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Pedro Brugarolas
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Sun Y, Ramos-Torres KM, Brugarolas P. Metabolic Stability of the Demyelination Positron Emission Tomography Tracer [ 18F]3-Fluoro-4-Aminopyridine and Identification of Its Metabolites. J Pharmacol Exp Ther 2023; 386:93-101. [PMID: 37024145 PMCID: PMC10289238 DOI: 10.1124/jpet.122.001462] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 03/05/2023] [Accepted: 03/23/2023] [Indexed: 04/08/2023] Open
Abstract
[18F]3-fluoro-4-aminopyridine ([18F]3F4AP) is a positron emission tomography (PET) tracer for imaging demyelination based on the multiple sclerosis drug 4-aminopyridine (4AP, dalfampridine). This radiotracer was found to be stable in rodents and nonhuman primates imaged under isoflurane anesthesia. However, recent findings indicate that its stability is greatly decreased in awake humans and mice. Since both 4AP and isoflurane are metabolized primarily by cytochrome P450 enzymes, particularly cytochrome P450 family 2 subfamily E member 1 (CYP2E1), we postulated that this enzyme may be responsible for the metabolism of 3F4AP. Here, we investigated the metabolism of [18F]3F4AP by CYP2E1 and identified its metabolites. We also investigated whether deuteration, a common approach to increase the stability of drugs, could improve its stability. Our results demonstrate that CYP2E1 readily metabolizes 3F4AP and its deuterated analogs and that the primary metabolites are 5-hydroxy-3F4AP and 3F4AP N-oxide. Although deuteration did not decrease the rate of the CYP2E1-mediated oxidation, our findings explain the diminished in vivo stability of 3F4AP compared with 4AP and further our understanding of when deuteration may improve the metabolic stability of drugs and PET ligands. SIGNIFICANCE STATEMENT: The demyelination tracer [18F]3F4AP was found to undergo rapid metabolism in humans, which could compromise its utility. Understanding the enzymes and metabolic products involved may offer strategies to reduce metabolism. Using a combination of in vitro assays and chemical syntheses, this report shows that cytochrome P450 enzyme CYP2E1 is likely responsible for [18F]3F4AP metabolism, that 4-amino-5-fluoroprydin-3-ol (5-hydroxy-3F4AP, 5OH3F4AP) and 4-amino-3-fluoropyridine 1-oxide (3F4AP N-oxide) are the main metabolites, and that deuteration is unlikely to improve the stability of the tracer in vivo.
Collapse
Affiliation(s)
- Yang Sun
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Karla M Ramos-Torres
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Pedro Brugarolas
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
10
|
Xia C, Liu C, Ren S, Cai Y, Zhang Q, Xia C. Potassium channels, tumorigenesis and targeted drugs. Biomed Pharmacother 2023; 162:114673. [PMID: 37031494 DOI: 10.1016/j.biopha.2023.114673] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023] Open
Abstract
Potassium channels play an important role in human physiological function. Recently, various molecular mechanisms have implicated abnormal functioning of potassium channels in the proliferation, migration, invasion, apoptosis, and cancer stem cell phenotype formation. Potassium channels also mediate the association of tumor cells with the tumor microenvironment. Meanwhile, potassium channels are important targets for cancer chemotherapy. A variety of drugs exert anti-cancer effects by modulating potassium channels in tumor cells. Therefore, there is a need to understand how potassium channels participate in tumor development and progression, which could reveal new, novel targets for cancer diagnosis and treatment. This review summarizes the roles of voltage-gated potassium channels, calcium-activated potassium channels, inwardly rectifying potassium channels, and two-pore domain potassium channels in tumorigenesis and the underlying mechanism of potassium channel-targeted drugs. Therefore, the study lays the foundation for rational and effective drug design and individualized clinical therapeutics.
Collapse
Affiliation(s)
- Cong Xia
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province 116023, China
| | - Can Liu
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong Province 528099, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Shuangyi Ren
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province 116023, China
| | - Yantao Cai
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong Province 528099, China
| | - Qianshi Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province 116023, China.
| | - Chenglai Xia
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong Province 528099, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China.
| |
Collapse
|
11
|
Zhou YP, Wilks MQ, Dhaynaut M, Guehl NJ, Moon SH, Fakhri GE, Normandin MD, Brugarolas P. Radiosynthesis automation, non-human primate biodistribution and dosimetry of K + channel tracer [ 11 C]3MeO4AP. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.28.534386. [PMID: 37034655 PMCID: PMC10081174 DOI: 10.1101/2023.03.28.534386] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Purpose 4-Aminopyridine (4AP) is a medication for the symptomatic treatment of multiple sclerosis. Several 4AP-based PET tracers have been developed for imaging demyelination. In preclinical studies, [ 11 C]3MeO4AP has shown promise due to its high brain permeability, high metabolic stability, high plasma availability, and high in vivo binding affinity. To prepare for the translation to human studies, we developed a cGMP-compliant automated radiosynthesis protocol and evaluated the whole-body biodistribution and radiation dosimetry of [ 11 C]3MeO4AP in non-human primates (NHPs). Methods Automated radiosynthesis was carried out using a GE TRACERlab FX-C Pro synthesis module. One male and one female adult rhesus macaques were used in the study. A high-resolution CT from cranial vertex to knee was acquired. PET data were collected using a dynamic acquisition protocol with 4 bed positions and 13 passes over a total scan time of ∼150 minutes. Based on the CT and PET images, volumes of interest (VOIs) were manually drawn for selected organs. Non-decay corrected time-activity curves (TACs) were extracted for each VOI. Radiation dosimetry and effective dose were calculated from the integrated TACs using OLINDA software. Results Fully automated radiosynthesis of [ 11 C]3MeO4AP was achieved with 7.3 ± 1.2 % (n = 4) of non-decay corrected radiochemical yield within 38 min of synthesis and purification time. [ 11 C]3MeO4AP distributed quickly throughout the body and into the brain. The organs with highest dose were the kidneys. The average effective dose of [ 11 C]3MeO4AP was 4.27 ± 0.57 μSv/MBq. No significant changes in vital signs were observed during the scan. Conclusion The cGMP compliant automated radiosynthesis of [ 11 C]3MeO4AP was developed. The whole-body biodistribution and radiation dosimetry of [ 11 C]3MeO4AP was successfully evaluated in NHPs. [ 11 C]3MeO4AP shows lower average effective dose than [ 18 F]3F4AP and similar average effective dose as other carbon-11 tracers.
Collapse
|
12
|
Sun Y, Guehl NJ, Zhou YP, Takahashi K, Belov V, Dhaynaut M, Moon SH, El Fakhri G, Normandin MD, Brugarolas P. Radiochemical Synthesis and Evaluation of 3-[ 11C]Methyl-4-aminopyridine in Rodents and Nonhuman Primates for Imaging Potassium Channels in the CNS. ACS Chem Neurosci 2022; 13:3342-3351. [PMID: 36417797 PMCID: PMC9732819 DOI: 10.1021/acschemneuro.2c00364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Demyelination, the loss of the insulating sheath of neurons, causes failed or slowed neuronal conduction and contributes to the neurological symptoms in multiple sclerosis, traumatic brain and spinal cord injuries, stroke, and dementia. In demyelinated neurons, the axonal potassium channels Kv1.1 and Kv1.2, generally under the myelin sheath, become exposed and upregulated. Therefore, imaging these channels using positron emission tomography can provide valuable information for disease diagnosis and monitoring. Here, we describe a novel tracer for Kv1 channels, [11C]3-methyl-4-aminopyridine ([11C]3Me4AP). [11C]3Me4AP was efficiently synthesized via Pd(0)-Cu(I) comediated Stille cross-coupling of a stannyl precursor containing a free amino group. Evaluation of its imaging properties in rats and nonhuman primates showed that [11C]3Me4AP has a moderate brain permeability and slow kinetics. Additional evaluation in monkeys showed that the tracer is metabolically stable and that a one-tissue compartment model can accurately model the regional brain time-activity curves. Compared to the related tracers [18F]3-fluoro-4-aminopyridine ([18F]3F4AP) and [11C]3-methoxy-4-aminopyridine ([11C]3MeO4AP), [11C]3Me4AP shows lower initial brain uptake, which indicates reduced permeability to the blood-brain barrier and slower kinetics, suggesting higher binding affinity consistent with in vitro studies. While the slow kinetics and strong binding affinity resulted in a tracer with less favorable properties for imaging the brain than its predecessors, these properties may make 3Me4AP useful as a therapeutic.
Collapse
|
13
|
Norcott PL. Selective NMR detection of individual reaction components hyperpolarised by reversible exchange with para-hydrogen. Phys Chem Chem Phys 2022; 24:13527-13533. [DOI: 10.1039/d2cp01657e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
NMR spectroscopy can sometimes be hampered by two inherent weaknesses: low sensitivity and overlap of signals in complex mixtures. Hyperpolarisation techniques using para-hydrogen (including the method known as SABRE) can...
Collapse
|
14
|
Meena P, Singh A, patel M, Verma AK. Transition-Metal-Free Regioselective Hydroamination of Styrenes with Amino-Heteroarenes. Chem Commun (Camb) 2022; 58:8424-8427. [DOI: 10.1039/d2cc02781j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Base-mediated anti-Markovnikov hydroamination of functionally varied styrenes with amino-substituted pyridine, quinoline, pyrimidine, pyrazine, and phenanthridine with excellent regioselectivity has been described. Double hydroamination was observed chemoselectively on secondary amine leaving...
Collapse
|
15
|
Positron emission tomography in multiple sclerosis - straight to the target. Nat Rev Neurol 2021; 17:663-675. [PMID: 34545219 DOI: 10.1038/s41582-021-00537-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 02/08/2023]
Abstract
Following the impressive progress in the treatment of relapsing-remitting multiple sclerosis (MS), the major challenge ahead is the development of treatments to prevent or delay the irreversible accumulation of clinical disability in progressive forms of the disease. The substrate of clinical progression is neuro-axonal degeneration, and a deep understanding of the mechanisms that underlie this process is a precondition for the development of therapies for progressive MS. PET imaging involves the use of radiolabelled compounds that bind to specific cellular and metabolic targets, thereby enabling direct in vivo measurement of several pathological processes. This approach can provide key insights into the clinical relevance of these processes and their chronological sequence during the disease course. In this Review, we focus on the contribution that PET is making to our understanding of extraneuronal and intraneuronal mechanisms that are involved in the pathogenesis of irreversible neuro-axonal damage in MS. We consider the major challenges with the use of PET in MS and the steps necessary to realize clinical benefits of the technique. In addition, we discuss the potential of emerging PET tracers and future applications of existing compounds to facilitate the identification of effective neuroprotective treatments for patients with MS.
Collapse
|
16
|
A Novel KCNA2 Variant in a Patient with Non-Progressive Congenital Ataxia and Epilepsy: Functional Characterization and Sensitivity to 4-Aminopyridine. Int J Mol Sci 2021; 22:ijms22189913. [PMID: 34576077 PMCID: PMC8469797 DOI: 10.3390/ijms22189913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/02/2021] [Accepted: 09/08/2021] [Indexed: 12/26/2022] Open
Abstract
Kv1.2 channels, encoded by the KCNA2 gene, are localized in the central and peripheral nervous system, where they regulate neuronal excitability. Recently, heterozygous mutations in KCNA2 have been associated with a spectrum of symptoms extending from epileptic encephalopathy, intellectual disability, and cerebellar ataxia. Patients are treated with a combination of antiepileptic drugs and 4-aminopyridine (4-AP) has been recently trialed in specific cases. We identified a novel variant in KCNA2, E236K, in a Serbian proband with non-progressive congenital ataxia and early onset epilepsy, treated with sodium valproate. To ascertain the pathogenicity of E236K mutation and to verify its sensitivity to 4-AP, we transfected HEK 293 cells with Kv1.2 WT or E236K cDNAs and recorded potassium currents through the whole-cell patch-clamp. In silico analysis supported the electrophysiological data. E236K channels showed voltage-dependent activation shifted towards negative potentials and slower kinetics of deactivation and activation compared with Kv1.2 WT. Heteromeric Kv1.2 WT+E236K channels, resembling the condition of the heterozygous patient, confirmed a mixed gain- and loss-of-function (GoF/LoF) biophysical phenotype. 4-AP inhibited both Kv1.2 and E236K channels with similar potency. Homology modeling studies of mutant channels suggested a reduced interaction between the residue K236 in the S2 segment and the gating charges at S4. Overall, the biophysical phenotype of E236K channels correlates with the mild end of the clinical spectrum reported in patients with GoF/LoF defects. The response to 4-AP corroborates existing evidence that KCNA2-disorders could benefit from variant-tailored therapeutic approaches, based on functional studies.
Collapse
|
17
|
Guehl NJ, Ramos-Torres KM, Linnman C, Moon SH, Dhaynaut M, Wilks MQ, Han PK, Ma C, Neelamegam R, Zhou YP, Popko B, Correia JA, Reich DS, Fakhri GE, Herscovitch P, Normandin MD, Brugarolas P. Evaluation of the potassium channel tracer [ 18F]3F4AP in rhesus macaques. J Cereb Blood Flow Metab 2021; 41:1721-1733. [PMID: 33090071 PMCID: PMC8221756 DOI: 10.1177/0271678x20963404] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Demyelination causes slowed or failed neuronal conduction and is a driver of disability in multiple sclerosis and other neurological diseases. Currently, the gold standard for imaging demyelination is MRI, but despite its high spatial resolution and sensitivity to demyelinated lesions, it remains challenging to obtain specific and quantitative measures of molecular changes involved in demyelination. To understand the contribution of demyelination in different diseases and to assess the efficacy of myelin-repair therapies, it is critical to develop new in vivo imaging tools sensitive to changes induced by demyelination. Upon demyelination, axonal K+ channels, normally located underneath the myelin sheath, become exposed and increase in expression, causing impaired conduction. Here, we investigate the properties of the K+ channel PET tracer [18F]3F4AP in primates and its sensitivity to a focal brain injury that occurred three years prior to imaging. [18F]3F4AP exhibited favorable properties for brain imaging including high brain penetration, high metabolic stability, high plasma availability, high reproducibility, high specificity, and fast kinetics. [18F]3F4AP showed preferential binding in areas of low myelin content as well as in the previously injured area. Sensitivity of [18F]3F4AP for the focal brain injury was higher than [18F]FDG, [11C]PiB, and [11C]PBR28, and compared favorably to currently used MRI methods.
Collapse
Affiliation(s)
- Nicolas J Guehl
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Karla M Ramos-Torres
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Clas Linnman
- Spaulding Neuroimaging Lab, Spaulding Rehabilitation Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Sung-Hyun Moon
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Maeva Dhaynaut
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Moses Q Wilks
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Paul K Han
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Chao Ma
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ramesh Neelamegam
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yu-Peng Zhou
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Brian Popko
- Department of Neurology, Northwestern Feinberg School of Medicine, Chicago, IL, USA
| | - John A Correia
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Herscovitch
- Positron Emission Tomography Department, NIH Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Marc D Normandin
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Pedro Brugarolas
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
18
|
De Jesús-Pérez JJ, Méndez-Maldonado GA, López-Romero AE, Esparza-Jasso D, González-Hernández IL, De la Rosa V, Gastélum-Garibaldi R, Sánchez-Rodríguez JE, Arreola J. Electro-steric opening of the CLC-2 chloride channel gate. Sci Rep 2021; 11:13127. [PMID: 34162897 PMCID: PMC8222222 DOI: 10.1038/s41598-021-92247-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 06/04/2021] [Indexed: 01/18/2023] Open
Abstract
The widely expressed two-pore homodimeric inward rectifier CLC-2 chloride channel regulates transepithelial chloride transport, extracellular chloride homeostasis, and neuronal excitability. Each pore is independently gated at hyperpolarized voltages by a conserved pore glutamate. Presumably, exiting chloride ions push glutamate outwardly while external protonation stabilizes it. To understand the mechanism of mouse CLC-2 opening we used homology modelling-guided structure-function analysis. Structural modelling suggests that glutamate E213 interacts with tyrosine Y561 to close a pore. Accordingly, Y561A and E213D mutants are activated at less hyperpolarized voltages, re-opened at depolarized voltages, and fast and common gating components are reduced. The double mutant cycle analysis showed that E213 and Y561 are energetically coupled to alter CLC-2 gating. In agreement, the anomalous mole fraction behaviour of the voltage dependence, measured by the voltage to induce half-open probability, was strongly altered in these mutants. Finally, cytosolic acidification or high extracellular chloride concentration, conditions that have little or no effect on WT CLC-2, induced reopening of Y561 mutants at positive voltages presumably by the inward opening of E213. We concluded that the CLC-2 gate is formed by Y561-E213 and that outward permeant anions open the gate by electrostatic and steric interactions.
Collapse
Affiliation(s)
- José J De Jesús-Pérez
- Physics Institute, Universidad Autónoma de San Luis Potosí, Ave. Dr. Manuel Nava #6, 78290, San Luis Potosí, SLP, Mexico
| | - G Arlette Méndez-Maldonado
- Departamento de Física, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Blvd. M. García Barragán #1421, 44430, Guadalajara, Jalisco, Mexico
| | - Ana E López-Romero
- Physics Institute, Universidad Autónoma de San Luis Potosí, Ave. Dr. Manuel Nava #6, 78290, San Luis Potosí, SLP, Mexico
| | - David Esparza-Jasso
- Physics Institute, Universidad Autónoma de San Luis Potosí, Ave. Dr. Manuel Nava #6, 78290, San Luis Potosí, SLP, Mexico
| | - Irma L González-Hernández
- Departamento de Física, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Blvd. M. García Barragán #1421, 44430, Guadalajara, Jalisco, Mexico
| | - Víctor De la Rosa
- CONACYT, School of Medicine, Universidad Autónoma de San Luis Potosí, Ave. V. Carranza 2005, Los Filtros, 78290, San Luis Potosí, SLP, Mexico
| | - Roberto Gastélum-Garibaldi
- Departamento de Física, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Blvd. M. García Barragán #1421, 44430, Guadalajara, Jalisco, Mexico
| | - Jorge E Sánchez-Rodríguez
- Departamento de Física, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Blvd. M. García Barragán #1421, 44430, Guadalajara, Jalisco, Mexico
| | - Jorge Arreola
- Physics Institute, Universidad Autónoma de San Luis Potosí, Ave. Dr. Manuel Nava #6, 78290, San Luis Potosí, SLP, Mexico.
| |
Collapse
|
19
|
Chitra R, Choudhury R, Capet F, Roussel P, Bhatt P. Crystal structure of 4-aminopyridinium 3-(4-aminopyridinium) succinate tetra hydrate: A new salt from 4-aminopyridine and maleic acid crystallization. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
20
|
Naz S, Shah FA, Nadeem H, Sarwar S, Tan Z, Imran M, Ali T, Li JB, Li S. Amino Acid Conjugates of Aminothiazole and Aminopyridine as Potential Anticancer Agents: Synthesis, Molecular Docking and in vitro Evaluation. Drug Des Devel Ther 2021; 15:1459-1476. [PMID: 33833504 PMCID: PMC8021256 DOI: 10.2147/dddt.s297013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/27/2021] [Indexed: 12/15/2022] Open
Abstract
PURPOSE The development of resistance to available anticancer drugs is increasingly becoming a major challenge and new chemical entities could be unveiled to compensate this therapeutic failure. The current study demonstrated the synthesis of 2-aminothiazole [S3(a-d) and S5(a-d)] and 2-aminopyridine [S4(a-d) and S6(a-d)] derivatives that can target multiple cellular networks implicated in cancer development. METHODS Biological assays were performed to investigate the antioxidant and anticancer potential of synthesized compounds. Redox imbalance and oxidative stress are hallmarks of cancer, therefore, synthesized compounds were preliminarily screened for their antioxidant activity using DPPH assay, and further five derivatives S3b, S3c, S4c, S5b, and S6c, with significant antioxidant potential, were selected for investigation of in vitro anticancer potential. The cytotoxic activities were evaluated against the parent (A2780) and cisplatin-resistant (A2780CISR) ovarian cancer cell lines. Further, Molecular docking studies of active compounds were performed to determine binding affinities. RESULTS Results revealed that S3c, S5b, and S6c displayed promising inhibition in cisplatin-resistant cell lines in comparison to parent cells in terms of both resistance factor (RF) and IC50 values. Moreover, S3c proved to be most active compound in both parent and resistant cell lines with IC50 values 15.57 µM and 11.52 µM respectively. Our docking studies demonstrated that compounds S3c, S5b, and S6c exhibited significant binding affinity with multiple protein targets of the signaling cascade. CONCLUSION Anticancer activities of compounds S3c, S5b, and S6c in cisplatin-resistant cell lines suggested that these ligands may contribute as lead compounds for the development of new anticancer drugs.
Collapse
Affiliation(s)
- Shagufta Naz
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, 44000, Pakistan
- Shenzhen University Clinical Research Center for Neurological Diseases, Health Management Center, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen, People's Republic of China
| | - Fawad Ali Shah
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, 44000, Pakistan
| | - Humaira Nadeem
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, 44000, Pakistan
| | - Sadia Sarwar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, 44000, Pakistan
| | - Zhen Tan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Muhammad Imran
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, 44000, Pakistan
| | - Tahir Ali
- Shenzhen University Clinical Research Center for Neurological Diseases, Health Management Center, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen, People's Republic of China
| | - Jing Bo Li
- Shenzhen University Clinical Research Center for Neurological Diseases, Health Management Center, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen, People's Republic of China
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, People’s Republic of China
| |
Collapse
|
21
|
Ryzhkina I, Murtazina L, Gainutdinov K, Konovalov A. Diluted Aqueous Dispersed Systems of 4-Aminopyridine: The Relationship of Self-Organization, Physicochemical Properties, and Influence on the Electrical Characteristics of Neurons. Front Chem 2021; 9:623860. [PMID: 33796504 PMCID: PMC8007878 DOI: 10.3389/fchem.2021.623860] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/01/2021] [Indexed: 11/22/2022] Open
Abstract
A variety of physicochemical methods were used to examine the self-organization, physicochemical, UV absorption, and fluorescent properties of diluted aqueous solutions (calculated concentrations from 1·10-20 to 1·10-2 M) of the membrane voltage-dependent potassium channels blocker 4-aminopyridine (4-AP). Using the dynamic light scattering method, it was shown that 4-AP solutions at concentrations in the range of 1·10-20-1·10-6 M are dispersed systems in which domains and nanoassociates of hundreds of nm in size are formed upon dilution. An interrelation between the non-monotonic concentration dependencies of the size of the dispersed phase, the fluorescence intensity (λ ex 225 nm, λ em 340 nm), specific electrical conductivity, and pH has been established. This allows us to predict the bioeffects of the 4-AP systems at low concentrations. The impact of these diluted aqueous systems on the electrical characteristics of identified neurons of Helix lucorum snails was studied. Incubation of neurons in the 4-AP systems for which the formation of domains and nanoassociates had been established lead to a nonmonotonic decrease of the resting potential by 7-13%. An analysis of the obtained results and published data allows for a conclusion that a consistent change in the nature and parameters of the dispersed phase, as well as the pH of the medium, apparently determines the nonmonotonic nature of the effect of the 4-AP systems in a 1·10-20-1·10-6 M concentration range on the resting membrane potential of neurons. It was found that the pre-incubation of neurons in the 4-AP system with a concentration of 1·10-12 M led to a 17.0% synergistic decrease in the membrane potential after a subsequent treatment with 1·10-2 M 4-AP solution. This finding demonstrates a significant modifying effect of self-organized dispersed systems of 4-AP in low concentrations on the neurons' sensitivity to 4-AP.
Collapse
Affiliation(s)
- Irina Ryzhkina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russia
| | - Lyaisan Murtazina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russia
| | - Khalil Gainutdinov
- Zavoisky Physical-Technical Institute, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russia
| | - Alexander Konovalov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russia
| |
Collapse
|
22
|
Dietrich M, Hartung HP, Albrecht P. Neuroprotective Properties of 4-Aminopyridine. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/3/e976. [PMID: 33653963 PMCID: PMC7931640 DOI: 10.1212/nxi.0000000000000976] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023]
Abstract
As an antagonist of voltage-gated potassium (Kv) channels, 4-aminopyridine (4-AP) is used as symptomatic therapy in several neurologic disorders. The improvement of visual function and motor skills and relieve of fatigue in patients with MS have been attributed to 4-AP. Its prolonged release formulation (fampridine) has been approved for the symptomatic treatment of walking disability in MS. The beneficial effects were explained by the blockade of axonal Kv channels, thereby enhancing conduction along demyelinated axons. However, an increasing body of evidence suggests that 4-AP may have additional properties beyond the symptomatic mode of action. In this review, we summarize preclinical and clinical data on possible neuroprotective features of 4-AP.
Collapse
Affiliation(s)
- Michael Dietrich
- From the Department of Neurology (M.D., H.-P.H., P.A.), Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany; and Brain and Mind Center (H.-P.H.), University of Sydney, Australia
| | - Hans-Peter Hartung
- From the Department of Neurology (M.D., H.-P.H., P.A.), Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany; and Brain and Mind Center (H.-P.H.), University of Sydney, Australia
| | - Philipp Albrecht
- From the Department of Neurology (M.D., H.-P.H., P.A.), Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany; and Brain and Mind Center (H.-P.H.), University of Sydney, Australia.
| |
Collapse
|
23
|
Guehl NJ, Neelamegam R, Zhou YP, Moon SH, Dhaynaut M, El Fakhri G, Normandin MD, Brugarolas P. Radiochemical Synthesis and Evaluation in Non-Human Primates of 3-[ 11C]methoxy-4-aminopyridine: A Novel PET Tracer for Imaging Potassium Channels in the CNS. ACS Chem Neurosci 2021; 12:756-765. [PMID: 33539063 DOI: 10.1021/acschemneuro.0c00791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Demyelination, the loss of the protecting sheath of neurons, contributes to disability in many neurological diseases. In order to fully understand its role in different diseases and to monitor treatments aiming at reversing this process, it would be valuable to have PET radiotracers that can detect and quantify molecular changes involved in demyelination such as the uncovering and upregulation of the axonal potassium channels Kv1.1 and Kv1.2. Carbon-11 labeled radiotracers present the advantage of allowing for multiple scans on the same subject in the same day. Here, we describe [11C]3MeO4AP, a novel 11C-labeled version of the K+ channel tracer [18F]3F4AP, and characterize its imaging properties in two non-human primates including a monkey with a focal brain injury sustained during a surgical procedure 3 years prior to imaging. Our findings show that [11C]3MeO4AP is brain permeable, metabolically stable and has high plasma availability. When compared with [18F]3F4AP, [11C]3MeO4AP shows very high correlation in volumes of distribution (VT), confirming a common target. [11C]3MeO4AP shows slower washout than [18F]3F4AP, suggesting stronger binding. Finally, similar to [18F]3F4AP, [11C]3MeO4AP is highly sensitive to the focal brain injury. All these features make it a promising radioligand for imaging demyelinated lesions.
Collapse
Affiliation(s)
- Nicolas J. Guehl
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Ramesh Neelamegam
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Yu-Peng Zhou
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Sung-Hyun Moon
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Maeva Dhaynaut
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Marc D. Normandin
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Pedro Brugarolas
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| |
Collapse
|
24
|
Türker F, Noma SAA, Aktaş A, Al-Khafaji K, Taşkın Tok T, Ateş B, Gök Y. The (NHC)PdBr2(2-aminopyridine) complexes: synthesis, characterization, molecular docking study, and inhibitor effects on the human serum carbonic anhydrase and serum bovine xanthine oxidase. MONATSHEFTE FUR CHEMIE 2020. [DOI: 10.1007/s00706-020-02687-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
25
|
Ramos-Torres KM, Zhou YP, Yang BY, Guehl NJ, Sung-Hyun M, Telu S, Normandin MD, Pike VW, Brugarolas P. Syntheses of [ 11C]2- and [ 11C]3-trifluoromethyl-4-aminopyridine: potential PET radioligands for demyelinating diseases. RSC Med Chem 2020; 11:1161-1167. [PMID: 33479620 PMCID: PMC7651860 DOI: 10.1039/d0md00190b] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/17/2020] [Indexed: 11/21/2022] Open
Abstract
Trifluoromethyl groups are of great interest in PET radiopharmaceuticals. Radiolabelled 4-aminopyridine (4AP) derivatives have been proposed for imaging demyelinating diseases. Here, we describe methods for producing 11C-trifluoromethylated derivatives of 4AP and present early imaging results with [11C]3-trifluoromethyl-4AP in a rhesus macaque. This study shows the utility of [11C]CuCF3 for labelling pyridines and provides initial evidence for the potential use of [11C]3-trifluoromethyl-4AP as a PET radioligand.
Collapse
Affiliation(s)
- Karla M Ramos-Torres
- Gordon Center for Medical Imaging , Department of Radiology , Massachusetts General Hospital and Harvard Medical School , Boston , MA , USA .
| | - Yu-Peng Zhou
- Gordon Center for Medical Imaging , Department of Radiology , Massachusetts General Hospital and Harvard Medical School , Boston , MA , USA .
| | - Bo Yeun Yang
- Molecular Imaging Branch , National Institute of Mental Health , National Institutes of Health , Bethesda , MD , USA .
| | - Nicolas J Guehl
- Gordon Center for Medical Imaging , Department of Radiology , Massachusetts General Hospital and Harvard Medical School , Boston , MA , USA .
| | - Moon Sung-Hyun
- Gordon Center for Medical Imaging , Department of Radiology , Massachusetts General Hospital and Harvard Medical School , Boston , MA , USA .
| | - Sanjay Telu
- Molecular Imaging Branch , National Institute of Mental Health , National Institutes of Health , Bethesda , MD , USA .
| | - Marc D Normandin
- Gordon Center for Medical Imaging , Department of Radiology , Massachusetts General Hospital and Harvard Medical School , Boston , MA , USA .
| | - Victor W Pike
- Molecular Imaging Branch , National Institute of Mental Health , National Institutes of Health , Bethesda , MD , USA .
| | - Pedro Brugarolas
- Gordon Center for Medical Imaging , Department of Radiology , Massachusetts General Hospital and Harvard Medical School , Boston , MA , USA .
| |
Collapse
|