Konstantinos Plakas, Chia-Ju Hsieh, Dinahlee Saturnino Guarino, Catherine Hou, Wai-Kit Chia, Anthony Young, Alexander Schmitz, Yi-Pei Ho, Chi-Chang Weng, Hsiaoju Lee, Shihong Li, Thomas J A Graham, Robert H Mach
{"title":"Toward a Small-Molecule Antagonist Radioligand for Positron Emission Tomography Imaging of the Mu Opioid Receptor.","authors":"Konstantinos Plakas, Chia-Ju Hsieh, Dinahlee Saturnino Guarino, Catherine Hou, Wai-Kit Chia, Anthony Young, Alexander Schmitz, Yi-Pei Ho, Chi-Chang Weng, Hsiaoju Lee, Shihong Li, Thomas J A Graham, Robert H Mach","doi":"10.1021/acschemneuro.5c00140","DOIUrl":"10.1021/acschemneuro.5c00140","url":null,"abstract":"<p><p>The opioid crisis is a catastrophic health emergency catalyzed by the misuse of opioids that target and activate the mu opioid receptor. Many traditional radioligands used to study the mu opioid receptor are often tightly regulated owing to their abuse and respiratory depression potential. Of those that are not regulated, a lack of opioid receptor subtype selectivity can cause confounding in interpreting results. In the present study, we sought to design and characterize a library of 24 antagonist ligands for the mu opioid receptor. Ligands were evaluated for the binding affinity, intrinsic activity, and predicted blood-brain barrier permeability. Several ligands demonstrated single-digit nM binding affinity for the mu opioid receptor while also demonstrating selectivity over the delta and kappa opioid receptors. The antagonist behavior of <b>1A</b> and <b>3A</b> at the mu opioid receptor indicate that these ligands would likely not induce opioid-dependent respiratory depression. Therefore, these ligands can enable a safer means to interrogate the endogenous opioid system. Based on binding affinity, selectivity, and potential off-target binding, [<sup>11</sup>C]<b>1A</b> was prepared via metallophotoredox of the aryl-bromide functional group to [<sup>11</sup>C]methyl iodide. The nascent radioligand demonstrated brain uptake in a rhesus macaque model and accumulation in the caudate and putamen. Naloxone was able to reduce [<sup>11</sup>C]<b>1A</b> binding, though the interactions were not as pronounced as naloxone's ability to displace [<sup>11</sup>C]carfentanil. These results suggest that GSK1521498 and related congeners are amenable to radioligand design and can offer a safer way to query opioid neurobiology.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2025-03-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143741719","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Enam Alhagh Gorgich, Zahra Heidari, Hamidreza Mahmoudzadeh-Sagheb, Auob Rustamzadeh, Arash Shabani, Ali Amirzadeh, Bahram Haghi Ashtiani
{"title":"Brain Metabolite Profiles are Associated with Selective Neuronal Vulnerability and Underlying Mechanisms in Amyotrophic Lateral Sclerosis.","authors":"Enam Alhagh Gorgich, Zahra Heidari, Hamidreza Mahmoudzadeh-Sagheb, Auob Rustamzadeh, Arash Shabani, Ali Amirzadeh, Bahram Haghi Ashtiani","doi":"10.1021/acschemneuro.4c00593","DOIUrl":"https://doi.org/10.1021/acschemneuro.4c00593","url":null,"abstract":"<p><p>Amyotrophic lateral sclerosis (ALS) is a lethal neurological syndrome accompanied by selective degeneration of somatic motor neurons and neurochemistry alterations. Nevertheless, eye movement's nuclei are relatively spared from ALS damage. This survey was to probe metabolite changes in the primary motor cortex (PMC) and interstitial nucleus of Cajal (INC) of ALS patients using proton magnetic resonance spectroscopy (<sup>1</sup>H-MRS). In this case-control study, 20 patients with ALS and 20 healthy controls underwent 1.5 T MRI and multivoxel <sup>1</sup>H-MRS. <sup>1</sup>H-MRS spectra to determine metabolite profiles including tNAA, mIns, tCr, tCho, and also tNAA/tCr, tNAA/tCho, and mIns/tNAA metabolite ratios from the PMC and INC were quantified via a point resolved spectroscopy pulse (PRESS) sequence in two groups. Further, the associations between <sup>1</sup>H-MRS markers with forced vital capacity (FVC), ALS functional rating scale (ALSFRS-R), and disease progression rate (ΔFS) were investigated. In the PMC, tNAA and tNAA/tCr were significantly lower in ALS patients than the healthy controls, but mIns and mIns/tNAA were significantly greater in these patients (<i>p</i> < 0.05). In the INC, tCho and mIns concentrations, and mIns/tNAA ratio were significantly increased (<i>p</i> < 0.05) in ALS patients, while tNAA and tNAA/tCr ratio did not show significant discriminations between the two groups (<i>p</i> > 0.05). The PMC tNAA/Cr ratio is associated with ALSFRS-R (<i>p</i> = 0.001, <i>r</i> = 0.71), FVC (<i>p</i> = 0.03, <i>r</i> = 0.58), and ΔFS (<i>p</i> = 0.01, <i>r</i> = -0.33). The mIns/tNAA ratio in PMC is also associated with ΔFS (<i>p</i> = 0.02, <i>r</i> = 0.41). In the INC, tCho concentrations (<i>p</i> = 0.04, <i>r</i> = -0.54) and mIns/tNAA ratio (<i>p</i> = 0.02, <i>r</i> = -0.38) were negatively associated with ALSFRS-R and positively correlated with ΔFS (<i>p</i> = 0.01, <i>r</i> = 0.33) and (<i>p</i> = 0.001, <i>r</i> = 0.61), respectively. The study suggests that neurochemistry changes in ALS patients' brains are linked to selective neuronal vulnerability and the underlying pathophysiology of the disease.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2025-03-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143741715","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Gabriel Cathoud, Mohtadin Hashemi, Yuri Lyubchenko, Pedro Simões
{"title":"Uncovering Amyloid-β Interactions: Gray versus White Matter.","authors":"Gabriel Cathoud, Mohtadin Hashemi, Yuri Lyubchenko, Pedro Simões","doi":"10.1021/acschemneuro.4c00439","DOIUrl":"https://doi.org/10.1021/acschemneuro.4c00439","url":null,"abstract":"<p><p>Alzheimer's disease is characterized by the accumulation of amyloid plaques in the brain. Recent studies suggest that amyloid-β (Aβ) peptides interact with cell membranes, potentially catalyzing plaque formation. However, the effect of varying cell membrane compositions on this catalytic process requires further investigation. Using molecular dynamics simulations, we demonstrate that a model gray matter membrane significantly influences the secondary structure of β-amyloid peptides. Notably, residues Asp1 and Glu22 play crucial roles in the membrane interaction. Glutamic acid at position 22, located in the middle of the peptide chain, appears to promote the formation of β-hairpin conformations, which are critical for aggregation. Additionally, our simulations reveal that the model white matter membrane allows a spontaneous insertion of segments of the peptide into the membrane, suggesting that membrane interaction not only alters the peptide structure but may also compromise membrane integrity. Our results show that the different membrane compositions in the brain may play different roles when interacting with β-amyloid peptides.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2025-03-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143717572","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Induction of Ferroptotic Cell Death by Neuromelanin Pigments in Dopaminergic Cells.","authors":"Gizem Kaftan Öcal, Güliz Armagan","doi":"10.1021/acschemneuro.5c00029","DOIUrl":"https://doi.org/10.1021/acschemneuro.5c00029","url":null,"abstract":"<p><p>Neuromelanin (NM) is an iron-rich, insoluble brown or black pigment that exhibits protective properties. However, its accumulation over time may render it a source of free radicals. In Parkinson's disease, dopaminergic neurons with the highest NM levels and increased iron content are preferentially vulnerable to degeneration. Considering NM's iron binding capacity and the critical role of iron in ferroptosis, we aimed to investigate the interplay between neuromelanin and ferroptosis in dopaminergic cells. We prepared two NM pigments: iron-free NM (ifNM) and iron-containing NM (Fe<sup>3+</sup>NM) and, exposed to cells. After verifying NM accumulation, cell viability was assessed in the absence or presence of antioxidants (NAC (1 mM), Trolox (100 μM)) and specific inhibitors of cell death types. Ferroptosis-related parameters, including lipid peroxidation byproducts (4-HNE), lipid ROS, glutathione, intracellular iron, GPX4, and ACSL4, and cellular iron metabolism-related proteins (TfR1, ferroportin, ferritin, IREB2) were evaluated following ifNM and Fe<sup>3+</sup>NM treatments, with or without Ferrostatin-1, Liproxstatin-1 and deferoxamine. Both NMs induced cell death via distinct mechanisms. Ferroptotic cell death by ifNM and Fe<sup>3+</sup>NM was reversed by ferrostatin-1 and NAC (<i>p</i> < 0.05). Significant alterations in lipid peroxidation, GPX4 levels, and iron metabolism were observed independent of NM's iron composition (<i>p</i> < 0.05). Ferritin levels increased following ifNM treatment, reflecting an adaptive response to iron overload, while Fe<sup>3+</sup>NM treatment led to ferritin depletion, possibly via ferritinophagy. Our findings reveal a distinct role of iron-rich and iron-free neuromelanin in modulating ferroptotic pathways, highlighting the potential of targeting neuromelanin-iron interactions as a therapeutic strategy to mitigate neuronal ferroptosis in Parkinson's disease.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2025-03-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143717571","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Troglitazone as a Novel Nrf2 Activator to Attenuate Oxidative Stress and Exert Neuroprotection.","authors":"Linjie Zhang, Shuang Wang, Yanxia Zhang, Xiaopeng Zhang, Junmin Xi, Jun Wu, Jianguo Fang, Haiyu Zhao, Baoxin Zhang","doi":"10.1021/acschemneuro.5c00163","DOIUrl":"https://doi.org/10.1021/acschemneuro.5c00163","url":null,"abstract":"<p><p>Nuclear factor erythroid 2 related factor 2 (Nrf2) is closely associated with neurodegenerative diseases, and the Nrf2-mediated activation of antioxidant response elements (AREs) brings about validated strategies for treating neurodegenerative diseases. Here, we discovered that troglitazone, a clinical medication for diabetes mellitus, could serve as a Nrf2 activator to rescue neuronal damages both in vitro and in vivo. The mechanism of troglitazone action involves binding with kelch-like ECH-associated protein 1 (Keap1) and the activation of Nrf2. This process leads to the migration of Nrf2 to the cell nucleus and transactivates the AREs. Troglitazone exhibits significant alleviation of oxidative stress in PC12 cells induced by hydrogen peroxide or 6-hydroxydopamine (6-OHDA). In vivo studies indicate that troglitazone could rescue the motor activity and neurodevelopmental deficiency in zebrafish induced by 6-OHDA. Additionally, mass spectrometry imaging demonstrates that troglitazone could cross the zebrafish blood-brain barrier, supporting the application of troglitazone in treating neurodegenerative diseases. Overall, this work reveals that the novel Nrf2 activator troglitazone has potential therapeutic value for neurodegeneration and provides a foundation for its repurposing.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2025-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143707763","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ACS Chemical NeurosciencePub Date : 2025-03-25DOI: 10.1021/acschemneuro.5c0007510.1021/acschemneuro.5c00075
Emanuel F. Lopes, Alyssa M. West, Jason L. Locke, Katherine Holleran, Leighelle A. Adrian, Monica H. Dawes, Alyson M. Curry, Harlie A. McKelvey, Thomas Martin and Sara R. Jones*,
{"title":"Morphine-Induced Antinociception Is Potentiated and Dopamine Elevations Are Inhibited by the Biased Kappa Opioid Receptor Agonist Triazole 1.1","authors":"Emanuel F. Lopes, Alyssa M. West, Jason L. Locke, Katherine Holleran, Leighelle A. Adrian, Monica H. Dawes, Alyson M. Curry, Harlie A. McKelvey, Thomas Martin and Sara R. Jones*, ","doi":"10.1021/acschemneuro.5c0007510.1021/acschemneuro.5c00075","DOIUrl":"https://doi.org/10.1021/acschemneuro.5c00075https://doi.org/10.1021/acschemneuro.5c00075","url":null,"abstract":"<p >Traditional analgesic opioid compounds, which act through μ opioid receptors (MORs), engender a high risk for misuse and dependence. κ opioid receptor (KOR) activation, a potential target for pain treatment, produces antinociception without euphoric side effects but results in dysphoria and aversion. Triazole 1.1 is a KOR agonist biased toward G-protein coupled signaling, potentially promoting antinociception without dysphoria. We tested whether triazole 1.1 could provide antinociception and its effects in combination with morphine. We employed a lactic acid abdominal pain model, which induced acute pain behaviors, decreased basal dopamine levels in the nucleus accumbens (NAc), and increased KOR function. We administered several interventions including triazole 1.1 (30 mg/kg) and morphine (12 or 24 mg/kg), individually and in combination. Triazole 1.1 alone reduced the pain behavioral response and changes to KOR function but did not prevent the reduction in basal dopamine levels. Morphine not only dose-dependently prevented behavioral pain responses but also elevated NAc dopamine and did not prevent the pain-induced increase in KOR function. However, combining low-dose morphine with triazole 1.1 prevents behavioral pain responses, changes to NAc dopamine levels, and changes to KOR function. Therefore, we present triazole 1.1 as a dose-sparing pain treatment to be used in combination with a lower dose of morphine, thus reducing the potential for opioid misuse.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":"16 7","pages":"1377–1387 1377–1387"},"PeriodicalIF":4.1,"publicationDate":"2025-03-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143746006","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Takato Hiranita, Weimin C Hong, Abhisheak Sharma, Jessica P Lopez, Christophe Mesangeau, Daniel A Whittaker, Walid Alsharif, Theresa A Kopajtic, Seshulatha Jamalapuram, Bonnie A Avery, Gianluigi Tanda, Christopher R McCurdy, Jonathan L Katz
{"title":"Preclinical Profile of CM699 as a Medication Candidate for Stimulant Use Disorder.","authors":"Takato Hiranita, Weimin C Hong, Abhisheak Sharma, Jessica P Lopez, Christophe Mesangeau, Daniel A Whittaker, Walid Alsharif, Theresa A Kopajtic, Seshulatha Jamalapuram, Bonnie A Avery, Gianluigi Tanda, Christopher R McCurdy, Jonathan L Katz","doi":"10.1021/acschemneuro.4c00589","DOIUrl":"https://doi.org/10.1021/acschemneuro.4c00589","url":null,"abstract":"<p><p>There currently are no medications proven to be effective for the treatment of stimulant-use disorder (SUD). Sigma-receptor (σR) antagonists block many effects of stimulant drugs but not the reinforcing effects assessed with self-administration in rats. However, a recent study suggests that σR antagonism combined with a dopamine (DA) transporter (DAT) blockade selectively attenuates stimulant self-administration. A compound with potential for dual DAT/σR inhibition, CM699, was synthesized and had the necessary <i>ex vivo</i> affinities of 311 and 14.1 nM at DAT and σ<sub>1</sub>Rs, respectively. CM699 inhibited DA uptake <i>ex vivo</i>. Antagonist effects at σ<sub>1</sub>Rs by CM699 were confirmed with a recently reported pharmacological assay: CM699 increased, whereas the σ<sub>1</sub>R agonist, (+)-pentazocine, decreased σ<sub>1</sub>R multimers detected in nondenaturing protein gels, and CM699 blocked the effects of (+)-pentazocine. CM699 after intravenous administration (5.0 mg/kg) in rats had an elimination half-life of 4.4 h. In rats, CM699 after intraperitoneal administration blunted the stimulatory effects of cocaine on DA levels in the nucleus accumbens and insurmountably blocked cocaine self-administration, indicating efficacy as a cocaine antagonist <i>in vivo</i>. When given alone, CM699 was not self-administered nor had significant effects on nucleus accumbens DA, suggesting minimal, if any, abuse potential. Further, in a biochemical assay designed to probe the conformation of DAT, (+)-pentazocine potentiated cocaine-induced cysteine accessibility of DAT transmembrane domain 6a, suggesting a shift in the conformational equilibrium of DAT toward outward-facing, whereas CM699 blocked this effect. The results provide preclinical proof of concept for dual DAT/σR inhibition as a novel DAT-conformational approach for the development of medications to treat SUD.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2025-03-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143707760","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ACS Chemical NeurosciencePub Date : 2025-03-25DOI: 10.1021/acschemneuro.4c0057710.1021/acschemneuro.4c00577
Sonia Podvin, Jazmin Florio, Brian Spencer, Michael Mante, Estefani Guzman, Carlos Arias, Charles Mosier, Von V. Phan, Michael C. Yoon, Jehad Almaliti, Anthony J. O’Donoghue, William H. Gerwick, Robert A. Rissman and Vivian Hook*,
{"title":"Activation of Cytosolic Cathepsin B Activity in the Brain by Traumatic Brain Injury and Inhibition by the Neutral pH Selective Inhibitor Probe Z-Arg-Lys-AOMK","authors":"Sonia Podvin, Jazmin Florio, Brian Spencer, Michael Mante, Estefani Guzman, Carlos Arias, Charles Mosier, Von V. Phan, Michael C. Yoon, Jehad Almaliti, Anthony J. O’Donoghue, William H. Gerwick, Robert A. Rissman and Vivian Hook*, ","doi":"10.1021/acschemneuro.4c0057710.1021/acschemneuro.4c00577","DOIUrl":"https://doi.org/10.1021/acschemneuro.4c00577https://doi.org/10.1021/acschemneuro.4c00577","url":null,"abstract":"<p >Cathepsin B has been shown to contribute to deficits in traumatic brain injury (TBI), an important risk factor for Alzheimer’s disease (AD). Cathepsin B is elevated in TBI and AD patients, as well as in animal models of these conditions. Knockout of the cathepsin B gene results in amelioration of TBI-induced motor dysfunction and improvement of AD memory deficit in mice. The mechanism of cathepsin B pathogenesis in these brain disorders has been hypothesized to involve its translocation to the cytosol from its normal lysosomal location. This study, therefore, evaluated brain cytosolic cathepsin B activity in the controlled cortical impact (CCI) mouse model of TBI. CCI-TBI resulted in motor deficits demonstrated by the rotarod assay, brain tissue lesions, and disorganization of the hippocampus. Significantly, CCI-TBI increased cytosolic cathepsin B activity in the brain cortex in the ipsilateral brain hemisphere that received the CCI-TBI injury, with a concomitant decrease in the lysosomal fraction. Cathepsin B activity was monitored using the substrate Z-Nle-Lys-Arg-AMC which specifically detects cathepsin B activity but not other cysteine proteases. The normal lysosomal distribution of cathepsin B was observed by its discrete localization in brain cortical cells. CCI-TBI resulted in a more diffuse cellular distribution of cathepsin B consistent with translocation to the cytosol. Further studies utilized the novel neutral pH-selective inhibitor, Z-Arg-Lys-AOMK, that specifically inhibits cathepsin B at neutral pH 7.2 of the cytosol but not at acidic pH 4.6 of lysosomes. Daily administration of Z-Arg-Lys-AOMK (ip), beginning 1 day before CCI-TBI, resulted in the reduction of the increased cytosolic cathepsin B activity induced by CCI-TBI. The inhibitor also reduced cathepsin B activities in homogenates of the brain cortex and hippocampus which were increased by CCI-TBI. Furthermore, the Z-Arg-Lys-AOMK inhibitor resulted in the reduction of motor function deficit resulting from CCI-TBI. These findings demonstrate the activation of cytosolic cathepsin B activity in CCI-TBI mouse brain injury.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":"16 7","pages":"1297–1308 1297–1308"},"PeriodicalIF":4.1,"publicationDate":"2025-03-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/epdf/10.1021/acschemneuro.4c00577","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143746001","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Julie L Engers, Jinming Li, Changho Han, Madeline F Long, Alison R Gregro, Christopher C Presley, Jonathan W Dickerson, Weimin Peng, Hyekyung P Cho, Alice L Rodriguez, Zixiu Xiang, Olivier Boutaud, Colin O'Carroll, P Markus Dey, Ethan S Burstein, Colleen M Niswender, Jerri M Rook, P Jeffrey Conn, Darren W Engers, Craig W Lindsley
{"title":"Application of Deuterium in an M<sub>1</sub> Positive Allosteric Modulator Back-Up Program: The Discovery of VU6045422.","authors":"Julie L Engers, Jinming Li, Changho Han, Madeline F Long, Alison R Gregro, Christopher C Presley, Jonathan W Dickerson, Weimin Peng, Hyekyung P Cho, Alice L Rodriguez, Zixiu Xiang, Olivier Boutaud, Colin O'Carroll, P Markus Dey, Ethan S Burstein, Colleen M Niswender, Jerri M Rook, P Jeffrey Conn, Darren W Engers, Craig W Lindsley","doi":"10.1021/acschemneuro.5c00119","DOIUrl":"https://doi.org/10.1021/acschemneuro.5c00119","url":null,"abstract":"<p><p>Recently, we disclosed VU0467319, an M<sub>1</sub> positive allosteric modulator (PAM) clinical candidate that had successfully completed a phase I single ascending dose clinical trial. Pharmacokinetic assessment revealed that, in humans upon increasing dose, a circulating, inactive metabolite constituted a major portion of the total drug-related area under the curve (AUC). One approach the team employed to reduce inactive metabolite formation in the back-up program was the kinetic isotope effect, replacing the metabolically labile C-H bonds with shorter, more stable C-D bonds. The C-D dipole afforded VU6045422, a more potent M<sub>1</sub> PAM (human EC<sub>50</sub> = 192 nM, 80% ACh Max) than its proteocongener VU0467319 (human EC<sub>50</sub> = 492 nM, 71% ACh Max), and retained the desired profile of minimal M<sub>1</sub> agonism. Overall, the profile of VU6045422 supported advancement, as did greater <i>in vitro</i> metabolic stability in both microsomes and hepatocytes than did VU0467319. In both rat and dog <i>in vivo</i>, low doses proved to mirror the <i>in vitro</i> profile; however, at higher doses in 14-day exploratory toxicology studies, the amount of the same undesired metabolite derived from VU6045422 was equivalent to that produced from VU0467319. This unexpected IVIVC result, coupled with less than dose-proportional increases in exposure and no improvement in solubility, led to discontinuation of VU0467319/VU6045422 development.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2025-03-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143707757","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ACS Chemical NeurosciencePub Date : 2025-03-24DOI: 10.1021/acschemneuro.5c0011210.1021/acschemneuro.5c00112
Ritu Soni, Vaishali Pankaj, Sudeep Roy, Amit Khairnar and Jigna Shah*,
{"title":"Upregulation of the PI3K/AKT and Nrf2 Pathways by the DPP-4 Inhibitor Sitagliptin Renders Neuroprotection in Chemically Induced Parkinson’s Disease Mouse Models","authors":"Ritu Soni, Vaishali Pankaj, Sudeep Roy, Amit Khairnar and Jigna Shah*, ","doi":"10.1021/acschemneuro.5c0011210.1021/acschemneuro.5c00112","DOIUrl":"https://doi.org/10.1021/acschemneuro.5c00112https://doi.org/10.1021/acschemneuro.5c00112","url":null,"abstract":"<p >Parkinson’s disease (PD) is one of the most common progressive neurodegenerative pathologies that leads to dopaminergic deficiency and motor manifestations. Alpha-synuclein aggregation is a characteristic hallmark of PD pathogenesis. These aggregates facilitate the formation of Lewy bodies and degeneration. The epidemiological evidence demonstrates a definitive association of diabetes with PD risk. Considering this, many antidiabetic agents such as GLP-1 agonists and DPP-4 inhibitors are being explored as alternative PD therapeutics. This study evaluated the neuroprotective effect of the DPP-4 inhibitor sitagliptin mediated by the PI3K/AKT and Nrf2 pathways in PD models. <i>In silico</i> studies were conducted to determine the binding affinity, stability, and ADMET properties of DPP-4 inhibitors with target proteins. Sitagliptin (15 mg/kg p.o.) was administered in rotenone (30 mg/kg p.o. for 28 days)-induced and MPTP/P (25 mg/kg i.p. MPTP and 100 mg/kg probenecid i.p. twice a week for 5 weeks)-induced PD mouse (C57/BL6) models. Neurobehavioral assessments were carried out throughout the study. Biochemical (GSH, MDA), molecular estimations (AKT, Nrf2, PI3K, GSK-3β, GLP1, CREB, BDNF, NF-κB, alpha-synuclein), histopathological studies, and immunohistochemistry were carried out at the end of the study. The <i>in silico</i> studies demonstrate better binding, stability, and ADMET profile of sitagliptin with both target proteins. Sitagliptin restored cognitive and motor deficits in both rotenone- and MPTP/P-induced mouse models. There was upregulation of PI3K, AKT, Nrf2, CREB, and BDNF levels and downregulation of GSK-3β, NF-κB, and alpha-synuclein levels in both models after treatment with sitagliptin. However, GLP1 levels were not significantly restored, indicating a GLP1-independent mechanism. It also restored histopathological alterations and TH+ neuronal loss induced by rotenone and MPTP/P. These findings demonstrate that sitagliptin exhibits neuroprotective action mediated by upregulation of the PI3K/AKT and Nrf2 pathways in rotenone and MPTP/P mouse models of PD.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":"16 7","pages":"1402–1417 1402–1417"},"PeriodicalIF":4.1,"publicationDate":"2025-03-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143746032","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}