1
|
Pewklang T, Thompson T, Sefiani A, Geoffroy CG, Kamkaew A, Burgess K. Selective, Intrinsically Fluorescent Trk Modulating Probes. ACS Chem Neurosci 2024; 15. [PMID: 39356215 PMCID: PMC11487604 DOI: 10.1021/acschemneuro.4c00290] [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: 05/10/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 10/03/2024] Open
Abstract
Neurotrophins (NTs) elicit the growth, survival, and differentiation of neurons and other neuroectoderm tissues via activation of Trk receptors. Hot spots for NT·Trk interactions involve three neurotrophin loops. Mimicry of these using "cyclo-organopeptides" comprising loop sequences cyclized onto endocyclic organic fragments accounts for a few of the low molecular mass Trk agonists or modulators reported so far; the majority are nonpeptidic small molecules accessed without molecular design and identified in random screens. It has proven difficult to verify activities induced by low molecular mass substances are due to Trk activation (rather than via other receptors), enhanced Trk expression, enhanced NT expression, or other pathways. Consequently, identification of selective probes for the various Trk receptors (e.g., A, B, and C) has been very challenging. Further, a key feature of probes for early stage assays is that they should be easily detectable, and none of the compounds reported to date are. In this work, we designed novel cyclo-organopeptide derivatives where the organic fragment is a BODIPY fluor and found ones that selectively, though not specifically, activate TrkA, B, or C. One of the assays used to reach this conclusion (binding to live Trk-expressing cells) relied on intrinsic fluorescence in the tested materials. Consequently, this work established low molecular mass Trk-selective probes exhibiting neuroprotective effects.
Collapse
Affiliation(s)
- Thitima Pewklang
- Department
of Chemistry, Texas A & M University, Box 30012, College Station, Texas 77842-3012, United States
- School
of Chemistry, Institute of Science, Suranaree
University of Technology, Nakhon
Ratchasima 30000, Thailand
| | - Tye Thompson
- Department
of Chemistry, Texas A & M University, Box 30012, College Station, Texas 77842-3012, United States
| | - Arthur Sefiani
- Department
of Neuroscience and Experimental Therapeutics, Texas A & M University Health Science Center, Bryan, Texas 77807, United States
- NeuroCreis,
Inc., College Station, Texas 77840, United States
| | - Cédric G. Geoffroy
- Department
of Neuroscience and Experimental Therapeutics, Texas A & M University Health Science Center, Bryan, Texas 77807, United States
- NeuroCreis,
Inc., College Station, Texas 77840, United States
| | - Anyanee Kamkaew
- School
of Chemistry, Institute of Science, Suranaree
University of Technology, Nakhon
Ratchasima 30000, Thailand
| | - Kevin Burgess
- Department
of Chemistry, Texas A & M University, Box 30012, College Station, Texas 77842-3012, United States
| |
Collapse
|
2
|
Kalabekova R, Quinn CM, Movellan KT, Gronenborn AM, Akke M, Polenova T. 19F Fast Magic-Angle Spinning NMR Spectroscopy on Microcrystalline Complexes of Fluorinated Ligands and the Carbohydrate Recognition Domain of Galectin-3. Biochemistry 2024; 63:2207-2216. [PMID: 39008798 DOI: 10.1021/acs.biochem.4c00232] [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: 07/17/2024]
Abstract
Structural characterization of protein-ligand binding interfaces at atomic resolution is essential for improving the design of specific and potent inhibitors. Herein, we explored fast 19F- and 1H-detected magic angle spinning NMR spectroscopy to investigate the interaction between two fluorinated ligand diastereomers with the microcrystalline galectin-3 carbohydrate recognition domain. The detailed environment around the fluorine atoms was mapped by 2D 13C-19F and 1H-19F dipolar correlation experiments and permitted characterization of the binding interface. Our results demonstrate that 19F MAS NMR is a powerful tool for detailed characterization of protein-ligand interfaces and protein interactions at the atomic level.
Collapse
Affiliation(s)
- Roza Kalabekova
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - Caitlin M Quinn
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - Kumar Tekwani Movellan
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - Angela M Gronenborn
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
- Department of Structural Biology, University of Pittsburgh School of Medicine, 3501 Fifth Ave., Pittsburgh, Pennsylvania 15261, United States
| | - Mikael Akke
- Division of Biophysical Chemistry, Center for Molecular Protein Science, Department of Chemistry, Lund University, P.O. Box 124, Lund SE-22100, Sweden
| | - Tatyana Polenova
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
- Department of Structural Biology, University of Pittsburgh School of Medicine, 3501 Fifth Ave., Pittsburgh, Pennsylvania 15261, United States
| |
Collapse
|
3
|
Madhubala D, Mahato R, Khan MR, Bala A, Mukherjee AK. Neurotrophin peptidomimetics for the treatment of neurodegenerative diseases. Drug Discov Today 2024; 29:104156. [PMID: 39233307 DOI: 10.1016/j.drudis.2024.104156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/21/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
Neurotrophins, such as nerve growth factor and brain-derived neurotrophic factor, play an essential role in the survival of neurons. However, incorporating better features can increase their therapeutic efficacy in neurodegenerative diseases (NDs). Peptidomimetics, which mimic these neurotrophins, show potential for treating NDs. This study emphasizes the use of peptidomimetics from neurotrophins for treating NDs and their benefits. By improving bioavailability and stability, these molecules can completely transform the therapy for NDs. This in-depth review guides researchers and pharmaceutical developers, providing insight into the changing field of neurodegenerative medicine.
Collapse
Affiliation(s)
- Dev Madhubala
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India; Division of Life Sciences, Institute of Advanced Study in Science and Technology, Vigyan Path, Paschim Boragaon, Garchuk, Guwahati 781035, Assam, India
| | - Rosy Mahato
- Division of Life Sciences, Institute of Advanced Study in Science and Technology, Vigyan Path, Paschim Boragaon, Garchuk, Guwahati 781035, Assam, India; Faculty of Science, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Mojibur R Khan
- Division of Life Sciences, Institute of Advanced Study in Science and Technology, Vigyan Path, Paschim Boragaon, Garchuk, Guwahati 781035, Assam, India; Faculty of Science, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Asis Bala
- Division of Life Sciences, Institute of Advanced Study in Science and Technology, Vigyan Path, Paschim Boragaon, Garchuk, Guwahati 781035, Assam, India; Faculty of Science, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Ashis K Mukherjee
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India; Division of Life Sciences, Institute of Advanced Study in Science and Technology, Vigyan Path, Paschim Boragaon, Garchuk, Guwahati 781035, Assam, India; Faculty of Science, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India.
| |
Collapse
|
4
|
Magrì A, Tomasello B, Naletova I, Tabbì G, Cairns WRL, Greco V, Sciuto S, La Mendola D, Rizzarelli E. New BDNF and NT-3 Cyclic Mimetics Concur with Copper to Activate Trophic Signaling Pathways as Potential Molecular Entities to Protect Old Brains from Neurodegeneration. Biomolecules 2024; 14:1104. [PMID: 39334869 PMCID: PMC11430436 DOI: 10.3390/biom14091104] [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: 08/05/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
A low level of Neurotrophins (NTs), their Tyrosine Kinase Receptors (Trks), Vascular Endothelial Growth Factors (VEGFs) and their receptors, mainly VEGFR1 and VEGFR2, characterizes AD brains. The use of NTs and VEGFs as drugs presents different issues due to their low permeability of the blood-brain barrier, the poor pharmacokinetic profile, and the relevant side effects. To overcome these issues, different functional and structural NT mimics have been employed. Being aware that the N-terminus domain as the key domain of NTs for the binding selectivity and activation of Trks and the need to avoid or delay proteolysis, we herein report on the mimicking ability of two cyclic peptide encompassing the N-terminus of Brain Derived Growth Factor (BDNF), (c-[HSDPARRGELSV-]), cBDNF(1-12) and of Neurotrophin3 (NT3), (c-[YAEHKSHRGEYSV-]), cNT3(1-13). The two cyclic peptide features were characterized by a combined thermodynamic and spectroscopic approach (potentiometry, NMR, UV-vis and CD) that was extended to their copper(II) ion complexes. SH-SY5Y cell assays show that the Cu2+ present at the sub-micromolar level in the complete culture media affects the treatments with the two peptides. cBDNF(1-12) and cNT3(1-13) act as ionophores, induce neuronal differentiation and promote Trks and CREB phosphorylation in a copper dependent manner. Consistently, both peptide and Cu2+ stimulate BDNF and VEGF expression as well as VEGF release; cBDNF(1-12) and cNT3(1-13) induce the expression of Trks and VEGFRs.
Collapse
Affiliation(s)
- Antonio Magrì
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy; (A.M.); (I.N.); (G.T.)
| | - Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy;
| | - Irina Naletova
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy; (A.M.); (I.N.); (G.T.)
| | - Giovanni Tabbì
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy; (A.M.); (I.N.); (G.T.)
| | - Warren R. L. Cairns
- CNR-Institute of Polar Sciences (CNR-ISP), 155 Via Torino, 30172 Venice, Italy;
| | - Valentina Greco
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| | - Sebastiano Sciuto
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, via Bonanno Pisano 6, 56126 Pisa, Italy;
| | - Enrico Rizzarelli
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy; (A.M.); (I.N.); (G.T.)
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| |
Collapse
|
5
|
Cheng J, Wu C, Wang Y, Wang Z, He Y, Shen J. The antidepressant-like effect and proposed mechanism of action of TPN672MA, a novel serotonin-dopamine receptor modulator for the treatment of schizophrenia. Pharmacol Biochem Behav 2024; 242:173809. [PMID: 38936482 DOI: 10.1016/j.pbb.2024.173809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/12/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
TPN672MA, an innovative antipsychotic drug candidate currently in clinical trials, acts as a dopamine D2/D3 receptor partial agonist, serotonin 5-HT1A receptor agonist, and serotonin 5-HT2A receptor antagonist. Preclinical investigations have demonstrated its potential in treating the core symptoms of schizophrenia. The present study highlights TPN672MA's significant antidepressant-like effects in classical behavioral models, such as the chronic social defeat stress paradigm. The pronounced 5-HT1A receptor agonism and D2/D3 receptor partial agonism of TPN672MA likely contribute to its therapeutic effects in depression. Additionally, TPN672MA's antidepressant-like efficacy may be linked to its ability to enhance the expression levels of brain-derived neurotrophic factor (BDNF) and postsynaptic density protein-95 (PSD95) in the hippocampus. Furthermore, TPN672MA displayed a more rapid onset of antidepressant-like action. In conclusion, TPN672MA represents a promising new drug candidate for the treatment of symptoms of schizophrenia and depression.
Collapse
Affiliation(s)
- Jiaxin Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Chunhui Wu
- Vigonvita (Shanghai) Life Sciences Co., Ltd., Shanghai 200000, China.
| | - Yu Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Zhen Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yang He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Jingshan Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
6
|
Wang L, Du J, Liu Q, Wang D, Wang W, Lei M, Li K, Li Y, Hao A, Sang Y, Yi F, Zhou W, Liu H, Mao C, Qiu J. Wrapping stem cells with wireless electrical nanopatches for traumatic brain injury therapy. Nat Commun 2024; 15:7223. [PMID: 39174514 PMCID: PMC11341554 DOI: 10.1038/s41467-024-51098-y] [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: 12/08/2023] [Accepted: 07/29/2024] [Indexed: 08/24/2024] Open
Abstract
Electrical stimulation holds promise for enhancing neuronal differentiation of neural stem cells to treat traumatic brain injury. However, once the stem cells leave the stimulating material and migrate post transplantation, electrical stimulation on them is diminished. Here, we wrap the stem cells with wireless electrical nanopatches, the conductive graphene nanosheets. Under electromagnetic induction, electrical stimulation can thus be applied in-situ to individual nanopatch-wrapped stem cells on demand, stimulating their neuronal differentiation through a MAPK/ERK signaling pathway. Consequently, 41% of the nanopatch-wrapped stem cells differentiate into functional neurons in 5 days, as opposed to only 16.3% of the unwrapped ones. The brain injury male mice implanted with the nanopatch-wrapped stem cells and exposed to a rotating magnetic field 30 min/day exhibit significant recovery of brain tissues, behaviors, and cognitions, within 28 days. This study opens up an avenue to individualized electrical stimulation of transplanted stem cells for treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Liang Wang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China
| | - Jingyi Du
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Qilu Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China
| | - Dongshuang Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Wenhan Wang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China
| | - Ming Lei
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China
| | - Keyi Li
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China
| | - Yiwei Li
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China
| | - Aijun Hao
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Yuanhua Sang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China
| | - Fan Yi
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Wenjuan Zhou
- Key Laboratory for Experimental Teratology of Ministry of Education, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China.
- Shandong Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China.
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China.
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China.
| | - Jichuan Qiu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China.
| |
Collapse
|
7
|
Latif‐Hernandez A, Yang T, Butler RR, Losada PM, Minhas PS, White H, Tran KC, Liu H, Simmons DA, Langness V, Andreasson KI, Wyss‐Coray T, Longo FM. A TrkB and TrkC partial agonist restores deficits in synaptic function and promotes activity-dependent synaptic and microglial transcriptomic changes in a late-stage Alzheimer's mouse model. Alzheimers Dement 2024; 20:4434-4460. [PMID: 38779814 PMCID: PMC11247716 DOI: 10.1002/alz.13857] [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: 10/09/2023] [Revised: 03/12/2024] [Accepted: 04/02/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION Tropomyosin related kinase B (TrkB) and C (TrkC) receptor signaling promotes synaptic plasticity and interacts with pathways affected by amyloid beta (Aβ) toxicity. Upregulating TrkB/C signaling could reduce Alzheimer's disease (AD)-related degenerative signaling, memory loss, and synaptic dysfunction. METHODS PTX-BD10-2 (BD10-2), a small molecule TrkB/C receptor partial agonist, was orally administered to aged London/Swedish-APP mutant mice (APPL/S) and wild-type controls. Effects on memory and hippocampal long-term potentiation (LTP) were assessed using electrophysiology, behavioral studies, immunoblotting, immunofluorescence staining, and RNA sequencing. RESULTS In APPL/S mice, BD10-2 treatment improved memory and LTP deficits. This was accompanied by normalized phosphorylation of protein kinase B (Akt), calcium-calmodulin-dependent kinase II (CaMKII), and AMPA-type glutamate receptors containing the subunit GluA1; enhanced activity-dependent recruitment of synaptic proteins; and increased excitatory synapse number. BD10-2 also had potentially favorable effects on LTP-dependent complement pathway and synaptic gene transcription. DISCUSSION BD10-2 prevented APPL/S/Aβ-associated memory and LTP deficits, reduced abnormalities in synapse-related signaling and activity-dependent transcription of synaptic genes, and bolstered transcriptional changes associated with microglial immune response. HIGHLIGHTS Small molecule modulation of tropomyosin related kinase B (TrkB) and C (TrkC) restores long-term potentiation (LTP) and behavior in an Alzheimer's disease (AD) model. Modulation of TrkB and TrkC regulates synaptic activity-dependent transcription. TrkB and TrkC receptors are candidate targets for translational therapeutics. Electrophysiology combined with transcriptomics elucidates synaptic restoration. LTP identifies neuron and microglia AD-relevant human-mouse co-expression modules.
Collapse
Affiliation(s)
- Amira Latif‐Hernandez
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Tao Yang
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Robert R. Butler
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Patricia Moran Losada
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordCaliforniaUSA
| | - Paras S. Minhas
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Halle White
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Kevin C. Tran
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Harry Liu
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Danielle A. Simmons
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Vanessa Langness
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Katrin I. Andreasson
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordCaliforniaUSA
- Chan Zuckerberg BiohubSan FranciscoCaliforniaUSA
| | - Tony Wyss‐Coray
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordCaliforniaUSA
- The Phil and Penny Knight Initiative for Brain ResilienceStanford UniversityStanfordCaliforniaUSA
| | - Frank M. Longo
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordCaliforniaUSA
| |
Collapse
|
8
|
Bernardes CP, Lopes Pinheiro E, Ferreira IG, de Oliveira IS, dos Santos NAG, Sampaio SV, Arantes EC, dos Santos AC. Fraction of C. d. collilineatus venom containing crotapotin protects PC12 cells against MPP + toxicity by activating the NGF-signaling pathway. J Venom Anim Toxins Incl Trop Dis 2024; 30:e20230056. [PMID: 38915449 PMCID: PMC11194915 DOI: 10.1590/1678-9199-jvatitd-2023-0056] [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: 08/23/2023] [Accepted: 05/08/2024] [Indexed: 06/26/2024] Open
Abstract
Background Parkinson's disease (PD) is the second most prevalent neurodegenerative disease. There is no effective treatment for neurodegenerative diseases. Snake venoms are a cocktail of proteins and peptides with great therapeutic potential and might be useful in the treatment of neurodegenerative diseases. Crotapotin is the acid chain of crotoxin, the major component of Crotalus durissus collilineatus venom. PD is characterized by low levels of neurotrophins, and synaptic and axonal degeneration; therefore, neurotrophic compounds might delay the progression of PD. The neurotrophic potential of crotapotin has not been studied yet. Methods We evaluated the neurotrophic potential of crotapotin in untreated PC12 cells, by assessing the induction of neurite outgrowth. The activation of the NGF signaling pathway was investigated through pharmacological inhibition of its main modulators. Additionally, its neuroprotective and neurorestorative effects were evaluated by assessing neurite outgrowth and cell viability in PC12 cells treated with the dopaminergic neurotoxin MPP+ (1-methyl-4-phenylpyridinium), known to induce Parkinsonism in humans and animal models. Results Crotapotin induced neuritogenesis in PC12 cells through the NGF-signaling pathway, more specifically, by activating the NGF-selective receptor trkA, and the PI3K/Akt and the MAPK/ERK cascades, which are involved in neuronal survival and differentiation. In addition, crotapotin had no cytotoxic effect and protected PC12 cells against the inhibitory effects of MPP+ on cell viability and differentiation. Conclusion These findings show, for the first time, that crotapotin has neurotrophic/neuroprotective/neurorestorative potential and might be beneficial in Parkinson's disease. Additional studies are necessary to evaluate the toxicity of crotapotin in other cell models.
Collapse
Affiliation(s)
- Carolina Petri Bernardes
- Department of Clinical Analyses, Toxicology and Food Science, School
of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP),
Ribeirão Preto, SP, Brazil
| | - Ernesto Lopes Pinheiro
- Department of Biomolecular Sciences, School of Pharmaceutical
Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP,
Brazil
| | - Isabela Gobbo Ferreira
- Department of Biomolecular Sciences, School of Pharmaceutical
Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP,
Brazil
| | - Isadora Sousa de Oliveira
- Department of Biomolecular Sciences, School of Pharmaceutical
Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP,
Brazil
| | - Neife Aparecida Guinaim dos Santos
- Department of Clinical Analyses, Toxicology and Food Science, School
of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP),
Ribeirão Preto, SP, Brazil
| | - Suely Vilela Sampaio
- Department of Clinical Analyses, Toxicology and Food Science, School
of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP),
Ribeirão Preto, SP, Brazil
| | - Eliane Candiani Arantes
- Department of Biomolecular Sciences, School of Pharmaceutical
Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP,
Brazil
| | - Antonio Cardozo dos Santos
- Department of Clinical Analyses, Toxicology and Food Science, School
of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP),
Ribeirão Preto, SP, Brazil
| |
Collapse
|
9
|
Wu X, Zhou Y, Xi Y, Zhou H, Tang Z, Xiong L, Qin D. Polyphenols: Natural Food-Grade Biomolecules for the Treatment of Nervous System Diseases from a Multi-Target Perspective. Pharmaceuticals (Basel) 2024; 17:775. [PMID: 38931442 PMCID: PMC11206395 DOI: 10.3390/ph17060775] [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: 05/24/2024] [Revised: 06/08/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Polyphenols are the most prevalent naturally occurring phytochemicals in the human diet and range in complexity from simple molecules to high-molecular-weight polymers. They have a broad range of chemical structures and are generally categorized as "neuroprotective", "anti-inflammatory", and "antioxidant" given their main function of halting disease onset and promoting health. Research has shown that some polyphenols and their metabolites can penetrate the blood-brain barrier and hence increase neuroprotective signaling and neurohormonal effects to provide anti-inflammatory and antioxidant effects. Therefore, multi-targeted modulation of polyphenols may prevent the progression of neuropsychiatric disorders and provide a new practical therapeutic strategy for difficult-to-treat neuropsychiatric disorders. Therefore, multi-target modulation of polyphenols has the potential to prevent the progression of neuropsychiatric disorders and provide a new practical therapeutic strategy for such nervous system diseases. Herein, we review the therapeutic benefits of polyphenols on autism-spectrum disorders, anxiety disorders, depression, and sleep disorders, along with in vitro and ex vivo experimental and clinical trials. Although their methods of action are still under investigation, polyphenols are still seldom employed directly as therapeutic agents for nervous system disorders. Comprehensive mechanistic investigations and large-scale multicenter randomized controlled trials are required to properly evaluate the safety, effectiveness, and side effects of polyphenols.
Collapse
Affiliation(s)
- Xinchen Wu
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (X.W.); (Y.Z.); (Y.X.)
| | - Yang Zhou
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (X.W.); (Y.Z.); (Y.X.)
| | - Yujiang Xi
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (X.W.); (Y.Z.); (Y.X.)
| | - Haimei Zhou
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming 650500, China; (H.Z.); (Z.T.)
| | - Zhengxiu Tang
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming 650500, China; (H.Z.); (Z.T.)
| | - Lei Xiong
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (X.W.); (Y.Z.); (Y.X.)
| | - Dongdong Qin
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming 650500, China; (H.Z.); (Z.T.)
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming 650500, China
| |
Collapse
|
10
|
Tomasello B, Bellia F, Naletova I, Magrì A, Tabbì G, Attanasio F, Tomasello MF, Cairns WRL, Fortino M, Pietropaolo A, Greco V, La Mendola D, Sciuto S, Arena G, Rizzarelli E. BDNF- and VEGF-Responsive Stimulus to an NGF Mimic Cyclic Peptide with Copper Ionophore Capability and Ctr1/CCS-Driven Signaling. ACS Chem Neurosci 2024; 15:1755-1769. [PMID: 38602894 DOI: 10.1021/acschemneuro.3c00716] [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] [Indexed: 04/13/2024] Open
Abstract
Neurotrophins are a family of growth factors that play a key role in the development and regulation of the functioning of the central nervous system. Their use as drugs is made difficult by their poor stability, cellular permeability, and side effects. Continuing our effort to use peptides that mimic the neurotrophic growth factor (NGF), the family model protein, and specifically the N-terminus of the protein, here we report on the spectroscopic characterization and resistance to hydrolysis of the 14-membered cyclic peptide reproducing the N-terminus sequence (SSSHPIFHRGEFSV (c-NGF(1-14)). Far-UV CD spectra and a computational study show that this peptide has a rigid conformation and left-handed chirality typical of polyproline II that favors its interaction with the D5 domain of the NGF receptor TrkA. c-NGF(1-14) is able to bind Cu2+ with good affinity; the resulting complexes have been characterized by potentiometric and spectroscopic measurements. Experiments on PC12 cells show that c-NGF(1-14) acts as an ionophore, influencing the degree and the localization of both the membrane transporter (Ctr1) and the copper intracellular transporter (CCS). c-NGF(1-14) induces PC12 differentiation, mimics the protein in TrkA phosphorylation, and activates the kinase cascade, inducing Erk1/2 phosphorylation. c-NGF(1-14) biological activities are enhanced when the peptide interacts with Cu2+ even with the submicromolar quantities present in the culture media as demonstrated by ICP-OES measurements. Finally, c-NGF(1-14) and Cu2+ concur to activate the cAMP response element-binding protein CREB that, in turn, induces the brain-derived neurotrophic factor (BDNF) and the vascular endothelial growth factor (VEGF) release.
Collapse
Affiliation(s)
- Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, V.le Andrea Doria 6, Catania 95125, Italy
| | - Francesco Bellia
- Institute of Crystallography, CNR, P. Gaifami 18, Catania 95126, Italy
| | - Irina Naletova
- Institute of Crystallography, CNR, P. Gaifami 18, Catania 95126, Italy
| | - Antonio Magrì
- Institute of Crystallography, CNR, P. Gaifami 18, Catania 95126, Italy
| | - Giovanni Tabbì
- Institute of Crystallography, CNR, P. Gaifami 18, Catania 95126, Italy
| | | | | | - Warren R L Cairns
- Istituto di Scienze Polari (ISP), c/o Campus Scientifico, Università Ca' Foscari Venezia Via Torino, Venezia Mestre 155-30170, Italy
| | - Mariagrazia Fortino
- Dipartimento di Scienze della Salute, Università di Catanzaro, Viale Europa, Catanzaro 88100, Italy
| | - Adriana Pietropaolo
- Dipartimento di Scienze della Salute, Università di Catanzaro, Viale Europa, Catanzaro 88100, Italy
| | - Valentina Greco
- Department of Chemical Sciences, University of Catania, A. Doria 6, Catania 95125, Italy
| | - Diego La Mendola
- Department of Pharmaceutical Sciences, University of Pisa, Bonanno Pisano 12, Pisa 56126, Italy
| | - Sebastiano Sciuto
- Department of Chemical Sciences, University of Catania, A. Doria 6, Catania 95125, Italy
| | - Giuseppe Arena
- Department of Chemical Sciences, University of Catania, A. Doria 6, Catania 95125, Italy
| | - Enrico Rizzarelli
- Institute of Crystallography, CNR, P. Gaifami 18, Catania 95126, Italy
- Department of Chemical Sciences, University of Catania, A. Doria 6, Catania 95125, Italy
| |
Collapse
|
11
|
Thompson T, Pewklang T, Piyanuch P, Wanichacheva N, Kamkaew A, Burgess K. A fluorescent electrophile for CLIPS: self indicating TrkB binders. Org Biomol Chem 2024; 22:506-512. [PMID: 38111346 PMCID: PMC10863675 DOI: 10.1039/d3ob01654d] [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: 12/20/2023]
Abstract
Combination of cysteine-containing peptides with electrophiles provides efficient access to cyclo-organopeptides. However, there are no routes to intrinsically fluorescent cyclo-organopeptides containing robust, brilliant fluorophores emitting at wavelengths longer than cellular autofluorescence. We show such fluorescent cyclo-organopeptides can be made via SNAr reactions of cysteine-containing peptides with a BODIPY system. Seven compounds of this type were prepared to test as probes; six contained peptide sequences corresponding to loop regions in brain-derived neurotrophic factor and neurotrophic factor 4 (BDNF and NT-4) which bind tropomyocin receptor kinase B (TrkB). Cellular assays in serum-free media indicated two of the six key compounds induced survival of HEK293 cells stably transfected with TrkB whereas a control did not. The two compounds inducing cell survival bound TrkB on those cells (Kd ∼40 and 47 nM), illustrating how intrinsically fluorescent cyclo-organopeptides can be assayed for quantifiable binding to surface receptors in cell membrane environments.
Collapse
Affiliation(s)
- Tye Thompson
- Department of Chemistry, Texas A & M University, Box 30012, College Station, TX 77842-3012, USA.
| | - Thitima Pewklang
- Department of Chemistry, Texas A & M University, Box 30012, College Station, TX 77842-3012, USA.
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Pornthip Piyanuch
- Department of Chemistry, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Nantanit Wanichacheva
- Department of Chemistry, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Anyanee Kamkaew
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Kevin Burgess
- Department of Chemistry, Texas A & M University, Box 30012, College Station, TX 77842-3012, USA.
| |
Collapse
|
12
|
Denenberg S, Machin KL, Landsberg GM. Behavior and Cognition of the Senior Cat and Its Interaction with Physical Disease. Vet Clin North Am Small Anim Pract 2024; 54:153-168. [PMID: 37865588 DOI: 10.1016/j.cvsm.2023.09.001] [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] [Indexed: 10/23/2023]
Abstract
In cats, age-related pathologic condition and neurologic degeneration can produce changes in activity, vocalization, appearance, appetite, litter box use, sleep-wake cycle, personality, and cognitive ability. These changes can influence the relationship between owner and pet. Although cognitive dysfunction syndrome (CDS) can cause altered behavior later in life, other medical or behavioral causes may mimic these clinical signs or complicate diagnosis. Management and treatment of CDS can be accomplished through pharmacologic intervention, diet and nutritional supplementation, and environmental enrichment aimed at slowing the progression of the disease.
Collapse
Affiliation(s)
- Sagi Denenberg
- North Toronto Veterinary Behaviour Specialty Clinic, 8705 Yonge Street, Richmond Hill, Ontario L4C 6Z1, Canada.
| | - Karen L Machin
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan S7N 5B4, Canada
| | | |
Collapse
|
13
|
Dejanovic B, Sheng M, Hanson JE. Targeting synapse function and loss for treatment of neurodegenerative diseases. Nat Rev Drug Discov 2024; 23:23-42. [PMID: 38012296 DOI: 10.1038/s41573-023-00823-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2023] [Indexed: 11/29/2023]
Abstract
Synapse dysfunction and loss are hallmarks of neurodegenerative diseases that correlate with cognitive decline. However, the mechanisms and therapeutic strategies to prevent or reverse synaptic damage remain elusive. In this Review, we discuss recent advances in understanding the molecular and cellular pathways that impair synapses in neurodegenerative diseases, including the effects of protein aggregation and neuroinflammation. We also highlight emerging therapeutic approaches that aim to restore synaptic function and integrity, such as enhancing synaptic plasticity, preventing synaptotoxicity, modulating neuronal network activity and targeting immune signalling. We discuss the preclinical and clinical evidence for each strategy, as well as the challenges and opportunities for developing effective synapse-targeting therapeutics for neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Morgan Sheng
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jesse E Hanson
- Department of Neuroscience, Genentech, South San Francisco, CA, USA.
| |
Collapse
|
14
|
Zagrebelsky M, Korte M. Are TrkB receptor agonists the right tool to fulfill the promises for a therapeutic value of the brain-derived neurotrophic factor? Neural Regen Res 2024; 19:29-34. [PMID: 37488840 PMCID: PMC10479861 DOI: 10.4103/1673-5374.374138] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/20/2023] [Accepted: 03/27/2023] [Indexed: 07/26/2023] Open
Abstract
Brain-derived neurotrophic factor signaling via its receptor tropomyosin receptor kinase B regulates several crucial physiological processes. It has been shown to act in the brain, promoting neuronal survival, growth, and plasticity as well as in the rest of the body where it is involved in regulating for instance aspects of the metabolism. Due to its crucial and very pleiotropic activity, reduction of brain-derived neurotrophic factor levels and alterations in the brain-derived neurotrophic factor/tropomyosin receptor kinase B signaling have been found to be associated with a wide spectrum of neurological diseases. However, because of its poor bioavailability and pharmacological properties, brain-derived neurotrophic factor itself has a very low therapeutic value. Moreover, the concomitant binding of exogenous brain-derived neurotrophic factor to the p75 neurotrophin receptor has the potential to elicit several unwanted and deleterious side effects. Therefore, developing tools and approaches to specifically promote tropomyosin receptor kinase B signaling has become an important goal of translational research. Among the newly developed tools are different categories of tropomyosin receptor kinase B receptor agonist molecules. In this review, we give a comprehensive description of the different tropomyosin receptor kinase B receptor agonist drugs developed so far and of the results of their application in animal models of several neurological diseases. Moreover, we discuss the main benefits of tropomyosin receptor kinase B receptor agonists, concentrating especially on the new tropomyosin receptor kinase B agonist antibodies. The benefits observed both in vitro and in vivo upon application of tropomyosin receptor kinase B receptor agonist drugs seem to predominantly depend on their general neuroprotective activity and their ability to promote neuronal plasticity. Moreover, tropomyosin receptor kinase B agonist antibodies have been shown to specifically bind the tropomyosin receptor kinase B receptor and not p75 neurotrophin receptor. Therefore, while, based on the current knowledge, the tropomyosin receptor kinase B receptor agonists do not seem to have the potential to reverse the disease pathology per se, promoting brain-derived neurotrophic factor/tropomyosin receptor kinase B signaling still has a very high therapeutic relevance.
Collapse
Affiliation(s)
- Marta Zagrebelsky
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
| | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
- Helmholtz Centre for Infection Research, AG NIND, Braunschweig, Germany
| |
Collapse
|
15
|
Vink HA, Ramekers D, Foster AC, Versnel H. The efficacy of a TrkB monoclonal antibody agonist in preserving the auditory nerve in deafened guinea pigs. Hear Res 2023; 439:108895. [PMID: 37837701 DOI: 10.1016/j.heares.2023.108895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 08/31/2023] [Accepted: 10/02/2023] [Indexed: 10/16/2023]
Abstract
The auditory nerve typically degenerates following loss of cochlear hair cells or synapses. In the case of hair cell loss neural degeneration hinders restoration of hearing through a cochlear implant, and in the case of synaptopathy suprathreshold hearing is affected, potentially degrading speech perception in noise. It has been established that neurotrophins such as brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) can mitigate auditory nerve degeneration. Several potential BDNF mimetics have also been investigated for neurotrophic effects in the cochlea. A recent in vitro study showed favorable effects of M3, a TrkB monoclonal antibody agonist, when compared with BDNF. In the present study we set out to examine the effect of M3 on auditory nerve preservation in vivo. Thirty-one guinea pigs were bilaterally deafened, and unilaterally treated with a single 3-µl dose of 7 mg/ml, 0.7 mg/ml M3 or vehicle-only by means of a small gelatin sponge two weeks later. During the experiment and analyses the experimenters were blinded to the three treatment groups. Four weeks after treatment, we assessed the treatment effect (1) histologically, by quantifying survival of SGCs and their peripheral processes (PPs); and (2) electrophysiologically, with two different paradigms of electrically evoked compound action potential (eCAP) recordings shown to be indicative of neural health: single-pulse stimulation with varying inter-phase gap (IPG), and pulse-train stimulation with varying inter-pulse interval. We observed a consistent and significant preservative effect of M3 on SGC survival in the lower basal turn (approximately 40% more survival than in the untreated contralateral cochlea), but also in the upper middle and lower apical turn of the cochlea. This effect was similar for the two treatment groups. Survival of PPs showed a trend similar to that of the SGCs, but was only significantly higher for the highest dose of M3. The protective effect of M3 on SGCs was not reflected in any of the eCAP measures: no statistically significant differences were observed between groups in IPG effect nor between the M3 treatment groups and the control group using the pulse-train stimulation paradigm. In short, while a clear effect of M3 was observed on SGC survival, this was not clearly translated into functional preservation.
Collapse
Affiliation(s)
- Henk A Vink
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Utrecht, Utrecht University, Room G.02.531, P.O. Box 85500, 3508 GA, Utrecht, the Netherlands; UMC Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Dyan Ramekers
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Utrecht, Utrecht University, Room G.02.531, P.O. Box 85500, 3508 GA, Utrecht, the Netherlands; UMC Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | | | - Huib Versnel
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Utrecht, Utrecht University, Room G.02.531, P.O. Box 85500, 3508 GA, Utrecht, the Netherlands; UMC Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
16
|
Narducci D, Charou D, Rogdakis T, Zota I, Bafiti V, Zervou M, Katsila T, Gravanis A, Prousis KC, Charalampopoulos I, Calogeropoulou T. A quest for the stereo-electronic requirements for selective agonism for the neurotrophin receptors TrkA and TrkB in 17-spirocyclic-dehydroepiandrosterone derivatives. Front Mol Neurosci 2023; 16:1244133. [PMID: 37840771 PMCID: PMC10568017 DOI: 10.3389/fnmol.2023.1244133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/31/2023] [Indexed: 10/17/2023] Open
Abstract
Introduction The neurotrophin system plays a pivotal role in the development, morphology, and survival of the nervous system, and its dysregulation has been manifested in numerous neurodegenerative and neuroinflammatory diseases. Neurotrophins NGF and BDNF are major growth factors that prevent neuronal death and synaptic loss through binding with high affinity to their specific tropomyosin-related kinase receptors namely, TrkA and TrkB, respectively. The poor pharmacokinetic properties prohibit the use of neurotrophins as therapeutic agents. Our group has previously synthesized BNN27, a prototype small molecule based on dehydroepiandrosterone, mimicking NGF through the activation of the TrkA receptor. Methods To obtain a better understanding of the stereo-electronic requirements for selective activation of TrkA and TrkB receptors, 27 new dehydroepiandrosterone derivatives bearing a C17-spiro-dihydropyran or cyclobutyl moiety were synthesized. The new compounds were evaluated for their ability (a) to selectively activate the TrkA receptor and its downstream signaling kinases Akt and Erk1/2 in PC12 cells, protecting these cells from serum deprivation-induced cell death, and (b) to induce phosphorylation of TrkB and to promote cell survival under serum deprivation conditions in NIH3T3 cells stable transfected with the TrkB receptor and primary cortical astrocytes. In addition the metabolic stability and CYP-mediated reaction was assessed. Results Among the novel derivatives, six were able to selectively protect PC12 cells through interaction with the TrkA receptor and five more to selectively protect TrkB-expressing cells via interaction with the TrkB receptor. In particular, compound ENT-A025 strongly induces TrkA and Erk1/2 phosphorylation, comparable to NGF, and can protect PC12 cells against serum deprivation-induced cell death. Furthermore, ENT-A065, ENT-A066, ENT-A068, ENT-A069, and ENT-A070 showed promising pro-survival effects in the PC12 cell line. Concerning TrkB agonists, ENT-A009 and ENT-A055 were able to induce phosphorylation of TrkB and reduce cell death levels in NIH3T3-TrkB cells. In addition, ENT-A076, ENT-A087, and ENT-A088 possessed antiapoptotic activity in NIH-3T3-TrkB cells exclusively mediated through the TrkB receptor. The metabolic stability and CYP-mediated reaction phenotyping of the potent analogs did not reveal any major liabilities. Discussion We have identified small molecule selective agonists of TrkA and TrkB receptors as promising lead neurotrophin mimetics for the development of potential therapeutics against neurodegenerative conditions.
Collapse
Affiliation(s)
- Daniele Narducci
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Despoina Charou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
| | - Thanasis Rogdakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
| | - Ioanna Zota
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
| | - Vivi Bafiti
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Maria Zervou
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Theodora Katsila
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Achille Gravanis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
| | - Kyriakos C. Prousis
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Ioannis Charalampopoulos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
| | | |
Collapse
|
17
|
Stansberry WM, Pierchala BA. Neurotrophic factors in the physiology of motor neurons and their role in the pathobiology and therapeutic approach to amyotrophic lateral sclerosis. Front Mol Neurosci 2023; 16:1238453. [PMID: 37692101 PMCID: PMC10483118 DOI: 10.3389/fnmol.2023.1238453] [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: 06/11/2023] [Accepted: 08/11/2023] [Indexed: 09/12/2023] Open
Abstract
The discovery of the neurotrophins and their potent survival and trophic effects led to great enthusiasm about their therapeutic potential to rescue dying neurons in neurodegenerative diseases. The further discovery that brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF) and glial cell line-derived neurotrophic factor (GDNF) had potent survival-promoting activity on motor neurons led to the proposal for their use in motor neuron diseases such as amyotrophic lateral sclerosis (ALS). In this review we synthesize the literature pertaining to the role of NGF, BDNF, CNTF and GDNF on the development and physiology of spinal motor neurons, as well as the preclinical studies that evaluated their potential for the treatment of ALS. Results from the clinical trials of these molecules will also be described and, with the aid of decades of hindsight, we will discuss what can reasonably be concluded and how this information can inform future clinical development of neurotrophic factors for ALS.
Collapse
Affiliation(s)
- Wesley M. Stansberry
- The Department of Anatomy, Cell Biology and Physiology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Brian A. Pierchala
- The Department of Anatomy, Cell Biology and Physiology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
18
|
Antipova TA, Logvinov IO, Deyev IE, Povarnina PY, Vakhitova YV, Gudasheva TA, Seredenin SB. Pharmacogenetic Analysis of the Interaction of the Low-Molecular-Weight BDNF Mimetic Dipeptide GSB-106 with TRK Receptors. DOKL BIOCHEM BIOPHYS 2023; 511:166-168. [PMID: 37833601 DOI: 10.1134/s1607672923700230] [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: 03/15/2023] [Revised: 04/03/2023] [Accepted: 04/03/2023] [Indexed: 10/15/2023]
Abstract
Using TrkA or TrkB receptor gene knockout HT-22 cells, the selectivity of the interaction of the low-molecular-weight dipeptide BDNF mimetic GSB-106 (hexamethylenediamide bis(N-monosuccinyl-L-seryl-L-lysine)) with TrkB receptors was shown.
Collapse
Affiliation(s)
- T A Antipova
- Zakusov Research Institute of Pharmacology, Moscow, Russia
| | - I O Logvinov
- Zakusov Research Institute of Pharmacology, Moscow, Russia
| | - I E Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - P Yu Povarnina
- Zakusov Research Institute of Pharmacology, Moscow, Russia
| | - Yu V Vakhitova
- Zakusov Research Institute of Pharmacology, Moscow, Russia
| | - T A Gudasheva
- Zakusov Research Institute of Pharmacology, Moscow, Russia.
| | - S B Seredenin
- Zakusov Research Institute of Pharmacology, Moscow, Russia
| |
Collapse
|
19
|
do Amaral L, Dos Santos NAG, Sisti FM, Del Bel E, Dos Santos AC. Doxycycline inhibits dopaminergic neurodegeneration through upregulation of axonal and synaptic proteins. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1787-1796. [PMID: 36843128 DOI: 10.1007/s00210-023-02435-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 02/18/2023] [Indexed: 02/28/2023]
Abstract
Doxycycline (DOX) is a widely used antibiotic that is able to cross the blood-brain barrier. Several studies have shown its neuroprotective effect against neurodegeneration and have associated it with antioxidant, anti-apoptotic, and anti-inflammatory mechanisms. We have recently demonstrated that DOX mimics nerve growth factor (NGF) signaling in PC12 cells. However, the involvement of this mechanism in the neuroprotective effect of DOX is unknown. Axonal degeneration and synaptic loss are key events at the early stages of neurodegeneration, and precede the neuronal death in neurodegenerative diseases, including Parkinson's disease (PD). Therefore, the regeneration of the axonal and synaptic network might be beneficial in PD. The effect of DOX in PC12 cells treated with the Parkinsonian neurotoxin 1-methyl-4-phenylpyridinium (MPP+) was addressed. Doxycycline reduced the inhibition of neuritogenesis induced by MPP+, even in cells deprived of NGF. The mechanism involved the upregulation of GAP-43, synapsin I, β-III-tubulin, F-actin, and neurofilament-200, proteins that are associated with axonal and synaptic plasticity. Considering the role of axonal degeneration and synaptic loss at the initial stages of PD, the recent advances in early diagnosis of neurodegeneration, and the advantages of drug repurposing, doxycycline is a promising candidate to treat PD.
Collapse
Affiliation(s)
- Lilian do Amaral
- Department of Clinical, Toxicological and Bromatological Analysis, School of Pharmaceutical Sciences of Ribeirão Preto, Av Do Café S/N, University of São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - Neife Aparecida Guinaim Dos Santos
- Department of Clinical, Toxicological and Bromatological Analysis, School of Pharmaceutical Sciences of Ribeirão Preto, Av Do Café S/N, University of São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - Flávia Malvestio Sisti
- Department of Clinical, Toxicological and Bromatological Analysis, School of Pharmaceutical Sciences of Ribeirão Preto, Av Do Café S/N, University of São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - Elaine Del Bel
- Department of Basic and Oral Biology, Faculty of Dentistry of Ribeirão Preto, USP, Av Do Café S/N, 14040-904, Ribeirão Preto, SP, Brazil
| | - Antônio Cardozo Dos Santos
- Department of Clinical, Toxicological and Bromatological Analysis, School of Pharmaceutical Sciences of Ribeirão Preto, Av Do Café S/N, University of São Paulo, Ribeirão Preto, SP, 14040-903, Brazil.
| |
Collapse
|
20
|
Martínez‐Mármol R, Chai Y, Conroy JN, Khan Z, Hong S, Kim SB, Gormal RS, Lee DH, Lee JK, Coulson EJ, Lee MK, Kim SY, Meunier FA. Hericerin derivatives activates a pan-neurotrophic pathway in central hippocampal neurons converging to ERK1/2 signaling enhancing spatial memory. J Neurochem 2023; 165:791-808. [PMID: 36660878 PMCID: PMC10952766 DOI: 10.1111/jnc.15767] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/31/2022] [Accepted: 01/04/2023] [Indexed: 01/21/2023]
Abstract
The traditional medicinal mushroom Hericium erinaceus is known for enhancing peripheral nerve regeneration through targeting nerve growth factor (NGF) neurotrophic activity. Here, we purified and identified biologically new active compounds from H. erinaceus, based on their ability to promote neurite outgrowth in hippocampal neurons. N-de phenylethyl isohericerin (NDPIH), an isoindoline compound from this mushroom, together with its hydrophobic derivative hericene A, were highly potent in promoting extensive axon outgrowth and neurite branching in cultured hippocampal neurons even in the absence of serum, demonstrating potent neurotrophic activity. Pharmacological inhibition of tropomyosin receptor kinase B (TrkB) by ANA-12 only partly prevented the NDPIH-induced neurotrophic activity, suggesting a potential link with BDNF signaling. However, we found that NDPIH activated ERK1/2 signaling in the absence of TrkB in HEK-293T cells, an effect that was not sensitive to ANA-12 in the presence of TrkB. Our results demonstrate that NDPIH acts via a complementary neurotrophic pathway independent of TrkB with converging downstream ERK1/2 activation. Mice fed with H. erinaceus crude extract and hericene A also exhibited increased neurotrophin expression and downstream signaling, resulting in significantly enhanced hippocampal memory. Hericene A therefore acts through a novel pan-neurotrophic signaling pathway, leading to improved cognitive performance.
Collapse
Affiliation(s)
- Ramón Martínez‐Mármol
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain Institute, The University of QueenslandBrisbaneQueenslandAustralia
| | - YeJin Chai
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain Institute, The University of QueenslandBrisbaneQueenslandAustralia
| | - Jacinta N. Conroy
- School of Biomedical SciencesThe University of QueenslandBrisbaneQueenslandAustralia
| | - Zahra Khan
- College of PharmacyGachon UniversityIncheonRepublic of Korea
| | - Seong‐Min Hong
- College of PharmacyGachon UniversityIncheonRepublic of Korea
| | - Seon Beom Kim
- College of PharmacyChungbuk National UniversityCheongjuRepublic of Korea
| | - Rachel S. Gormal
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain Institute, The University of QueenslandBrisbaneQueenslandAustralia
| | - Dae Hee Lee
- CNGBio corpCheongju‐siChungcheongbuk‐doRepublic of Korea
| | - Jae Kang Lee
- CNGBio corpCheongju‐siChungcheongbuk‐doRepublic of Korea
| | - Elizabeth J. Coulson
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain Institute, The University of QueenslandBrisbaneQueenslandAustralia
- School of Biomedical SciencesThe University of QueenslandBrisbaneQueenslandAustralia
| | - Mi Kyeong Lee
- College of PharmacyChungbuk National UniversityCheongjuRepublic of Korea
| | - Sun Yeou Kim
- College of PharmacyGachon UniversityIncheonRepublic of Korea
- Gachon Institute of Pharmaceutical ScienceGachon UniversityIncheonRepublic of Korea
| | - Frédéric A. Meunier
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain Institute, The University of QueenslandBrisbaneQueenslandAustralia
- School of Biomedical SciencesThe University of QueenslandBrisbaneQueenslandAustralia
| |
Collapse
|
21
|
Madhubala D, Patra A, Islam T, Saikia K, Khan MR, Ahmed SA, Borah JC, Mukherjee AK. Snake venom nerve growth factor-inspired designing of novel peptide therapeutics for the prevention of paraquat-induced apoptosis, neurodegeneration, and alteration of metabolic pathway genes in the rat pheochromocytoma PC-12 cell. Free Radic Biol Med 2023; 197:23-45. [PMID: 36669545 DOI: 10.1016/j.freeradbiomed.2023.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/03/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023]
Abstract
Neurodegenerative disorders (ND), associated with the progressive loss of neurons, oxidative stress-mediated production of reactive oxygen species (ROS), and mitochondrial dysfunction, can be treated with synthetic peptides possessing innate neurotrophic effects and neuroprotective activity. Computational analysis of two small synthetic peptides (trideca-neuropeptide, TNP; heptadeca-neuropeptide, HNP) developed from the nerve growth factors from snake venoms predicted their significant interaction with the human TrkA receptor (TrkA). In silico results were validated by an in vitro binding study of the FITC-conjugated custom peptides to rat pheochromocytoma PC-12 cell TrkA receptors. Pre-treatment of PC-12 cells with TNP and HNP induced neuritogenesis and significantly reduced the paraquat (PT)-induced cellular toxicity, the release of lactate dehydrogenase from the cell cytoplasm, production of intracellular ROS, restored the level of antioxidants, prevented alteration of mitochondrial transmembrane potential (ΔΨm) and adenosine triphosphate (ATP) production, and inhibited cellular apoptosis. These peptides lack in vitro cytotoxicity, haemolytic activity, and platelet-modulating properties and do not interfere with the blood coagulation system. Functional proteomic analyses demonstrated the reversal of PT-induced upregulated and downregulated metabolic pathway genes in PC-12 cells that were pre-treated with HNP and revealed the metabolic pathways regulated by HNP to induce neuritogenesis and confer protection against PT-induced neuronal damage in PC-12. The quantitative RT-PCR analysis confirmed that the PT-induced increased and decreased expression of critical pro-apoptotic and anti-apoptotic genes had been restored in the PC-12 cells pre-treated with the custom peptides. A network gene expression profile was proposed to elucidate the molecular interactions among the regulatory proteins for HNP to salvage the PT-induced damage. Taken together, our results show how the peptides can rescue PT-induced oxidative stress, mitochondrial dysfunction, and cellular death and suggest new opportunities for developing neuroprotective drugs.
Collapse
Affiliation(s)
- Dev Madhubala
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, 784028, Assam, India; Microbial Biotechnology and Protein Research Laboratory, Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati, 781035, Assam, India
| | - Aparup Patra
- Microbial Biotechnology and Protein Research Laboratory, Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati, 781035, Assam, India
| | - Taufikul Islam
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, 784028, Assam, India
| | - Kangkon Saikia
- Microbial Biotechnology and Protein Research Laboratory, Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati, 781035, Assam, India
| | - Mojibur R Khan
- Microbial Biotechnology and Protein Research Laboratory, Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati, 781035, Assam, India
| | - Semim Akhtar Ahmed
- Microbial Biotechnology and Protein Research Laboratory, Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati, 781035, Assam, India
| | - Jagat C Borah
- Microbial Biotechnology and Protein Research Laboratory, Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati, 781035, Assam, India
| | - Ashis K Mukherjee
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, 784028, Assam, India; Microbial Biotechnology and Protein Research Laboratory, Institute of Advanced Studies in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati, 781035, Assam, India.
| |
Collapse
|
22
|
Saadh MJ. Potential protective effects of red grape seed extract in a rat model of malathion-induced neurotoxicity. Vet World 2023; 16:380-385. [PMID: 37042003 PMCID: PMC10082724 DOI: 10.14202/vetworld.2023.380-385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/18/2023] [Indexed: 02/27/2023] Open
Abstract
Background and Aim: Exposure to pesticide mixtures used in agricultural practice poses a grave risk to non-target animals. This study aimed to determine whether red grape seed extract (RGSE, which is 95% bioflavonoids and equal to 12,000 mg of fresh red grape seed, and 150 mg of vitamin C) alleviated the changes in brain-derived neurotrophic factor (BDNF) level, acetylcholinesterase activity, oxidative stress, and apoptosis induced by orally administered malathion in a rat model of malathion-induced neurotoxicity.
Materials and Methods: Thirty-two adult male Wistar albino rats were divided into four groups and exposed to malathion with or without 4 weeks of RGSE treatment, treated with RGSE alone, or left untreated as controls. The animals were euthanized 24 h after last treatment. Brain samples were collected to measure acetylcholinesterase, superoxide dismutase (SOD), and caspase 3 activity, total antioxidant capacity (TAC), and BDNF levels.
Results: Malathion significantly reduced acetylcholinesterase and SOD activity and TAC and significantly increased caspase 3 activity. In comparison, acetylcholinesterase and SOC activity, BDNF level, and TAC were improved and caspase 3 activity was decreased in the malathion-RGSE group, indicating that RGSE corrected the alterations detected in these biochemical parameters.
Conclusion: Oxidative stress and apoptosis in the brains of rats exposed to oral malathion were substantially controlled by RGSE treatment.
Collapse
Affiliation(s)
- Mohamed Jamal Saadh
- Department of Basic Science, Faculty of Pharmacy, Middle East University, Amman, Jordan; Applied Science Research Center, Applied Science Private University, Amman, Jordan
| |
Collapse
|
23
|
NGF and Its Role in Immunoendocrine Communication during Metabolic Syndrome. Int J Mol Sci 2023; 24:ijms24031957. [PMID: 36768281 PMCID: PMC9916855 DOI: 10.3390/ijms24031957] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 01/20/2023] Open
Abstract
Nerve growth factor (NGF) was the first neurotrophin described. This neurotrophin contributes to organogenesis by promoting sensory innervation and angiogenesis in the endocrine and immune systems. Neuronal and non-neuronal cells produce and secrete NGF, and several cell types throughout the body express the high-affinity neurotrophin receptor TrkA and the low-affinity receptor p75NTR. NGF is essential for glucose-stimulated insulin secretion and the complete development of pancreatic islets. Plus, this factor is involved in regulating lipolysis and thermogenesis in adipose tissue. Immune cells produce and respond to NGF, modulating their inflammatory phenotype and the secretion of cytokines, contributing to insulin resistance and metabolic homeostasis. This neurotrophin regulates the synthesis of gonadal steroid hormones, which ultimately participate in the metabolic homeostasis of other tissues. Therefore, we propose that this neurotrophin's imbalance in concentrations and signaling during metabolic syndrome contribute to its pathophysiology. In the present work, we describe the multiple roles of NGF in immunoendocrine organs that are important in metabolic homeostasis and related to the pathophysiology of metabolic syndrome.
Collapse
|
24
|
Simmons DA, Belichenko NP, Longo FM. Pharmacological Co-Activation of TrkB and TrkC Receptor Signaling Ameliorates Striatal Neuropathology and Motor Deficits in Mouse Models of Huntington's Disease. J Huntingtons Dis 2023; 12:215-239. [PMID: 37638447 DOI: 10.3233/jhd-230589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
BACKGROUND Loss of neurotrophic support in the striatum, particularly reduced brain-derived neurotrophic factor (BDNF) levels, contributes importantly to Huntington's disease (HD) pathogenesis. Another neurotrophin (NT), NT-3, is reduced in the cortex of HD patients; however, its role in HD is unknown. BDNF and NT-3 bind with high affinity to the tropomyosin receptor-kinases (Trk) B and TrkC, respectively. Targeting TrkB/TrkC may be an effective HD therapeutic strategy, as multiple links exist between their signaling pathways and HD degenerative mechanisms. We developed a small molecule ligand, LM22B-10, that activates TrkB and TrkC to promote cell survival. OBJECTIVE This study aimed to determine if upregulating TrkB/TrkC signaling with LM22B-10 would alleviate the HD phenotype in R6/2 and Q140 mice. METHODS LM22B-10 was delivered by concomitant intranasal-intraperitoneal routes to R6/2 and Q140 mice and then motor performance and striatal pathology were evaluated. RESULTS NT-3 levels, TrkB/TrkC phosphorylation, and AKT signaling were reduced in the R6/2 striatum; LM22B-10 counteracted these deficits. LM22B-10 also reduced intranuclear huntingtin aggregates, dendritic spine loss, microglial activation, and degeneration of dopamine- and cyclic AMP-regulated phosphoprotein with a molecular weight of 32 kDa (DARPP-32) and parvalbumin-containing neurons in the R6/2 and/or Q140 striatum. Moreover, both HD mouse models showed improved motor performance after LM22B-10 treatment. CONCLUSIONS These results reveal an NT-3/TrkC signaling deficiency in the striatum of R6/2 mice, support the idea that targeting TrkB/TrkC alleviates HD-related neurodegeneration and motor dysfunction, and suggest a novel, disease-modifying, multi-target strategy for treating HD.
Collapse
Affiliation(s)
- Danielle A Simmons
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Nadia P Belichenko
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
25
|
NGF regulates sertoli cell growth and prevents LPS-induced junction protein damage via PI3K/AKT/NFκB signaling. Theriogenology 2023; 195:138-148. [DOI: 10.1016/j.theriogenology.2022.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/22/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022]
|
26
|
Qian L, Rawashdeh O, Kasas L, Milne MR, Garner N, Sankorrakul K, Marks N, Dean MW, Kim PR, Sharma A, Bellingham MC, Coulson EJ. Cholinergic basal forebrain degeneration due to sleep-disordered breathing exacerbates pathology in a mouse model of Alzheimer's disease. Nat Commun 2022; 13:6543. [PMID: 36323689 PMCID: PMC9630433 DOI: 10.1038/s41467-022-33624-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 09/26/2022] [Indexed: 11/07/2022] Open
Abstract
Although epidemiological studies indicate that sleep-disordered breathing (SDB) such as obstructive sleep apnea is a strong risk factor for the development of Alzheimer's disease (AD), the mechanisms of the risk remain unclear. Here we developed a method of modeling SDB in mice that replicates key features of the human condition: altered breathing during sleep, sleep disruption, moderate hypoxemia, and cognitive impairment. When we induced SDB in a familial AD model, the mice displayed exacerbation of cognitive impairment and the pathological features of AD, including increased levels of amyloid-beta and inflammatory markers, as well as selective degeneration of cholinergic basal forebrain neurons. These pathological features were not induced by chronic hypoxia or sleep disruption alone. Our results also revealed that the cholinergic neurodegeneration was mediated by the accumulation of nuclear hypoxia inducible factor 1 alpha. Furthermore, restoring blood oxygen levels during sleep to prevent hypoxia prevented the pathological changes induced by the SDB. These findings suggest a signaling mechanism whereby SDB induces cholinergic basal forebrain degeneration.
Collapse
Affiliation(s)
- Lei Qian
- grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072 Australia ,grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072 Australia ,grid.1003.20000 0000 9320 7537School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072 Australia
| | - Oliver Rawashdeh
- grid.1003.20000 0000 9320 7537School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072 Australia
| | - Leda Kasas
- grid.1003.20000 0000 9320 7537School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072 Australia
| | - Michael R. Milne
- grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072 Australia ,grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072 Australia ,grid.1003.20000 0000 9320 7537School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072 Australia
| | - Nicholas Garner
- grid.1003.20000 0000 9320 7537School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072 Australia
| | - Kornraviya Sankorrakul
- grid.1003.20000 0000 9320 7537School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072 Australia ,grid.10223.320000 0004 1937 0490Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Thailand
| | - Nicola Marks
- grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072 Australia
| | - Matthew W. Dean
- grid.1003.20000 0000 9320 7537School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072 Australia
| | - Pu Reum Kim
- grid.1003.20000 0000 9320 7537School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072 Australia
| | - Aanchal Sharma
- grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072 Australia
| | - Mark C. Bellingham
- grid.1003.20000 0000 9320 7537School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072 Australia
| | - Elizabeth J. Coulson
- grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072 Australia ,grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072 Australia ,grid.1003.20000 0000 9320 7537School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072 Australia
| |
Collapse
|
27
|
Xiong LL, Chen L, Deng IB, Zhou XF, Wang TH. P75 neurotrophin receptor as a therapeutic target for drug development to treat neurological diseases. Eur J Neurosci 2022; 56:5299-5318. [PMID: 36017737 DOI: 10.1111/ejn.15810] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/11/2022] [Accepted: 08/23/2022] [Indexed: 12/14/2022]
Abstract
The interaction of neurotrophins with their receptors is involved in the pathogenesis and progression of various neurological diseases, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, spinal cord injury and acute and chronic cerebral damage. The p75 neurotrophin receptor (p75NTR) plays a pivotal role in the development of neurological dysfunctions as a result of its high expression, abnormal processing and signalling. Therefore, p75NTR represents as a vital therapeutic target for the treatment of neurodegeneration, neuropsychiatric disorders and cerebrovascular insufficiency. This review summarizes the current research progress on the p75NTR signalling in neurological deficits. We also summarize the present therapeutic approaches by genetically and pharmacologically targeting p75NTR for the attenuation of pathological changes. Based on the evolving knowledge, the role of p75NTR in the regulation of tau hyperphosphorylation, Aβ metabolism, the degeneration of motor neurons and dopaminergic neurons has been discussed. Its position as a biomarker to evaluate the severity of diseases and as a druggable target for drug development has also been elucidated. Several prototype small molecule compounds were introduced to be crucial in neuronal survival and functional recovery via targeting p75NTR. These small molecule compounds represent desirable agents in attenuating neurodegeneration and cell death as they abolish activation-induced neurotoxicity of neurotrophins via modulating p75NTR signalling. More comprehensive and in-depth investigations on p75NTR-based drug development are required to shed light on effective treatment of numerous neurological disorders.
Collapse
Affiliation(s)
- Liu-Lin Xiong
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, China.,Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia.,Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Li Chen
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Isaac Bul Deng
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Xin-Fu Zhou
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Ting-Hua Wang
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
28
|
Povarnina PY, Volkova AA, Vorontsova ON, Kamensky AA, Gudasheva TA, Seredenin SB. A Low-Molecular-Weight BDNF Mimetic, Dipeptide GSB-214, Prevents Memory Impairment in Rat Models of Alzheimer's Disease. Acta Naturae 2022; 14:94-100. [PMID: 36694902 PMCID: PMC9844091 DOI: 10.32607/actanaturae.11755] [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: 06/16/2022] [Accepted: 09/30/2022] [Indexed: 01/22/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is known to be involved in the pathogenesis of Alzheimer's disease (AD). However, the pharmacological use of full-length neurotrophin is limited, because of its macromolecular protein nature. A dimeric dipeptide mimetic of the BDNF loop 1, bis-(N-monosuccinyl-L-methionyl-L-serine) heptamethylene diamide (GSB-214), was designed at the Zakusov Research Institute of Pharmacology. GSB-214 activates TrkB, PI3K/AKT, and PLC-γ1 in vitro. GSB-214 exhibited a neuroprotective activity during middle cerebral artery occlusion in rats when administered intraperitoneally (i.p.) at a dose of 0.1 mg/kg and improved memory in the novel object recognition test (0.1 and 1.0 mg/kg, i.p.). In the present study, we investigated the effects of GSB-214 on memory in the scopolamine- and steptozotocin-induced AD models, with reference to activation of TrkB receptors. AD was modeled in rats using a chronic i.p. scopolamine injection or a single streptozotocin injection into the cerebral ventricles. GSB-214 was administered within 10 days after the exposure to scopolamine at doses of 0.05, 0.1, and 1 mg/kg (i.p.) or within 14 days after the exposure to streptozotocin at a dose of 0.1 mg/kg (i.p.). The effect of the dipeptide was evaluated in the novel object recognition test; K252A, a selective inhibitor of tyrosine kinase receptors, was used to reveal a dependence between the mnemotropic action and Trk receptors. GSB-214 at doses of 0.05 and 0.1 mg/kg statistically significantly prevented scopolamine-induced long-term memory impairment, while not affecting short-term memory. In the streptozotocin-induced model, GSB-214 completely eliminated the impairment of short-term memory. No mnemotropic effect of GSB-214 was registered when Trk receptors were inhibited by K252A.
Collapse
Affiliation(s)
- P Yu Povarnina
- Research Zakusov Institute of Pharmacology, Moscow, 125315 Russia
| | - A A Volkova
- Research Zakusov Institute of Pharmacology, Moscow, 125315 Russia
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991 Russia
| | - O N Vorontsova
- Research Zakusov Institute of Pharmacology, Moscow, 125315 Russia
| | - A A Kamensky
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991 Russia
| | - T A Gudasheva
- Research Zakusov Institute of Pharmacology, Moscow, 125315 Russia
| | - S B Seredenin
- Research Zakusov Institute of Pharmacology, Moscow, 125315 Russia
| |
Collapse
|
29
|
Szarowicz CA, Steece-Collier K, Caulfield ME. New Frontiers in Neurodegeneration and Regeneration Associated with Brain-Derived Neurotrophic Factor and the rs6265 Single Nucleotide Polymorphism. Int J Mol Sci 2022; 23:ijms23148011. [PMID: 35887357 PMCID: PMC9319713 DOI: 10.3390/ijms23148011] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/20/2022] Open
Abstract
Brain-derived neurotrophic factor is an extensively studied neurotrophin implicated in the pathology of multiple neurodegenerative and psychiatric disorders including, but not limited to, Parkinson’s disease, Alzheimer’s disease, Huntington’s disease, traumatic brain injury, major de-pressive disorder, and schizophrenia. Here we provide a brief summary of current knowledge on the role of BDNF and the common human single nucleotide polymorphism, rs6265, in driving the pathogenesis and rehabilitation in these disorders, as well as the status of BDNF-targeted therapies. A common trend has emerged correlating low BDNF levels, either detected within the central nervous system or peripherally, to disease states, suggesting that BDNF replacement therapies may hold clinical promise. In addition, we introduce evidence for a distinct role of the BDNF pro-peptide as a biologically active ligand and the need for continuing studies on its neurological function outside of that as a molecular chaperone. Finally, we highlight the latest research describing the role of rs6265 expression in mechanisms of neurodegeneration as well as paradoxical advances in the understanding of this genetic variant in neuroregeneration. All of this is discussed in the context of personalized medicine, acknowledging there is no “one size fits all” therapy for neurodegenerative or psychiatric disorders and that continued study of the multiple BDNF isoforms and genetic variants represents an avenue for discovery ripe with therapeutic potential.
Collapse
Affiliation(s)
- Carlye A. Szarowicz
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (C.A.S.); (K.S.-C.)
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (C.A.S.); (K.S.-C.)
| | - Margaret E. Caulfield
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (C.A.S.); (K.S.-C.)
- Correspondence: ; Tel.: +1-616-234-0969; Fax: +1- 616-234-0991
| |
Collapse
|
30
|
Suppression of HIV-associated Macrophage Activation by a p75 Neurotrophin Receptor Ligand. J Neuroimmune Pharmacol 2022; 17:242-260. [PMID: 34296391 PMCID: PMC9386897 DOI: 10.1007/s11481-021-10002-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/22/2021] [Indexed: 12/29/2022]
Abstract
Previous studies indicated that nerve growth factor (NGF) and proNGF differentially regulate the phenotype of macrophages and microglia via actions at tropomyosin receptor kinase A (TrkA) and p75 neurotrophin receptors (p75NTR), respectively. The ability of HIV gp120 and virions to induce the secretion of factors toxic to neurons was suppressed by NGF and enhanced by proNGF, suggesting the potential for neurotrophin based "anti-inflammatory" interventions. To investigate the "anti-inflammatory" potential of the p75NTR ligand, LM11A-31, we treated cultured macrophages and microglia with HIV gp120 in the presence or absence of the ligand and evaluated the morphological phenotype, intrinsic calcium signaling, neurotoxic activity and proteins in the secretome. LM11A-31 at 10 nM was able to suppress the release of neurotoxic factors from both monocyte-derived macrophages (MDM) and microglia. The protective effects correlated with a shift in morphology and a unique secretory phenotype rich in growth factors that overrode the actions of HIV gp120. The protein pattern was generally consistent with anti-inflammatory, phagocytic and tissue remodeling functions. Although the toxic factor(s) and the source of the neuroprotection were not identified, the data indicated that an increased degradation of NGF induced by HIV gp120 was likely to contribute to neuronal vulnerability. Although substantial work is still needed to reveal the functions of many proteins in the mononuclear phagocyte secretome, such as growth and differentiation factors, the data clearly indicate that the ligand LM11A-31 has excellent therapeutic potential due to its ability to induce a more protective phenotype that restricts activation by HIV.
Collapse
|
31
|
Triaca V, Imbimbo BP, Nisticò R. Editorial: Neurotrophins Biodelivery to CNS: Innovative Approaches for Disease-Modifying Therapy. Front Neurosci 2022; 16:916563. [PMID: 35620662 PMCID: PMC9128590 DOI: 10.3389/fnins.2022.916563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Viviana Triaca
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
- *Correspondence: Viviana Triaca
| | | | - Robert Nisticò
- Department of Biology, School of Pharmacy, University of Tor Vergata, Rome, Italy
- Laboratory of Pharmacology of Synaptic Plasticity, Fondazione EBRI Rita Levi Montalcini, Rome, Italy
| |
Collapse
|
32
|
Hany R, Leyris JP, Bret G, Mallié S, Sar C, Thouaye M, Hamze A, Provot O, Sokoloff P, Valmier J, Villa P, Rognan D. High-Throughput Screening for Extracellular Inhibitors of the FLT3 Receptor Tyrosine Kinase Reveals Chemically Diverse and Druggable Negative Allosteric Modulators. ACS Chem Biol 2022; 17:709-722. [PMID: 35227060 DOI: 10.1021/acschembio.2c00048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Inhibiting receptor tyrosine kinases is commonly achieved by two main strategies targeting either the intracellular kinase domain by low molecular weight compounds or the extracellular ligand-binding domain by monoclonal antibodies. Identifying small molecules able to inhibit RTKs at the extracellular level would be highly desirable to gain exquisite selectivity but is believed to be challenging owing to the size of RTK endogenous ligands (cytokines, growth factors) and the topology of RTK extracellular domains. We here report the high-throughput screening of the French Chemical Library (48K compounds) for extracellular inhibitors of the Fms-like tyrosine kinase 3 (FLT3) receptor tyrosine kinase, by a homogeneous time-resolved fluorescence competition assay. A total of 679 small molecular weight ligands (1.4%) were confirmed to strongly inhibit (>75%) the binding of the fluorescent labeled FLT3 ligand (FL cytokine) to FLT3 overexpressed in HEK-293 cells, at two different concentrations (5 and 20 μM). Concentration-response curves, obtained for 111 lead-like molecules, confirmed the unexpected tolerance of the FLT3 extracellular domain for low molecular weight druggable inhibitors exhibiting submicromolar potencies, chemical diversity, and promising pharmacokinetic properties. Further investigation of one hit confirmed inhibitory properties in dorsal root ganglia neurons and in a mouse model of neuropathic pain.
Collapse
Affiliation(s)
- Romain Hany
- Plate-forme de Chimie Biologique Intégrative de Strasbourg (PCBIS), UAR3286 CNRS-Université de Strasbourg, Institut du Médicament de Strasbourg, ESBS Pôle API, Bld Sébastien Brant, 67412 Illkirch Cedex, France
| | - Jean-Philippe Leyris
- Institut des Neurosciences de Montpellier (INM), INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, 34000 Montpellier, France
- Université de Montpellier, 34000 Montpellier, France
- BIODOL Therapeutics, CAP Alpha, 34830 Clapiers, France
| | - Guillaume Bret
- Laboratoire d’Innovation Thérapeutique (LIT), UMR7200 CNRS-Université de Strasbourg, 67400 Illkirch, France
| | - Sylvie Mallié
- Institut des Neurosciences de Montpellier (INM), INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, 34000 Montpellier, France
- Université de Montpellier, 34000 Montpellier, France
| | - Chamroeun Sar
- Institut des Neurosciences de Montpellier (INM), INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, 34000 Montpellier, France
- Université de Montpellier, 34000 Montpellier, France
| | - Maxime Thouaye
- Institut des Neurosciences de Montpellier (INM), INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, 34000 Montpellier, France
- Université de Montpellier, 34000 Montpellier, France
| | - Abdallah Hamze
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France
| | - Olivier Provot
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France
| | | | - Jean Valmier
- Institut des Neurosciences de Montpellier (INM), INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, 34000 Montpellier, France
- Université de Montpellier, 34000 Montpellier, France
| | - Pascal Villa
- Plate-forme de Chimie Biologique Intégrative de Strasbourg (PCBIS), UAR3286 CNRS-Université de Strasbourg, Institut du Médicament de Strasbourg, ESBS Pôle API, Bld Sébastien Brant, 67412 Illkirch Cedex, France
| | - Didier Rognan
- Laboratoire d’Innovation Thérapeutique (LIT), UMR7200 CNRS-Université de Strasbourg, 67400 Illkirch, France
| |
Collapse
|
33
|
Gohil K, Kazmi MZH, Williams FJ. Structure-activity relationship and bioactivity studies of neurotrophic trans-banglene. Org Biomol Chem 2022; 20:2187-2193. [PMID: 35229853 DOI: 10.1039/d2ob00016d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
The synthesis and bioactivity of neurotrophic banglenes and derivatives is described, establishing a structure-activity relationship which enables future mechanistic studies. Neuritogenesis assays indicate that (-) trans-banglene is the active enantiomer. Assays performed with and without NGF protein suggest that neurotrophic activity and potentiation of NGF activity by (-) trans-banglene might be distinct unassociated processes. Interestingly, (-) trans-banglene potentiation of NGF-induced neuritogenesis is unaffected by the presence of Erk1/2, Akt and Pkc inhibitors.
Collapse
Affiliation(s)
- Khyati Gohil
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - M Zain H Kazmi
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | | |
Collapse
|
34
|
ENT-A010, a Novel Steroid Derivative, Displays Neuroprotective Functions and Modulates Microglial Responses. Biomolecules 2022; 12:biom12030424. [PMID: 35327616 PMCID: PMC8946810 DOI: 10.3390/biom12030424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/02/2022] [Accepted: 03/05/2022] [Indexed: 11/17/2022] Open
Abstract
Tackling neurodegeneration and neuroinflammation is particularly challenging due to the complexity of central nervous system (CNS) disorders, as well as the limited drug accessibility to the brain. The activation of tropomyosin-related kinase A (TRKA) receptor signaling by the nerve growth factor (NGF) or the neurosteroid dehydroepiandrosterone (DHEA) may combat neurodegeneration and regulate microglial function. In the present study, we synthesized a C-17-spiro-cyclopropyl DHEA derivative (ENT-A010), which was capable of activating TRKA. ENT-A010 protected PC12 cells against serum starvation-induced cell death, dorsal root ganglia (DRG) neurons against NGF deprivation-induced apoptosis and hippocampal neurons against Aβ-induced apoptosis. In addition, ENT-A010 pretreatment partially restored homeostatic features of microglia in the hippocampus of lipopolysaccharide (LPS)-treated mice, enhanced Aβ phagocytosis, and increased Ngf expression in microglia in vitro. In conclusion, the small molecule ENT-A010 elicited neuroprotective effects and modulated microglial function, thereby emerging as an interesting compound, which merits further study in the treatment of CNS disorders.
Collapse
|
35
|
Rogdakis T, Charou D, Latorrata A, Papadimitriou E, Tsengenes A, Athanasiou C, Papadopoulou M, Chalikiopoulou C, Katsila T, Ramos I, Prousis KC, Wade RC, Sidiropoulou K, Calogeropoulou T, Gravanis A, Charalampopoulos I. Development and Biological Characterization of a Novel Selective TrkA Agonist with Neuroprotective Properties against Amyloid Toxicity. Biomedicines 2022; 10:614. [PMID: 35327415 PMCID: PMC8945229 DOI: 10.3390/biomedicines10030614] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/23/2022] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Neurotrophins are growth factors that exert important neuroprotective effects by preventing neuronal death and synaptic loss. Nerve Growth Factor (NGF) acts through the activation of its high-affinity, pro-survival TrkA and low-affinity, pro-apoptotic p75NTR receptors. NGF has been shown to slow or prevent neurodegenerative signals in Alzheimer's Disease (AD) progression. However, its low bioavailability and its blood-brain-barrier impermeability limit the use of NGF as a potential therapeutic agent against AD. Based on our previous findings on synthetic dehydroepiandrosterone derivatives, we identified a novel NGF mimetic, named ENT-A013, which selectively activates TrkA and exerts neuroprotective, anti-amyloid-β actions. We now report the chemical synthesis, in silico modelling, metabolic stability, CYP-mediated reaction phenotyping and biological characterization of ENT-A013 under physiological and neurodegenerative conditions. We show that ENT-A013 selectively activates the TrkA receptor and its downstream kinases Akt and Erk1/2 in PC12 cells, protecting these cells from serum deprivation-induced cell death. Moreover, ENT-A013 promotes survival of primary Dorsal Root Ganglion (DRG) neurons upon NGF withdrawal and protects hippocampal neurons against Amyloid β-induced apoptosis and synaptic loss. Furthermore, this neurotrophin mimetic partially restores LTP impairment. In conclusion, ENT-A013 represents a promising new lead molecule for developing therapeutics against neurodegenerative disorders, such as Alzheimer's Disease, selectively targeting TrkA-mediated pro-survival signals.
Collapse
Affiliation(s)
- Thanasis Rogdakis
- Department of Pharmacology, Medical School, University of Crete, 71003 Heraklion, Greece; (T.R.); (D.C.); (E.P.); (M.P.); (A.G.)
- Foundation for Research & Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology & Biotechnology, 70013 Heraklion, Greece;
| | - Despoina Charou
- Department of Pharmacology, Medical School, University of Crete, 71003 Heraklion, Greece; (T.R.); (D.C.); (E.P.); (M.P.); (A.G.)
- Foundation for Research & Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology & Biotechnology, 70013 Heraklion, Greece;
| | - Alessia Latorrata
- National Hellenic Research Foundation, Institute of Chemical Biology, 11635 Athens, Greece; (A.L.); (C.C.); (T.K.); (K.C.P.); (T.C.)
| | - Eleni Papadimitriou
- Department of Pharmacology, Medical School, University of Crete, 71003 Heraklion, Greece; (T.R.); (D.C.); (E.P.); (M.P.); (A.G.)
- Foundation for Research & Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology & Biotechnology, 70013 Heraklion, Greece;
| | - Alexandros Tsengenes
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), 69118 Heidelberg, Germany; (A.T.); (C.A.); (R.C.W.)
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
- Heidelberg Biosciences International Graduate School, Heidelberg University, 69120 Heidelberg, Germany
| | - Christina Athanasiou
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), 69118 Heidelberg, Germany; (A.T.); (C.A.); (R.C.W.)
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
- Heidelberg Biosciences International Graduate School, Heidelberg University, 69120 Heidelberg, Germany
| | - Marianna Papadopoulou
- Department of Pharmacology, Medical School, University of Crete, 71003 Heraklion, Greece; (T.R.); (D.C.); (E.P.); (M.P.); (A.G.)
- Foundation for Research & Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology & Biotechnology, 70013 Heraklion, Greece;
| | - Constantina Chalikiopoulou
- National Hellenic Research Foundation, Institute of Chemical Biology, 11635 Athens, Greece; (A.L.); (C.C.); (T.K.); (K.C.P.); (T.C.)
| | - Theodora Katsila
- National Hellenic Research Foundation, Institute of Chemical Biology, 11635 Athens, Greece; (A.L.); (C.C.); (T.K.); (K.C.P.); (T.C.)
| | - Isbaal Ramos
- Innovative Technologies in Biological Systems SL (INNOPROT), 48160 Bizkaia, Spain;
| | - Kyriakos C. Prousis
- National Hellenic Research Foundation, Institute of Chemical Biology, 11635 Athens, Greece; (A.L.); (C.C.); (T.K.); (K.C.P.); (T.C.)
| | - Rebecca C. Wade
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), 69118 Heidelberg, Germany; (A.T.); (C.A.); (R.C.W.)
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, 69120 Heidelberg, Germany
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, 69120 Heidelberg, Germany
| | - Kyriaki Sidiropoulou
- Foundation for Research & Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology & Biotechnology, 70013 Heraklion, Greece;
- Department of Biology, University of Crete, 71113 Heraklion, Greece
| | - Theodora Calogeropoulou
- National Hellenic Research Foundation, Institute of Chemical Biology, 11635 Athens, Greece; (A.L.); (C.C.); (T.K.); (K.C.P.); (T.C.)
| | - Achille Gravanis
- Department of Pharmacology, Medical School, University of Crete, 71003 Heraklion, Greece; (T.R.); (D.C.); (E.P.); (M.P.); (A.G.)
- Foundation for Research & Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology & Biotechnology, 70013 Heraklion, Greece;
| | - Ioannis Charalampopoulos
- Department of Pharmacology, Medical School, University of Crete, 71003 Heraklion, Greece; (T.R.); (D.C.); (E.P.); (M.P.); (A.G.)
- Foundation for Research & Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology & Biotechnology, 70013 Heraklion, Greece;
| |
Collapse
|
36
|
Mezhlumyan AG, Tallerova AV, Povarnina PY, Tarasiuk AV, Sazonova NM, Gudasheva TA, Seredenin SB. Antidepressant-like Effects of BDNF and NGF Individual Loop Dipeptide Mimetics Depend on the Signal Transmission Patterns Associated with Trk. Pharmaceuticals (Basel) 2022; 15:ph15030284. [PMID: 35337082 PMCID: PMC8950955 DOI: 10.3390/ph15030284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 11/29/2022] Open
Abstract
Neurotrophins are considered as an attractive target for the development of antidepressants with a novel mechanism of action. Previously, the dimeric dipeptide mimetics of individual loops of nerve growth factor, NGF (GK-6, loop 1; GK-2, loop 4) and brain-derived neurotrophic factor, BDNF (GSB-214, loop 1; GTS-201, loop 2; GSB-106, loop 4) were designed and synthesized. All the mimetics of NGF and BDNF in vitro after a 5–180 min incubation in a HT-22 cell culture were able to phosphorylate the tropomyosin-related kinase A (TrkA) or B (TrkB) receptors, respectively, but had different post-receptor signaling patterns. In the present study, we conduct comparative research of the antidepressant-like activity of these mimetics at acute and subchronic administration in the forced swim test in mice. Only the dipeptide GSB-106 that in vitro activates mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK), phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) and phospholipase C-gamma (PLCγ) post-receptor pathways exhibited antidepressant-like activity (0.1 and 1.0 mg/kg, ip) at acute administration. At the same time, the inhibition of any one of these signaling pathways completely prevented the antidepressant-like effects of GSB-106 in the forced swim test. All the NGF mimetics were inactive after a single injection regardless of post-receptor in vitro signaling patterns. All the investigated dipeptides, except GTS-201, not activating PI3K/AKT in vitro unlike the other compounds, were active at subchronic administration. The data obtained demonstrate that the low-molecular weight BDNF mimetic GSB-106 that activates all three main post-receptor TrkB signaling pathways is the most promising for the development as an antidepressant.
Collapse
Affiliation(s)
- Armen G. Mezhlumyan
- Department of Medicinal Chemistry, V.V. Zakusov Research Institute of Pharmacology, 125315 Moscow, Russia; (A.G.M.); (A.V.T.); (P.Y.P.); (A.V.T.); (N.M.S.)
| | - Anna V. Tallerova
- Department of Medicinal Chemistry, V.V. Zakusov Research Institute of Pharmacology, 125315 Moscow, Russia; (A.G.M.); (A.V.T.); (P.Y.P.); (A.V.T.); (N.M.S.)
| | - Polina Y. Povarnina
- Department of Medicinal Chemistry, V.V. Zakusov Research Institute of Pharmacology, 125315 Moscow, Russia; (A.G.M.); (A.V.T.); (P.Y.P.); (A.V.T.); (N.M.S.)
| | - Aleksey V. Tarasiuk
- Department of Medicinal Chemistry, V.V. Zakusov Research Institute of Pharmacology, 125315 Moscow, Russia; (A.G.M.); (A.V.T.); (P.Y.P.); (A.V.T.); (N.M.S.)
| | - Nellya M. Sazonova
- Department of Medicinal Chemistry, V.V. Zakusov Research Institute of Pharmacology, 125315 Moscow, Russia; (A.G.M.); (A.V.T.); (P.Y.P.); (A.V.T.); (N.M.S.)
| | - Tatiana A. Gudasheva
- Department of Medicinal Chemistry, V.V. Zakusov Research Institute of Pharmacology, 125315 Moscow, Russia; (A.G.M.); (A.V.T.); (P.Y.P.); (A.V.T.); (N.M.S.)
- Correspondence:
| | - Sergey B. Seredenin
- Department of Pharmacogenetics, V.V. Zakusov Research Institute of Pharmacology, 25315 Moscow, Russia;
| |
Collapse
|
37
|
Fleury S, Schnitzer ME, Ledoux-Hutchinson L, Boukhatem I, Bélanger JC, Welman M, Busseuil D, Tardif JC, D’Antono B, Lordkipanidzé M. Clinical Correlates Identify ProBDNF and Thrombo-Inflammatory Markers as Key Predictors of Circulating p75NTR Extracellular Domain Levels in Older Adults. Front Aging Neurosci 2022; 14:821865. [PMID: 35264944 PMCID: PMC8899540 DOI: 10.3389/fnagi.2022.821865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
The p75NTR receptor binds all neurotrophins and is mostly known for its role in neuronal survival and apoptosis. Recently, the extracellular domain (ECD) of p75NTR has been reported in plasma, its levels being dysregulated in numerous neurological diseases. However, the factors associated with p75NTR ECD levels remain unknown. We investigated clinical correlates of plasma p75NTR ECD levels in older adults without clinically manifested neurological disorders. Circulating p75NTR levels were measured by enzyme-linked immunosorbent assay in plasma obtained from participants in the BEL-AGE cohort (n = 1,280). Determinants of plasma p75NTR ECD levels were explored using linear and non-linear statistical models. Plasma p75NTR ECD levels were higher in male participants; were positively correlated with circulating concentrations of pro-brain-derived neurotrophic factor, and inflammatory markers interleukin-6 and CD40 Ligand; and were negatively correlated with the platelet activation marker P-selectin. While most individuals had p75NTR levels ranging from 43 to 358 pg/ml, high p75NTR levels reaching up to 9,000 pg/ml were detectable in a subgroup representing 15% of the individuals studied. In this cohort of older adults without clinically manifested neurological disorders, there was no association between plasma p75NTR ECD levels and cognitive performance, as assessed by the Montreal Cognitive Assessment score. The physiological relevance of high p75NTR ECD levels in plasma warrants further investigation. Further research assessing the source of circulating p75NTR is needed for a deeper understanding of the direction of effect, and to investigate whether high p75NTR ECD levels are predictive biomarkers or consequences of neuropathology.
Collapse
Affiliation(s)
- Samuel Fleury
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
| | - Mireille E. Schnitzer
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
- Department of Social and Preventive Medicine, School of Public Health, Université de Montréal, Montreal, QC, Canada
| | | | - Imane Boukhatem
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
| | - Jean-Christophe Bélanger
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
| | - Mélanie Welman
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
| | - David Busseuil
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
| | - Jean-Claude Tardif
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Medicine, Montreal Heart Institute, Montreal, QC, Canada
| | - Bianca D’Antono
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
- Department of Psychology, Faculty of Arts and Sciences, Université de Montréal, Montreal, QC, Canada
- *Correspondence: Bianca D’Antono,
| | - Marie Lordkipanidzé
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
- Marie Lordkipanidzé,
| |
Collapse
|
38
|
Li T, Li X, Huang X, Yu H, Li S, Zhang Z, Xie Y, Song X, Liu J, Yang X, Liu G. Mitochondriomics reveals the underlying neuroprotective mechanism of TrkB receptor agonist R13 in the 5×FAD mice. Neuropharmacology 2022; 204:108899. [PMID: 34838815 DOI: 10.1016/j.neuropharm.2021.108899] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023]
Abstract
Decreased energy metabolism and mitochondrial biogenesis defects are implicated in the pathogenesis of Alzheimer's disease (AD). In present study, mitochondriomics analysis revealed significant effects of R13, a prodrug of 7,8-dihydroxyflavone, on mitochondrial protein expression profile, including the proteins related to the biological processes: fatty acid beta-oxidation, fatty acid metabolic process, mitochondrial electron transport, and mitochondrial respiratory chain. Cluster analysis demonstrated that R13 promoted mitochondrial oxidative phosphorylation (OXPHOS). The functional analysis showed that R13 increased ATP levels and enhanced OXPHOS including complex Ⅰ, Ⅱ, Ⅲ and Ⅳ. R13 treatment increased mitochondrial biogenesis by regulating the levels of p-AMPKα, p-CREB, PGC-1α, NRF1 and TFAM as a consequence of activation of TrkB receptor in the 5 × FAD mice. Finally, R13 significantly reduced the levels of tau phosphorylation and Aβ plaque. Our data suggest that R13 may be used for treating AD via enhancing mitochondrial biogenesis and metabolism.
Collapse
Affiliation(s)
- Ting Li
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Li
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China; Department of Pathology, Wuhan No. 1 Hospital, Wuhan, 430022, China
| | - Xi Huang
- Department of Neurology,The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China; The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Hao Yu
- Shenzhen Polytechnic, Shenzhen, 518055, China
| | - Shupeng Li
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Zaijun Zhang
- Institute of New Drug Research and Guangzhou, Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Yongmei Xie
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Xiangrong Song
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jianjun Liu
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China.
| | - Gongping Liu
- Department of Pathology, Wuhan No. 1 Hospital, Wuhan, 430022, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, China.
| |
Collapse
|
39
|
Chronic partial TrkB activation reduces seizures and mortality in a mouse model of Dravet syndrome. Proc Natl Acad Sci U S A 2022; 119:2022726119. [PMID: 35165147 PMCID: PMC8851461 DOI: 10.1073/pnas.2022726119] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2021] [Indexed: 12/03/2022] Open
Abstract
Dravet syndrome (DS) is a severe childhood epileptic encephalopathy characterized by intractable seizures and comorbidities, including a high rate of premature mortality. DS is mainly caused by loss-of-function mutations of the Scn1a gene encoding sodium channel Nav1.1 that is predominantly expressed in inhibitory parvalbumin-containing (PV) interneurons. Decreased Nav1.1 impairs PV cell function, causing DS phenotypes. Effective pharmacological therapy targeting defective PV interneurons is currently not available. This study demonstrated that early treatment with a partial TrkB receptor agonist, LM22A-4, increased Nav1.1 expression, improved PV interneuron function, and reduced seizure occurrence and mortality rate in DS mice, suggesting a potential therapy for DS. Dravet syndrome (DS) is one of the most severe childhood epilepsies, characterized by intractable seizures and comorbidities including cognitive and social dysfunction and high premature mortality. DS is mainly caused by loss-of-function mutations in the Scn1a gene encoding Nav1.1 that is predominantly expressed in inhibitory parvalbumin-containing (PV) interneurons. Decreased Nav1.1 impairs PV cell function, contributing to DS phenotypes. Effective pharmacological therapy that targets defective PV interneurons is not available. The known role of brain-derived neurotrophic factor (BDNF) in the development and maintenance of interneurons, together with our previous results showing improved PV interneuronal function and antiepileptogenic effects of a TrkB receptor agonist in a posttraumatic epilepsy model, led to the hypothesis that early treatment with a TrkB receptor agonist might prevent or reduce seizure activity in DS mice. To test this hypothesis, we treated DS mice with LM22A-4 (LM), a partial agonist at the BDNF TrkB receptor, for 7 d starting at postnatal day 13 (P13), before the onset of spontaneous seizures. Results from immunohistochemistry, Western blot, whole-cell patch-clamp recording, and in vivo seizure monitoring showed that LM treatment increased the number of perisomatic PV interneuronal synapses around cortical pyramidal cells in layer V, upregulated Nav1.1 in PV neurons, increased inhibitory synaptic transmission, and decreased seizures and the mortality rate in DS mice. The results suggest that early treatment with a partial TrkB receptor agonist may be a promising therapeutic approach to enhance PV interneuron function and reduce epileptogenesis and premature death in DS.
Collapse
|
40
|
Nguyen TVV, Crumpacker RH, Calderon KE, Garcia FG, Zbesko JC, Frye JB, Gonzalez S, Becktel DA, Yang T, Tavera-Garcia MA, Morrison HW, Schnellmann RG, Longo FM, Doyle KP. Post-Stroke Administration of the p75 Neurotrophin Receptor Modulator, LM11A-31, Attenuates Chronic Changes in Brain Metabolism, Increases Neurotransmitter Levels, and Improves Recovery. J Pharmacol Exp Ther 2022; 380:126-141. [PMID: 34893553 PMCID: PMC11048261 DOI: 10.1124/jpet.121.000711] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 11/29/2021] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to test whether poststroke oral administration of a small molecule p75 neurotrophin receptor (p75NTR) modulator (LM11A-31) can augment neuronal survival and improve recovery in a mouse model of stroke. Mice were administered LM11A-31 for up to 12 weeks, beginning 1 week after stroke. Metabolomic analysis revealed that after 2 weeks of daily treatment, mice that received LM11A-31 were distinct from vehicle-treated mice by principal component analysis and had higher levels of serotonin, acetylcholine, and dopamine in their ipsilateral hemisphere. LM11A-31 treatment also improved redox homeostasis by restoring reduced glutathione. It also offset a stroke-induced reduction in glycolysis by increasing acetyl-CoA. There was no effect on cytokine levels in the infarct. At 13 weeks after stroke, adaptive immune cell infiltration in the infarct was unchanged in LM11A-31-treated mice, indicating that LM11A-31 does not alter the chronic inflammatory response to stroke at the site of the infarct. However, LM11A-31-treated mice had less brain atrophy, neurodegeneration, tau pathology, and microglial activation in other regions of the ipsilateral hemisphere. These findings correlated with improved recovery of motor function on a ladder test, improved sensorimotor and cognitive abilities on a nest construction test, and less impulsivity in an open field test. These data support small molecule modulation of the p75NTR for preserving neuronal health and function during stroke recovery. SIGNIFICANCE STATEMENT: The findings from this study introduce the p75 neurotrophin receptor as a novel small molecule target for promotion of stroke recovery. Given that LM11A-31 is in clinical trials as a potential therapy for Alzheimer's disease, it could be considered as a candidate for assessment in stroke or vascular dementia studies.
Collapse
Affiliation(s)
- Thuy-Vi V Nguyen
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Rachel H Crumpacker
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Kylie E Calderon
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Frankie G Garcia
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Jacob C Zbesko
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Jennifer B Frye
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Selena Gonzalez
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Danielle A Becktel
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Tao Yang
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Marco A Tavera-Garcia
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Helena W Morrison
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Rick G Schnellmann
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Frank M Longo
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Kristian P Doyle
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| |
Collapse
|
41
|
Semisynthesis and neurotrophic activity studies of novel neomajucin/majucin derivatives as neurotrophin small molecule mimetics. Bioorg Med Chem Lett 2022; 60:128580. [PMID: 35066142 DOI: 10.1016/j.bmcl.2022.128580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/02/2022] [Accepted: 01/16/2022] [Indexed: 11/20/2022]
Abstract
Majucin-type Illicium sesquiterpenes with potent neurotrophic activity are considered to be promising candidates for the treatment of various neurodegenerative disease. Owing to the low-abundance metabolites in Illicium genus, there are few studies on their structural modifications, structure-activity relationships, and pharmacophoric motif. Herein, structural modifications were conducted on the hydroxyl groups at C-3 and C-6 positions of two majucin-type compounds neomajucin (1) and majucin (2), and 39 neomajucin/majucin based esters were synthesized and evaluated for their neurite outgrowth-promoting activities. Among all the target derivatives, compounds 1a, 1j, 1r, 2b, 2d, 3a, 3b, 3d and 3h displayed more potent neurite outgrowth-promoting activity than their precursors. Some interesting structure-activity relationships (SARs) were also observed. Moreover, compound 1a showed good neuroprotective effect on MPP+-induced PC12 cell damage. Finally, compounds 1a and 3a exhibited relatively no cytotoxicity to normal human H9C2 cardiac cells. This work will shed light on the development of neomajucin/majucin derivatives as potential neurotrophic agents.
Collapse
|
42
|
Español P, Luna R, Soler C, Caruana P, Altés-Arranz A, Rodríguez F, Porta O, Sanchez O, Llurba E, Rovira R, Céspedes MV. Neural plasticity of the uterus: New targets for endometrial cancer? WOMEN'S HEALTH (LONDON, ENGLAND) 2022; 18:17455057221095537. [PMID: 35465787 PMCID: PMC9047769 DOI: 10.1177/17455057221095537] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Endometrial carcinoma is the most common gynecological malignancy in Western countries and is expected to increase in the following years because of the high index of obesity in the population. Recently, neural signaling has been recognized as part of the tumor microenvironment, playing an active role in tumor progression and invasion of different solid tumor types. The uterus stands out for the physiological plasticity of its peripheral nerves due to cyclic remodeling brought on by estrogen and progesterone hormones throughout the reproductive cycle. Therefore, a precise understanding of nerve-cancer crosstalk and the contribution of the organ-intrinsic neuroplasticity, mediated by estrogen and progesterone, of the uterine is urgently needed. The development of new and innovative medicines for patients with endometrial cancer would increase their quality of life and health. This review compiles information on the architecture and function of autonomous uterine neural innervations and the influence of hormone-dependent nerves in normal uterus and tumor progression. It also explores new therapeutic possibilities for endometrial cancer using these endocrine and neural advantages.
Collapse
Affiliation(s)
- Pia Español
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Rocio Luna
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Cristina Soler
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Pablo Caruana
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Amanda Altés-Arranz
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Francisco Rodríguez
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Oriol Porta
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Olga Sanchez
- Women and Perinatal Health Research Group, Obstetrics and Gynaecology Department, Hospital Sant Pau and Universitat Autònoma de Barcelona, Barcelona, Spain.,Maternal and Child Health and Development Network, Instituto Salud Carlos III, Madrid, Spain
| | - Elisa Llurba
- Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Women and Perinatal Health Research Group, Obstetrics and Gynaecology Department, Hospital Sant Pau and Universitat Autònoma de Barcelona, Barcelona, Spain.,Maternal and Child Health and Development Network, Instituto Salud Carlos III, Madrid, Spain
| | - Ramón Rovira
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - María Virtudes Céspedes
- Gynecology and Oncology Peritoneal Group, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| |
Collapse
|
43
|
Naletova I, Greco V, Sciuto S, Attanasio F, Rizzarelli E. Ionophore Ability of Carnosine and Its Trehalose Conjugate Assists Copper Signal in Triggering Brain-Derived Neurotrophic Factor and Vascular Endothelial Growth Factor Activation In Vitro. Int J Mol Sci 2021; 22:13504. [PMID: 34948299 PMCID: PMC8706131 DOI: 10.3390/ijms222413504] [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: 10/25/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022] Open
Abstract
l-carnosine (β-alanyl-l-histidine) (Car hereafter) is a natural dipeptide widely distributed in mammalian tissues and reaching high concentrations (0.7-2.0 mM) in the brain. The molecular features of the dipeptide underlie the antioxidant, anti-aggregating and metal chelating ability showed in a large number of physiological effects, while the biological mechanisms involved in the protective role found against several diseases cannot be explained on the basis of the above-mentioned properties alone, requiring further research efforts. It has been reported that l-carnosine increases the secretion and expression of various neurotrophic factors and affects copper homeostasis in nervous cells inducing Cu cellular uptake in keeping with the key metal-sensing system. Having in mind this l-carnosine ability, here we report the copper-binding and ionophore ability of l-carnosine to activate tyrosine kinase cascade pathways in PC12 cells and stimulate the expression of BDNF. Furthermore, the study was extended to verify the ability of the dipeptide to favor copper signaling inducing the expression of VEGF. Being aware that the potential protective action of l-carnosine is drastically hampered by its hydrolysis, we also report on the behavior of a conjugate of l-carnosine with trehalose that blocks the carnosinase degradative activity. Overall, our findings describe a copper tuning effect on the ability of l-carnosine and, particularly its conjugate, to activate tyrosine kinase cascade pathways.
Collapse
Affiliation(s)
- Irina Naletova
- Institute of Crystallography, National Council of Research—CNR, Via Paolo Gaifami 18, 95126 Catania, Italy;
- National Inter-University Consortium Metals Chemistry in Biological Systems (CIRCMSB), Via Celso Ulpiani 27, 70126 Bari, Italy
| | - Valentina Greco
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| | - Sebastiano Sciuto
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| | - Francesco Attanasio
- Institute of Crystallography, National Council of Research—CNR, Via Paolo Gaifami 18, 95126 Catania, Italy;
| | - Enrico Rizzarelli
- Institute of Crystallography, National Council of Research—CNR, Via Paolo Gaifami 18, 95126 Catania, Italy;
- National Inter-University Consortium Metals Chemistry in Biological Systems (CIRCMSB), Via Celso Ulpiani 27, 70126 Bari, Italy
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| |
Collapse
|
44
|
Analysis of Antidepressant-like Effects and Action Mechanisms of GSB-106, a Small Molecule, Affecting the TrkB Signaling. Int J Mol Sci 2021; 22:ijms222413381. [PMID: 34948177 PMCID: PMC8704497 DOI: 10.3390/ijms222413381] [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: 10/27/2021] [Revised: 12/01/2021] [Accepted: 12/10/2021] [Indexed: 12/13/2022] Open
Abstract
Induction of BDNF-TrkB signaling is associated with the action mechanisms of conventional and fast-acting antidepressants. GSB-106, developed as a small dimeric dipeptide mimetic of BDNF, was previously shown to produce antidepressant-like effects in the mouse Porsolt test, tail suspension test, Nomura water wheel test, in the chronic social defeat stress model and in the inflammation-induced model of depression. In the present study, we evaluated the effect of chronic per os administration of GSB-106 to Balb/c mice under unpredictable chronic mild stress (UCMS). It was observed for the first time that long term GSB-106 treatment (1 mg/kg, 26 days) during ongoing UCMS procedure ameliorated the depressive-like behaviors in mice as indicated by the Porsolt test. In addition, chronic per os administration of GSB-106 resulted in an increase in BDNF levels, which were found to be decreased in the prefrontal cortex and hippocampus of mice after UCMS. Furthermore, prolonged GSB-106 treatment was accompanied by an increase in the content of pTrkB706/707 in the prefrontal cortex and by a pronounced increase in the level of pTrkB816 in both studied brain structures of mice subjected to UCMS procedure. In summary, the present data show that chronic GSB-106 treatment produces an antidepressant-like effect in the unpredictable chronic mild stress model, which is likely to be associated with the regulation of the BDNF-TrkB signaling.
Collapse
|
45
|
Gonzalez S, McHugh TLM, Yang T, Syriani W, Massa SM, Longo FM, Simmons DA. Small molecule modulation of TrkB and TrkC neurotrophin receptors prevents cholinergic neuron atrophy in an Alzheimer's disease mouse model at an advanced pathological stage. Neurobiol Dis 2021; 162:105563. [PMID: 34838668 DOI: 10.1016/j.nbd.2021.105563] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/05/2021] [Accepted: 11/22/2021] [Indexed: 12/23/2022] Open
Abstract
Degeneration of basal forebrain cholinergic neurons (BFCNs) in the nucleus basalis of Meynert (NBM) and vertical diagonal band (VDB) along with their connections is a key pathological event leading to memory impairment in Alzheimer's disease (AD). Aberrant neurotrophin signaling via Trks and the p75 neurotrophin receptor (p75NTR) contributes importantly to BFCN dystrophy. While NGF/TrkA signaling has received the most attention in this regard, TrkB and TrkC signaling also provide trophic support to BFCNs and these receptors may be well located to preserve BFCN connectivity. We previously identified a small molecule TrkB/TrkC ligand, LM22B-10, that promotes cell survival and neurite outgrowth in vitro and activates TrkB/TrkC signaling in the hippocampus of aged mice when given intranasally, but shows poor oral bioavailability. An LM22B-10 derivative, PTX-BD10-2, with improved oral bioavailability has been developed and this study examined its effects on BFCN atrophy in the hAPPLond/Swe (APPL/S) AD mouse model. Oral delivery of PTX-BD10-2 was started after appreciable amyloid and cholinergic pathology was present to parallel the clinical context, as most AD patients start treatment at advanced pathological stages. PTX-BD10-2 restored cholinergic neurite integrity in the NBM and VDB, and reduced NBM neuronal atrophy in symptomatic APPL/S mice. Dystrophy of cholinergic neurites in BF target regions, including the cortex, hippocampus, and amygdala, was also reduced with treatment. Finally, PTX-BD10-2 reduced NBM tau pathology and improved the survival of cholinergic neurons derived from human induced pluripotent stem cells (iPSCs) after amyloid-β exposure. These data provide evidence that targeting TrkB and TrkC signaling with PTX-BD10-2 may be an effective disease-modifying strategy for combating cholinergic dysfunction in AD. The potential for clinical translation is further supported by the compound's reduction of AD-related degenerative processes that have progressed beyond early stages and its neuroprotective effects in human iPSC-derived cholinergic neurons.
Collapse
Affiliation(s)
- Selena Gonzalez
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Tyne L M McHugh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Tao Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Wassim Syriani
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Stephen M Massa
- Department of Neurology, Laboratory for Computational Neurochemistry and Drug Discovery, Veterans Affairs Health Care System and Department of Neurology, University of California-San Francisco, San Francisco, CA 94121, United States of America
| | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America
| | - Danielle A Simmons
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, United States of America.
| |
Collapse
|
46
|
Saeger HN, Olson DE. Psychedelic-inspired approaches for treating neurodegenerative disorders. J Neurochem 2021; 162:109-127. [PMID: 34816433 DOI: 10.1111/jnc.15544] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/19/2021] [Accepted: 11/21/2021] [Indexed: 12/21/2022]
Abstract
Psychedelics are increasingly being recognized for their potential to treat a wide range of brain disorders including depression, post-traumatic stress disorder (PTSD), and substance use disorder. Their broad therapeutic potential might result from an ability to rescue cortical atrophy common to many neuropsychiatric and neurodegenerative diseases by impacting neurotrophic factor gene expression, activating neuronal growth and survival mechanisms, and modulating the immune system. While the therapeutic potential of psychedelics has not yet been extended to neurodegenerative disorders, we provide evidence suggesting that approaches based on psychedelic science might prove useful for treating these diseases. The primary target of psychedelics, the 5-HT2A receptor, plays key roles in cortical neuron health and is dysregulated in Alzheimer's disease. Moreover, evidence suggests that psychedelics and related compounds could prove useful for treating the behavioral and psychological symptoms of dementia (BPSD). While more research is needed to probe the effects of psychedelics in models of neurodegenerative diseases, the robust effects of these compounds on structural and functional neuroplasticity and inflammation clearly warrant further investigation.
Collapse
Affiliation(s)
- Hannah N Saeger
- Pharmacology and Toxicology Graduate Group, University of California, Davis, Davis, California, USA
| | - David E Olson
- Department of Chemistry, University of California, Davis, Davis, California, USA.,Department of Biochemistry & Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California, USA.,Center for Neuroscience, University of California, Davis, Davis, California, USA
| |
Collapse
|
47
|
Zhang D, Zhao S, Zhang Z, Xu D, Lian D, Wu J, He D, Sun K, Li L. Regulation of the p75 neurotrophin receptor attenuates neuroinflammation and stimulates hippocampal neurogenesis in experimental Streptococcus pneumoniae meningitis. J Neuroinflammation 2021; 18:253. [PMID: 34727939 PMCID: PMC8561879 DOI: 10.1186/s12974-021-02294-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/09/2021] [Indexed: 12/12/2022] Open
Abstract
Background Streptococcus pneumoniae meningitis is a destructive central nervous system (CNS) infection with acute and long-term neurological disorders. Previous studies suggest that p75NTR signaling influences cell survival, apoptosis, and proliferation in brain-injured conditions. However, the role of p75NTR signaling in regulating pneumococcal meningitis (PM)-induced neuroinflammation and altered neurogenesis remains largely to be elucidated. Methods p75NTR signaling activation in the pathological process of PM was assessed. During acute PM, a small-molecule p75NTR modulator LM11A-31 or vehicle was intranasally administered for 3 days prior to S. pneumoniae exposure. At 24 h post-infection, clinical severity, histopathology, astrocytes/microglia activation, neuronal apoptosis and necrosis, inflammation-related transcription factors and proinflammatory cytokines/mediators were evaluated. Additionally, p75NTR was knocked down by the adenovirus-mediated short-hairpin RNA (shRNA) to ascertain the role of p75NTR in PM. During long-term PM, the intranasal administration of LM11A-31 or vehicle was continued for 7 days after successfully establishing the PM model. Dynamic changes in inflammation and hippocampal neurogenesis were assessed. Results Our results revealed that both 24 h (acute) and 7, 14, 28 day (long-term) groups of infected rats showed increased p75NTR expression in the brain. During acute PM, modulation of p75NTR through pretreatment of PM model with LM11A-31 significantly alleviated S. pneumoniae-induced clinical severity, histopathological injury and the activation of astrocytes and microglia. LM11A-31 pretreatment also significantly ameliorated neuronal apoptosis and necrosis. Moreover, we found that blocking p75NTR with LM11A-31 decreased the expression of inflammation-related transcription factors (NF-κBp65, C/EBPβ) and proinflammatory cytokines/mediators (IL-1β, TNF-α, IL-6 and iNOS). Furthermore, p75NTR knockdown induced significant changes in histopathology and inflammation-related transcription factors expression. Importantly, long-term LM11A-31 treatment accelerated the resolution of PM-induced inflammation and significantly improved hippocampal neurogenesis. Conclusion Our findings suggest that the p75NTR signaling plays an essential role in the pathogenesis of PM. Targeting p75NTR has beneficial effects on PM rats by alleviating neuroinflammation and promoting hippocampal neurogenesis. Thus, the p75NTR signaling may be a potential therapeutic target to improve the outcome of PM. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02294-w.
Collapse
Affiliation(s)
- Dandan Zhang
- Department of Pediatric Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Shanghai, 200092, China
| | - Shengnan Zhao
- Department of Pediatric Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Shanghai, 200092, China
| | - Zhijie Zhang
- Department of Pediatric Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Shanghai, 200092, China
| | - Danfeng Xu
- Department of Pediatric Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Shanghai, 200092, China
| | - Di Lian
- Department of Pediatric Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Shanghai, 200092, China
| | - Jing Wu
- Department of Pediatric Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Shanghai, 200092, China
| | - Dake He
- Department of Pediatric Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Shanghai, 200092, China
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Shanghai, 200092, China.
| | - Ling Li
- Department of Pediatric Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Shanghai, 200092, China.
| |
Collapse
|
48
|
Brahimi F, Galan A, Siegel S, Szobota S, Sarunic MV, Foster AC, Saragovi HU. Therapeutic Neuroprotection by an Engineered Neurotrophin that Selectively Activates Tropomyosin Receptor Kinase (Trk) Family Neurotrophin Receptors but Not the p75 Neurotrophin Receptor. Mol Pharmacol 2021; 100:491-501. [PMID: 34470776 DOI: 10.1124/molpharm.121.000301] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/12/2021] [Indexed: 11/22/2022] Open
Abstract
The neurotrophin growth factors bind and activate two types of cell surface receptors: the tropomyosin receptor kinase (Trk) family and p75. TrkA, TrkB, and TrkC are bound preferentially by nerve growth factor, brain-derived neurotrophic factor, and neurotrophin 3 (NT3), respectively, to activate neuroprotective signals. The p75 receptors are activated by all neurotrophins, and paradoxically in neurodegenerative disease p75 is upregulated and mediates neurotoxic signals. To test neuroprotection strategies, we engineered NT3 to broadly activate Trk receptors (mutant D) or to reduce p75 binding (mutant RK). We also combined these features in a molecule that activates TrkA, TrkB, and TrkC but has reduced p75 binding (mutant DRK). In neurodegenerative disease mouse models in vivo, the DRK protein is a superior therapeutic agent compared with mutant D, mutant RK, and wild-type neurotrophins and protects a broader range of stressed neurons. This work rationalizes a therapeutic strategy based on the biology of each type of receptor, avoiding activation of p75 toxicity while broadly activating neuroprotection in stressed neuronal populations expressing different Trk receptors. SIGNIFICANCE STATEMENT: The neurotrophins nerve growth factor, brain-derived neurotrophic factor, and neurotrophin 3 each can activate a tropomyosin receptor kinase (Trk) A, TrkB, or TrkC receptor, respectively, and all can activate a p75 receptor. Trks and p75 mediate opposite signals. We report the engineering of a protein that activates all Trks, combined with low p75 binding, as an effective therapeutic agent in vivo.
Collapse
Affiliation(s)
- Fouad Brahimi
- Lady Davis Institute-Jewish General Hospital (F.B., A.G., H.U.S.), Pharmacology and Therapeutics (H.U.S.), and Ophthalmology and Vision Science (H.U.S.), McGill University, Montreal, Quebec, Canada; Otonomy, Inc., San Diego, California (S.Si., S.Sz., A.C.F.); and School of Engineering Science, Simon Fraser University, British Columbia, Canada (M.V.S.)
| | - Alba Galan
- Lady Davis Institute-Jewish General Hospital (F.B., A.G., H.U.S.), Pharmacology and Therapeutics (H.U.S.), and Ophthalmology and Vision Science (H.U.S.), McGill University, Montreal, Quebec, Canada; Otonomy, Inc., San Diego, California (S.Si., S.Sz., A.C.F.); and School of Engineering Science, Simon Fraser University, British Columbia, Canada (M.V.S.)
| | - Sairey Siegel
- Lady Davis Institute-Jewish General Hospital (F.B., A.G., H.U.S.), Pharmacology and Therapeutics (H.U.S.), and Ophthalmology and Vision Science (H.U.S.), McGill University, Montreal, Quebec, Canada; Otonomy, Inc., San Diego, California (S.Si., S.Sz., A.C.F.); and School of Engineering Science, Simon Fraser University, British Columbia, Canada (M.V.S.)
| | - Stephanie Szobota
- Lady Davis Institute-Jewish General Hospital (F.B., A.G., H.U.S.), Pharmacology and Therapeutics (H.U.S.), and Ophthalmology and Vision Science (H.U.S.), McGill University, Montreal, Quebec, Canada; Otonomy, Inc., San Diego, California (S.Si., S.Sz., A.C.F.); and School of Engineering Science, Simon Fraser University, British Columbia, Canada (M.V.S.)
| | - Marinko V Sarunic
- Lady Davis Institute-Jewish General Hospital (F.B., A.G., H.U.S.), Pharmacology and Therapeutics (H.U.S.), and Ophthalmology and Vision Science (H.U.S.), McGill University, Montreal, Quebec, Canada; Otonomy, Inc., San Diego, California (S.Si., S.Sz., A.C.F.); and School of Engineering Science, Simon Fraser University, British Columbia, Canada (M.V.S.)
| | - Alan C Foster
- Lady Davis Institute-Jewish General Hospital (F.B., A.G., H.U.S.), Pharmacology and Therapeutics (H.U.S.), and Ophthalmology and Vision Science (H.U.S.), McGill University, Montreal, Quebec, Canada; Otonomy, Inc., San Diego, California (S.Si., S.Sz., A.C.F.); and School of Engineering Science, Simon Fraser University, British Columbia, Canada (M.V.S.)
| | - H Uri Saragovi
- Lady Davis Institute-Jewish General Hospital (F.B., A.G., H.U.S.), Pharmacology and Therapeutics (H.U.S.), and Ophthalmology and Vision Science (H.U.S.), McGill University, Montreal, Quebec, Canada; Otonomy, Inc., San Diego, California (S.Si., S.Sz., A.C.F.); and School of Engineering Science, Simon Fraser University, British Columbia, Canada (M.V.S.)
| |
Collapse
|
49
|
Mitra S, Gera R, Linderoth B, Lind G, Wahlberg L, Almqvist P, Behbahani H, Eriksdotter M. A Review of Techniques for Biodelivery of Nerve Growth Factor (NGF) to the Brain in Relation to Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1331:167-191. [PMID: 34453298 DOI: 10.1007/978-3-030-74046-7_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Age-dependent progressive neurodegeneration and associated cognitive dysfunction represent a serious concern worldwide. Currently, dementia accounts for the fifth highest cause of death, among which Alzheimer's disease (AD) represents more than 60% of the cases. AD is associated with progressive cognitive dysfunction which affects daily life of the affected individual and associated family. The cognitive dysfunctions are at least partially due to the degeneration of a specific set of neurons (cholinergic neurons) whose cell bodies are situated in the basal forebrain region (basal forebrain cholinergic neurons, BFCNs) but innervate wide areas of the brain. It has been explicitly shown that the delivery of the neurotrophic protein nerve growth factor (NGF) can rescue BFCNs and restore cognitive dysfunction, making NGF interesting as a potential therapeutic substance for AD. Unfortunately, NGF cannot pass through the blood-brain barrier (BBB) and thus peripheral administration of NGF protein is not viable therapeutically. NGF must be delivered in a way which will allow its brain penetration and availability to the BFCNs to modulate BFCN activity and viability. Over the past few decades, various methodologies have been developed to deliver NGF to the brain tissue. In this chapter, NGF delivery methods are discussed in the context of AD.
Collapse
Affiliation(s)
- Sumonto Mitra
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden.
| | - Ruchi Gera
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Linderoth
- Section of Neurosurgery, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Göran Lind
- Section of Neurosurgery, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Per Almqvist
- Section of Neurosurgery, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Homira Behbahani
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden.,Karolinska Universitets laboratoriet (LNP5), Karolinska University Hospital, Stockholm, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
50
|
In vivo functions of p75 NTR: challenges and opportunities for an emerging therapeutic target. Trends Pharmacol Sci 2021; 42:772-788. [PMID: 34334250 DOI: 10.1016/j.tips.2021.06.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/31/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022]
Abstract
The p75 neurotrophin receptor (p75NTR) functions at the molecular nexus of cell death, survival, and differentiation. In addition to its contribution to neurodegenerative diseases and nervous system injuries, recent studies have revealed unanticipated roles of p75NTR in liver repair, fibrinolysis, lung fibrosis, muscle regeneration, and metabolism. Linking these various p75NTR functions more precisely to specific mechanisms marks p75NTR as an emerging candidate for therapeutic intervention in a wide range of disorders. Indeed, small molecule inhibitors of p75NTR binding to neurotrophins have shown efficacy in models of Alzheimer's disease (AD) and neurodegeneration. Here, we outline recent advances in understanding p75NTR pleiotropic functions in vivo, and propose an integrated view of p75NTR and its challenges and opportunities as a pharmacological target.
Collapse
|