1
|
Lillethorup IA, Hemmingsen AV, Qvortrup K. Prodrugs and their activation mechanisms for brain drug delivery. RSC Med Chem 2025:d4md00788c. [PMID: 39829971 PMCID: PMC11740913 DOI: 10.1039/d4md00788c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/15/2024] [Indexed: 01/22/2025] Open
Abstract
Prodrugs are masked drugs that first become pharmacologically active after undergoing a structural change in vivo. They are designed to improve physicochemical/biopharmaceutical drug properties and increase site specificity. The prodrug approach is important when developing brain-targeting drugs due to the presence of the brain barriers that seriously limit the brain entry of highly polar, multifunctional drug entities. While several excellent reviews summarize the structural modifications facilitating transport across the brain barriers, a summary of mechanisms used for the activation of the prodrug in the brain is missing. Given the high need for innovative discoveries in brain drug development, we here review the most important tools being developed since 2000 for CNS prodrug activation.
Collapse
Affiliation(s)
| | | | - Katrine Qvortrup
- Department of Chemistry, Technical University of Denmark 2800 Kongens Lyngby Denmark
| |
Collapse
|
2
|
Zheng H, Wu H, Wang D, Wang S, Ji D, Liu X, Gao G, Su X, Zhang Y, Ling Y. Research progress of prodrugs for the treatment of cerebral ischemia. Eur J Med Chem 2024; 272:116457. [PMID: 38704941 DOI: 10.1016/j.ejmech.2024.116457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/20/2024] [Accepted: 04/25/2024] [Indexed: 05/07/2024]
Abstract
It is well-known that pharmacotherapy plays a pivotal role in the treatment and prevention of cerebral ischemia. Nevertheless, existing drugs, including numerous natural products, encounter various challenges when applied in cerebral ischemia treatment. These challenges comprise poor brain absorption due to low blood-brain barrier (BBB) permeability, limited water solubility, inadequate bioavailability, poor stability, and rapid metabolism. To address these issues, researchers have turned to prodrug strategies, aiming to mitigate or eliminate the adverse properties of parent drug molecules. In vivo metabolism or enzymatic reactions convert prodrugs into active parent drugs, thereby augmenting BBB permeability, improving bioavailability and stability, and reducing toxicity to normal tissues, ultimately aiming to enhance treatment efficacy and safety. This comprehensive review delves into multiple effective prodrug strategies, providing a detailed description of representative prodrugs developed over the past two decades. It underscores the potential of prodrug approaches to improve the therapeutic outcomes of currently available drugs for cerebral ischemia. The publication of this review serves to enrich current research progress on prodrug strategies for the treatment and prevention of cerebral ischemia. Furthermore, it seeks to offer valuable insights for pharmaceutical chemists in this field, offer guidance for the development of drugs for cerebral ischemia, and provide patients with safer and more effective drug treatment options.
Collapse
Affiliation(s)
- Hongwei Zheng
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Hongmei Wu
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China; Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Dezhi Wang
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China; Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Sijia Wang
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China; Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Dongliang Ji
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China; Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Xiao Liu
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Ge Gao
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Xing Su
- Department of Neurosurgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China.
| | - Yanan Zhang
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China.
| | - Yong Ling
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001, Nantong, Jiangsu, PR China.
| |
Collapse
|
3
|
Ding Y, Jin Y, Peng T, Gao Y, Zang Y, He H, Li F, Zhang Y, Zhang H, Chen L. Fabrication of multifunctional metal-organic frameworks nanoparticles via layer-by-layer self-assembly to efficiently discover PSD95-nNOS uncouplers for stroke treatment. J Nanobiotechnology 2022; 20:379. [PMID: 35964123 PMCID: PMC9375364 DOI: 10.1186/s12951-022-01583-7] [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: 05/17/2022] [Accepted: 07/31/2022] [Indexed: 11/24/2022] Open
Abstract
Background Disruption of the postsynaptic density protein-95 (PSD95)—neuronal nitric oxide synthase (nNOS) coupling is an effective way to treat ischemic stroke, however, it still faces some challenges, especially lack of satisfactory PSD95-nNOS uncouplers and the efficient high throughput screening model to discover them. Results Herein, the multifunctional metal–organic framework (MMOF) nanoparticles as a new screening system were innovatively fabricated via layer-by-layer self-assembly in which His-tagged nNOS was selectively immobilized on the surface of magnetic MOF, and then PSD95 with green fluorescent protein (GFP-PSD95) was specifically bound on it. It was found that MMOF nanoparticles not only exhibited the superior performances including the high loading efficiency, reusability, and anti-interference ability, but also possessed the good fluorescent sensitivity to detect the coupled GFP-PSD95. After MMOF nanoparticles interacted with the uncouplers, they would be rapidly separated from uncoupled GFP-PSD95 by magnet, and the fluorescent intensities could be determined to assay the uncoupling efficiency at high throughput level. Conclusions In conclusion, MMOF nanoparticles were successfully fabricated and applied to screen the natural actives as potential PSD95-nNOS uncouplers. Taken together, our newly developed method provided a new material as a platform for efficiently discovering PSD95-nNOS uncouplers for stoke treatment. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01583-7.
Collapse
Affiliation(s)
- Yingying Ding
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Yang Jin
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Tao Peng
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Yankun Gao
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Yang Zang
- College of Economics and Management, Anhui Agricultural University, Hefei, Anhui, 230036, People's Republic of China
| | - Hongliang He
- Department of Pharmacy, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Fei Li
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Yu Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, People's Republic of China.
| | - Hongjuan Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, People's Republic of China.
| | - Lina Chen
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, People's Republic of China.
| |
Collapse
|
4
|
Wei W, Liu W, Du S, Govindarajalu G, Irungu A, Bekker A, Tao YX. A Compound Mitigates Cancer Pain and Chemotherapy-Induced Neuropathic Pain by Dually Targeting nNOS-PSD-95 Interaction and GABA A Receptor. Neurotherapeutics 2021; 18:2436-2448. [PMID: 34796458 PMCID: PMC8804143 DOI: 10.1007/s13311-021-01158-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2021] [Indexed: 10/19/2022] Open
Abstract
Metastatic bone pain and chemotherapy-induced peripheral neuropathic pain are the most common clinical symptoms in cancer patients. The current clinical management of these two disorders is ineffective and/or produces severe side effects. The present study employed a dual-target compound named as ZL006-05 and examined the effect of systemic administration of ZL006-05 on RM-1-induced bone cancer pain and paclitaxel-induced neuropathic pain. Intravenous injection of ZL006-05 dose-dependently alleviated RM-1-induced mechanical allodynia, heat hyperalgesia, cold hyperalgesia, and spontaneously ongoing nociceptive responses during both induction and maintenance periods, without analgesic tolerance, affecting basal/acute pain and locomotor function. Similar behavioral results were observed in paclitaxel-induced neuropathic pain. This injection also decreased neuronal and astrocyte hyperactivities in the lumbar dorsal horn after RM-1 tibial inoculation or paclitaxel intraperitoneal injection. Mechanistically, intravenous injection of ZL006-05 potentiated the GABAA receptor agonist-evoked currents in the neurons of the dorsal horn and anterior cingulate cortex and also blocked the paclitaxel-induced increase in postsynaptic density-95-neuronal nitric oxide synthase interaction in dorsal horn. Our findings strongly suggest that ZL006-05 may be a new candidate for the management of cancer pain and chemotherapy-induced peripheral neuropathic pain.
Collapse
Affiliation(s)
- Wei Wei
- Department of Anesthesiology, Rutgers New Jersey Medical School, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA
| | - Weili Liu
- Department of Anesthesiology, Rutgers New Jersey Medical School, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA
| | - Shibin Du
- Department of Anesthesiology, Rutgers New Jersey Medical School, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA
| | - Gokulapriya Govindarajalu
- Department of Anesthesiology, Rutgers New Jersey Medical School, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA
| | - Antony Irungu
- Department of Anesthesiology, Rutgers New Jersey Medical School, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA
| | - Alex Bekker
- Department of Anesthesiology, Rutgers New Jersey Medical School, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA
| | - Yuan-Xiang Tao
- Department of Anesthesiology, Rutgers New Jersey Medical School, The State University of New Jersey, 185 S. Orange Ave., MSB, F-661, Newark, NJ, 07103, USA.
- Department of Physiology, Rutgers New Jersey Medical School, The State University of New Jersey, Pharmacology & Neuroscience, Newark, NJ, 07103, USA.
- Department of Cell Biology & Molecular Medicine, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, 07103, USA.
| |
Collapse
|
5
|
Qin Y, Feng L, Fan X, Zheng L, Zhang Y, Chang L, Li T. Neuroprotective Effect of N-Cyclohexylethyl-[A/G]-[D/E]-X-V Peptides on Ischemic Stroke by Blocking nNOS-CAPON Interaction. ACS Chem Neurosci 2021; 12:244-255. [PMID: 33356131 DOI: 10.1021/acschemneuro.0c00739] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The protein-protein interaction between neuronal nitric oxide syntheses (nNOS) and the carboxy-terminal PDZ ligand of nNOS (CAPON) is a potential target for the treatment of ischemic stroke. Our previous study had identified ZLc-002 as a promising lead compound for inhibiting nNOS-CAPON coupling. To find better neuroprotective agents disrupting the ischemia-induced nNOS-CAPON interaction, a series of N-cyclohexylethyl-[A/G]-[D/E]-X-V peptides based on the carboxy-terminal tetrapeptide of CAPON was designed, synthesized, and evaluated in this study. Herein, we reported an affinity-based fluorescence polarization (FP) method using 5-carboxyfluorescein (5-FAM) labeled CAPON (496-506) peptide as the probe for high-throughput screening of the small-molecule inhibitors of the PDZ domain of nNOS. N-Cyclohexylethyl-ADAV displayed the most potent affinity for the nNOS PDZ domain in the FP and isothermal titration calorimetry (ITC) (ΔH = -1670 ± 151.0 cal/mol) assays. To improve bioavailability, lipophilicity, and membrane permeability, the Asp methylation was employed to get N-cyclohexylethyl-AD(OMe)AV, which possesses good blood-brain barrier (BBB) permeability in vitro parallel artificial membrane permeability assay (PAMPA)-BBB (Pe = 6.07 cm/s) and in vivo assays. In addition, N-cyclohexylethyl-AD(OMe)AV (10 mg/kg body weight, i.v., immediately after reperfusion) substantially reduced infarct size in rats, which was measured 24 h after reperfusion and subjected to 120 min of middle cerebral artery occlusion (MCAO).
Collapse
Affiliation(s)
- Yajuan Qin
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Lingling Feng
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xin Fan
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Liping Zheng
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yu Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Lei Chang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Tingyou Li
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
6
|
Gu Y, Zhu D. nNOS-mediated protein-protein interactions: promising targets for treating neurological and neuropsychiatric disorders. J Biomed Res 2020; 35:1-10. [PMID: 33402546 PMCID: PMC7874267 DOI: 10.7555/jbr.34.20200108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neurological and neuropsychiatric disorders are one of the leading causes of disability worldwide and affect the health of billions of people. Nitric oxide (NO), a free gas with multitudinous bioactivities, is mainly produced from the oxidation of L-arginine by neuronal nitric oxide synthase (nNOS) in the brain. Inhibiting nNOS benefits a variety of neurological and neuropsychiatric disorders, including stroke, depression and anxiety disorders, post-traumatic stress disorder, Parkinson's disease, Alzheimer's disease, chronic pain, and drug addiction. Due to critical roles of nNOS in learning and memory and synaptic plasticity, direct inhibition of nNOS may cause severe side effects. Importantly, interactions of several proteins, including post-synaptic density 95 (PSD-95), carboxy-terminal PDZ ligand of nNOS (CAPON) and serotonin transporter (SERT), with the PSD/Disc-large/ZO-1 homologous (PDZ) domain of nNOS have been demonstrated to influence the subcellular distribution and activity of the enzyme in the brain. Therefore, it will be a preferable means to interfere with nNOS-mediated protein-protein interactions (PPIs), which do not lead to undesirable effects. Herein, we summarize the current literatures on nNOS-mediated PPIs involved in neurological and neuropsychiatric disorders, and the discovery of drugs targeting the PPIs, which is expected to provide potential targets for developing novel drugs and new strategy for the treatment of neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yuanyuan Gu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Dongya Zhu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Institution of Stem Cell and Neuroregeneration, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
7
|
Pan L, Ding Y, Ni X, Wang CZ, Jiang B, Zhang Y, Jiang N, Tang Y, Chen L, Yuan CS. Modeling rapid and selective capture of nNOS–PSD-95 uncouplers from Sanhuang Xiexin decoction by novel molecularly imprinted polymers based on metal–organic frameworks. RSC Adv 2020; 10:7671-7681. [PMID: 35492204 PMCID: PMC9049783 DOI: 10.1039/c9ra10537a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/05/2020] [Indexed: 12/12/2022] Open
Abstract
Novel and highly selective molecularly imprinted polymers based on the surface of metal–organic frameworks, NH2-MIL-101(Cr) (MIL@MIPS), were successfully fabricated to capture neuronal nitric oxide synthase–postsynaptic density protein-95 (nNOS–PSD-95) uncouplers from Sanhuang Xiexin Decoction (SXD) for stroke treatment. The resultant polymers were characterized by Fourier transform infrared spectroscopy, scanning electron microscopy, thermogravimetric analysis, and X-ray diffraction. The performance tests revealed that MIL@MIPs had a large binding capacity, fast kinetics, and excellent selectivity. Then the obtained polymers were satisfactorily applied to solid-phase extraction coupled with high-performance liquid chromatography to selectively capture nNOS–PSD-95 uncouplers from SXD. Furthermore, the biological activities of components obtained from SXD were evaluated in vivo and in vitro. As a consequence, the components showed a potent neuroprotective effect from the MTS assay and uncoupling activity from the co-immunoprecipitation experiment. In addition, the anti-ischemic stroke assay in vivo was further investigated to determine the effect of reducing infarct size and ameliorating neurological deficit by the active components. Therefore, this present study contributes a valuable new method and new tendency to selectively capture active components for stroke treatment from SXD and other natural medicines. Novel MIL@MIPs were prepared to rapidly capture nNOS–PSD-95 uncouplers from Sanhuang Xiexin decoction, coupled with SPE and HPLC.![]()
Collapse
Affiliation(s)
- Linli Pan
- School of Pharmacy
- Nanjing Medical University
- Nanjing
- China
| | - Yingying Ding
- School of Pharmacy
- Nanjing Medical University
- Nanjing
- China
| | - Xiaoting Ni
- School of Pharmacy
- Nanjing Medical University
- Nanjing
- China
| | - Chong-Zhi Wang
- Tang Center for Herbal Medicine Research
- Department of Anesthesia & Critical Care
- University of Chicago
- Chicago
- USA
| | - Bo Jiang
- School of Pharmacy
- Nanjing Medical University
- Nanjing
- China
| | - Yu Zhang
- School of Pharmacy
- Nanjing Medical University
- Nanjing
- China
| | - Nan Jiang
- School of Pharmacy
- Nanjing Medical University
- Nanjing
- China
| | - Yulin Tang
- School of Pharmacy
- Nanjing Medical University
- Nanjing
- China
| | - Lina Chen
- School of Pharmacy
- Nanjing Medical University
- Nanjing
- China
| | - Chun-Su Yuan
- Tang Center for Herbal Medicine Research
- Department of Anesthesia & Critical Care
- University of Chicago
- Chicago
- USA
| |
Collapse
|
8
|
Translating preclinical findings in clinically relevant new antipsychotic targets: focus on the glutamatergic postsynaptic density. Implications for treatment resistant schizophrenia. Neurosci Biobehav Rev 2019; 107:795-827. [DOI: 10.1016/j.neubiorev.2019.08.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 07/20/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023]
|
9
|
Yao D, Zhang L, Huang J, Sun C, Zhang Y, Gu X, Wang CZ, Li F, Chen L, Yuan CS. A surface magnetic imprinted polymers as artificial receptors for selective and efficient capturing of new neuronal nitric oxide synthase-post synaptic density protein-95 uncouplers. J Pharm Biomed Anal 2018; 154:180-190. [PMID: 29550707 DOI: 10.1016/j.jpba.2018.03.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 03/01/2018] [Accepted: 03/02/2018] [Indexed: 02/06/2023]
Abstract
In this work, surface magnetic molecularly imprinted polymers (SMMIPs) were synthesized and used as artificial receptors in the dispersive magnetic solid phase extraction (DMSPE) for capturing potential neuronal nitric oxide synthase-post synaptic density protein-95 (nNOS-PSD-95) uncouplers, which is known as neuroprotection against stroke. Factors that affected selective separation and adsorption of the artificial receptors, such as the amount of template, the types of functional monomer and porogen solvents, and the molar ratio of template/functional monomer/cross-linker were optimized. The artificial receptors were also characterized using fourier transformed infrared, scanning electron microscope, thermal gravimetric analysis and physical property measurement systems. Multiple interactions between template and SMMIPs led to larger binding capacities, faster binding kinetics, quicker separation abilities and more efficient selectivity than the surface magnetic nonimprinted polymers (SMNIPs). The SMMIPs were successfully applied to capture potential nNOS-PSD-95 uncouplers from complex samples, and eight compounds were seized and confirmed rapidly when combined with HPLC and MS. The detection of the new nNOS-PSD-95 uncouplers ranged from 0.001 to 1.500 mg/mL with correlation coefficients of 0.9990-0.9995. The LOD and LOQ were 0.10-0.68 μg/mL and 0.47-2.11 μg/mL, respectively. The neuroprotective effect and co-immunoprecipitation test in vitro revealed that Emodin-1-O-β-d-glucoside, Rhaponticin, Gnetol and 2,3,5,4'-Tetrahydroxystilbene-2-O-β-d-glucoside have neuroprotective and uncoupling activities, and that they may be the new uncouplers of nNOS-PSD-95.
Collapse
Affiliation(s)
- Dandan Yao
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Lei Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jiaojiao Huang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Chenghong Sun
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yu Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiaoli Gu
- Department of Pharmacy, The Second Affiliated Hospital of Nantong University, China
| | - Chong-Zhi Wang
- Tang Center for Herbal Medicine Research, and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA
| | - Fei Li
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Lina Chen
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| | - Chun-Su Yuan
- Tang Center for Herbal Medicine Research, and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
10
|
Huang J, Sun C, Yao D, Wang CZ, Zhang L, Zhang Y, Chen L, Yuan CS. Novel surface imprinted magnetic mesoporous silica as artificial antibodies for efficient discovery and capture of candidate nNOS–PSD-95 uncouplers for stroke treatment. J Mater Chem B 2018; 6:1531-1542. [DOI: 10.1039/c7tb03044d] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Magnetic mesoporous silica imprinted materials as artificial antibodies for the discovery and capture of candidate nNOS–PSD-95 uncouplers for stroke treatment.
Collapse
Affiliation(s)
- Jiaojiao Huang
- School of Pharmacy, Nanjing Medical University
- Nanjing 211166
- China
| | - Chenghong Sun
- School of Pharmacy, Nanjing Medical University
- Nanjing 211166
- China
| | - Dandan Yao
- School of Pharmacy, Nanjing Medical University
- Nanjing 211166
- China
| | - Chong-Zhi Wang
- Tang Center for Herbal Medicine Research, and Department of Anesthesia & Critical Care, University of Chicago
- Chicago
- USA
| | - Lei Zhang
- School of Pharmacy, Nanjing Medical University
- Nanjing 211166
- China
| | - Yu Zhang
- School of Pharmacy, Nanjing Medical University
- Nanjing 211166
- China
| | - Lina Chen
- School of Pharmacy, Nanjing Medical University
- Nanjing 211166
- China
| | - Chun-Su Yuan
- Tang Center for Herbal Medicine Research, and Department of Anesthesia & Critical Care, University of Chicago
- Chicago
- USA
| |
Collapse
|
11
|
Tillmann S, Pereira VS, Liebenberg N, Christensen AK, Wegener G. ZL006, a small molecule inhibitor of PSD-95/nNOS interaction, does not induce antidepressant-like effects in two genetically predisposed rat models of depression and control animals. PLoS One 2017; 12:e0182698. [PMID: 28771575 PMCID: PMC5542618 DOI: 10.1371/journal.pone.0182698] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/21/2017] [Indexed: 11/26/2022] Open
Abstract
N-methyl-D-aspartate receptor (NMDA-R) antagonists and nitric oxide inhibitors have shown promising efficacy in depression but commonly induce adverse events. To circumvent these, a more indirect disruption of the nitric oxide synthase/postsynaptic density protein 95 kDa complex at the NMDA-R has been proposed. This disruption can be achieved using small molecule inhibitors such as ZL006, which has attracted attention as ischemic stroke therapy in rodents and has been proposed as a potential novel treatment for depression. Based on this, our aim was to translate these findings to animal models of depression to elucidate antidepressant-like properties in more detail. In the present study, we administered ZL006 to two established animal models of depression and control rodents. Following treatment, we measured locomotion in the Open Field and depressive-like behavior in the Forced Swim Test and Tail Suspension Test. Our experimental designs included the use of different species (rats, mice), strains (Flinders Sensitive Line rats, Flinders Resistant Line rats, Wistar Kyoto rats, Wistar Hanover rats, Sprague Dawley rats, B6NTac mice), routes of administration (intraperitoneal, intracerebroventricular), times of administration (single injection, repeated injections), treatment regimens (acute, sustained), and doses (5, 10, 15, 50 mg/kg). ZL006 did not affect behavior in any of the described settings. On a molecular level, ZL006 significantly reduced total nitrate/nitrite concentrations in the cerebellum, supporting that it is capable of reducing nitric oxide metabolites in the brain. Future studies using different experimental parameters are needed to further investigate the behavioral profile of ZL006.
Collapse
Affiliation(s)
- Sandra Tillmann
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
- * E-mail:
| | - Vitor Silva Pereira
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| | - Nico Liebenberg
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| | - Anne Karina Christensen
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| | - Gregers Wegener
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| |
Collapse
|
12
|
Liu SG, Wang YM, Zhang YJ, He XJ, Ma T, Song W, Zhang YM. ZL006 protects spinal cord neurons against ischemia-induced oxidative stress through AMPK-PGC-1α-Sirt3 pathway. Neurochem Int 2017; 108:230-237. [PMID: 28411102 DOI: 10.1016/j.neuint.2017.04.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/03/2017] [Accepted: 04/10/2017] [Indexed: 11/16/2022]
Abstract
Spinal cord ischemia (SCI) induces a range of cellular and molecular cascades, including activation of glutamate receptors and downstream signaling. Post-synaptic density protein 95 (PSD-95) links neuronal nitric oxide synthase (nNOS) with the N-methyl-d-aspartic acid (NMDA) receptors to form a ternary complex in the CNS. This molecular complex-mediated cytotoxicity has been implicated in brain ischemia, but its role in SCI has not been determined. The goal of the study was to investigate the potential protective effects of ZL006, a small-molecule inhibitor of the PSD-95/nNOS interaction, in an in vitro SCI model induced by oxygen and glucose deprivation (OGD) in cultured spinal cord neurons. We found that ZL006 reduced OGD-induced lactate dehydrogenase (LDH) release, neuronal apoptosis and loss of cell viability. This protection was accompanied by the preservation of mitochondrial function, as evidenced by reduced mitochondrial oxidative stress, attenuated mitochondrial membrane potential (MMP) loss, and enhanced ATP generation. In addition, ZL006 stimulated mitochondrial enzyme activities and SOD2 deacetylation in a Sirt3-dependent manner. The results of western blot analysis showed that ZL006 increased the activation of AMPK-PGC-1α-Sirt3 pathway, and the beneficial effects of ZL006 was partially abolished by AMPK inhibitor and PGC-1α knockdown. Therefore, our present data showed that, by the AMPK-PGC-1α-Sirt3 pathway, ZL006 protects spinal cord neurons against ischemia through reducing mitochondrial oxidative stress to prevent apoptosis.
Collapse
Affiliation(s)
- Shu-Guang Liu
- Department of Joint Surgery, Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province, 710054, China
| | - Yun-Mei Wang
- Department of Medical Oncology, Shaanxi Provincial Cancer Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province, 710061, China
| | - Yan-Jun Zhang
- Department of Medical Oncology, Shaanxi Provincial Cancer Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province, 710061, China
| | - Xi-Jing He
- Department of Orthopaedics, The Second Affiliated Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province, 710004, China
| | - Tao Ma
- Department of Joint Surgery, Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province, 710054, China
| | - Wei Song
- Department of Joint Surgery, Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province, 710054, China
| | - Yu-Min Zhang
- Department of Joint Surgery, Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province, 710054, China.
| |
Collapse
|
13
|
Ramakrishna K, Sivasankar C. Iridium catalyzed acceptor/acceptor carbene insertion into N–H bonds in water. Org Biomol Chem 2017; 15:2392-2396. [DOI: 10.1039/c7ob00177k] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Carbenes from highly stable acceptor/acceptor diazo compounds can be inserted into the N–H bonds of aromatic amines using an Ir(i) catalyst in an aqueous medium.
Collapse
Affiliation(s)
- Kankanala Ramakrishna
- Catalysis and Energy Laboratory
- Department of Chemistry
- Pondicherry University (A Central University)
- Puducherry 605014
- India
| | - Chinnappan Sivasankar
- Catalysis and Energy Laboratory
- Department of Chemistry
- Pondicherry University (A Central University)
- Puducherry 605014
- India
| |
Collapse
|