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
|
Merighi A. Brain-Derived Neurotrophic Factor, Nociception, and Pain. Biomolecules 2024; 14:539. [PMID: 38785946 PMCID: PMC11118093 DOI: 10.3390/biom14050539] [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: 02/08/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
This article examines the involvement of the brain-derived neurotrophic factor (BDNF) in the control of nociception and pain. BDNF, a neurotrophin known for its essential role in neuronal survival and plasticity, has garnered significant attention for its potential implications as a modulator of synaptic transmission. This comprehensive review aims to provide insights into the multifaceted interactions between BDNF and pain pathways, encompassing both physiological and pathological pain conditions. I delve into the molecular mechanisms underlying BDNF's involvement in pain processing and discuss potential therapeutic applications of BDNF and its mimetics in managing pain. Furthermore, I highlight recent advancements and challenges in translating BDNF-related research into clinical practice.
Collapse
Affiliation(s)
- Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, 10095 Turin, Italy
| |
Collapse
|
3
|
Kim JH, Irfan M, Hossain MA, George A, Chung S. BDNF/TrkB Is a Crucial Regulator in the Inflammation-Mediated Odontoblastic Differentiation of Dental Pulp Stem Cells. Cells 2023; 12:1851. [PMID: 37508514 PMCID: PMC10378460 DOI: 10.3390/cells12141851] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
The odontoblastic differentiation of dental pulp stem cells (DPSCs) associated with caries injury happens in an inflammatory context. We recently demonstrated that there is a link between inflammation and dental tissue regeneration, identified via enhanced DPSC-mediated dentinogenesis in vitro. Brain-derived neurotrophic factor (BDNF) is a nerve growth factor-related gene family molecule which functions through tropomyosin receptor kinase B (TrkB). While the roles of BDNF in neural tissue repair and other regeneration processes are well identified, its role in dentinogenesis has not been explored. Furthermore, the role of BDNF receptor-TrkB in inflammation-induced dentinogenesis remains unknown. The role of BDNF/TrkB was examined during a 17-day odontogenic differentiation of DPSCs. Human DPSCs were subjected to odontogenic differentiation in dentinogenic media treated with inflammation inducers (LTA or TNFα), BDNF, and a TrkB agonist (LM22A-4) and/or antagonist (CTX-B). Our data show that BDNF and TrkB receptors affect the early and late stages of the odontogenic differentiation of DPSCs. Immunofluorescent data confirmed the expression of BDNF and TrkB in DPSCs. Our ELISA and qPCR data demonstrate that TrkB agonist treatment increased the expression of dentin matrix protein-1 (DMP-1) during early DPSC odontoblastic differentiation. Coherently, the expression levels of runt-related transcription factor 2 (RUNX-2) and osteocalcin (OCN) were increased. TNFα, which is responsible for a diverse range of inflammation signaling, increased the levels of expression of dentin sialophosphoprotein (DSPP) and DMP1. Furthermore, BDNF significantly potentiated its effect. The application of CTX-B reversed this effect, suggesting TrkB`s critical role in TNFα-mediated dentinogenesis. Our studies provide novel findings on the role of BDNF-TrkB in the inflammation-induced odontoblastic differentiation of DPSCs. This finding will address a novel regulatory pathway and a therapeutic approach in dentin tissue engineering using DPSCs.
Collapse
Affiliation(s)
| | | | | | | | - Seung Chung
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL 60612, USA; (J.-H.K.); (M.I.); (M.A.H.); (A.G.)
| |
Collapse
|
4
|
Liu B, Liu Y, Li S, Chen P, Zhang J, Feng L. BDNF promotes mouse follicular development and reverses ovarian aging by promoting cell proliferation. J Ovarian Res 2023; 16:83. [PMID: 37106468 PMCID: PMC10134588 DOI: 10.1186/s13048-023-01163-9] [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: 11/21/2022] [Accepted: 04/16/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) plays an important role in ovarian function including follicle development and oocyte maturation, and embryonic development. However, whether BDNF treatment can reimpose ovarian aging and impaired fertility remains elusive. In this study, we investigated the reproductive outcomes of BDNF treatment and potential mechanisms in aged mice. METHOD "Aged" mice (35-37 weeks old, n = 68) were treated with recombinant human BDNF protein (rhBDNF, 1 µg/200 µL) through daily intraperitoneal (IP) injection for 10 days with/without ovulation induction. Reproductive age mice (8-10 weeks old, n = 28) were treated with ANA 12 (a selective BDNF receptor, TrkB antagonist) through daily IP injection for 5 days with/without ovulation induction. Ovarian function was assessed by ovarian weight, number of follicles, and sex hormone productions. Following induction of ovulation, the number of total oocytes or abnormal oocytes, and blastocyst formation were assessed. Reproductive functions of the mice were evaluated, including pregnancy rate, mating duration for conception, implantation sites, litter size, and weight of offspring. Finally, the molecular mechanism of the effects of BDNF on ovarian cell functions in mice were examined by Western blot and immunofluorescence. RESULTS rhBDNF treatment increased the ovarian weight, number of follicles, number and quality of oocytes including increased blastocysts formation, blood estrogen levels, and pregnancy rate in 35-37-week-old mice. Conversely, BDNF receptor antagonist, ANA 12, treatment decreased the ovarian volume and number of antral follicles and increased the proportion of abnormal oocytes in 8-10-week-old mice. We further demonstrated that BDNF treatment promoted ovarian cell proliferation as well as activation of TrkB and cyclinD1-creb signalling. CONCLUSION We demonstrated that ten consecutive days of daily IP injection of rhBDNF rescued ovarian function in aged mice. Our results further indicate that TrkB and cyclin D1-creb signaling may underlie the BDNF function in ovaries. Targeting BDNF-TrkB signaling is a potential novel therapeutic strategy to reverse ovarian aging.
Collapse
Affiliation(s)
- Bin Liu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Reproduction, Xinhua Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai, China
| | - Yongjie Liu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuman Li
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Pingping Chen
- Department of Reproduction, Xinhua Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai, China
| | - Jun Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Liping Feng
- Department of Obstetrics and Gynaecology, Duke University, Durham, NC, USA.
| |
Collapse
|
5
|
Tang C, Liu M, Zhou Z, Li H, Yang C, Yang L, Xiang J. Treadmill Exercise Alleviates Cognition Disorder by Activating the FNDC5: Dual Role of Integrin αV/β5 in Parkinson's Disease. Int J Mol Sci 2023; 24:ijms24097830. [PMID: 37175535 PMCID: PMC10178565 DOI: 10.3390/ijms24097830] [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: 02/20/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Parkinson's disease with cognitive impairment (PD-CI) results in several clinical outcomes for which specific treatment is lacking. Although the pathogenesis of PD-CI has not yet been fully elucidated, it is related to neuronal plasticity decline in the hippocampus region. The dopaminergic projections from the substantia nigra to the hippocampus are critical in regulating hippocampal plasticity. Recently, aerobic exercise has been recognized as an effective therapeutic strategy for enhancing plasticity through the secretion of various muscle factors. The exact role of FNDC5-an upregulated, newly identified myokine produced after exercise-in mediating hippocampal plasticity and regional dopaminergic projections in PD-CI remains unclear. In this study, the effect of treadmill exercise on hippocampal synaptic plasticity was evaluated in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced chronic PD models. The results showed that treadmill exercise substantially alleviated the motor dysfunction, cognition disorder, and dopaminergic neuron degeneration induced by MPTP. Here, we discovered that the quadriceps, serum, and brain FNDC5 levels were lower in PD mice and that intervention with treadmill exercise restored FNDC5 levels. Moreover, treadmill exercise enhanced the synaptic plasticity of hippocampal pyramidal neurons via increased dopamine levels and BDNF in the PD mice. The direct protective effect of FNDC5 is achieved by promoting the secretion of BDNF in the hippocampal neurons via binding the integrin αVβ5 receptor, thereby improving synaptic plasticity. Regarding the indirect protection effect, FNDC5 promotes the dopaminergic connection from the substantia nigra to the hippocampus by mediating the interaction between the integrin αVβ5 of the hippocampal neurons and the CD90 molecules on the membrane of dopaminergic terminals. Our findings demonstrated that treadmill exercise could effectively alleviate cognitive disorders via the activation of the FNDC5-BDNF pathway and enhance the dopaminergic synaptic connection from SNpc to the hippocampus in the MPTP-induced chronic PD model.
Collapse
Affiliation(s)
- Chuanxi Tang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Mengting Liu
- Department of Rehabilitation, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, China
- The College of Medical Technology, Xuzhou Medical University, Xuzhou 221004, China
| | - Zihang Zhou
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Hao Li
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Chenglin Yang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Li Yang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Jie Xiang
- Department of Rehabilitation, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, China
| |
Collapse
|
6
|
Li Y, Wei C, Wang W, Li Q, Wang Z. Tropomyosin receptor kinase B (TrkB) signalling: targeted therapy in neurogenic tumours. J Pathol Clin Res 2022; 9:89-99. [PMID: 36533776 PMCID: PMC9896160 DOI: 10.1002/cjp2.307] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
Tropomyosin receptor kinase B (TrkB), a transmembrane receptor protein, has been found to play a pivotal role in neural development. This protein is encoded by the neurotrophic receptor tyrosine kinase 2 (NTRK2) gene, and its abnormal activation caused by NTRK2 overexpression or fusion can contribute to tumour initiation, progression, and resistance to therapeutics in multiple types of neurogenic tumours. Targeted therapies for this mechanism have been designed and developed in preclinical and clinical studies, including selective TrkB inhibitors and pan-TRK inhibitors. This review describes the gene structure, biological function, abnormal TrkB activation mechanism, and current-related targeted therapies in neurogenic tumours.
Collapse
Affiliation(s)
- Yuehua Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Chengjiang Wei
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Wei Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Zhi‐Chao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| |
Collapse
|
7
|
Izumi Y, Hsu FF, Conway CR, Nagele P, Mennerick SJ, Zorumski CF. Nitrous Oxide, a Rapid Antidepressant, Has Ketamine-like Effects on Excitatory Transmission in the Adult Hippocampus. Biol Psychiatry 2022; 92:964-972. [PMID: 36050137 PMCID: PMC10107749 DOI: 10.1016/j.biopsych.2022.06.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/03/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Nitrous oxide (N2O) is a noncompetitive inhibitor of NMDA receptors that appears to have ketamine-like rapid antidepressant effects in patients with treatment-resistant major depression. In preclinical studies, ketamine enhances glutamate-mediated synaptic transmission in the hippocampus and prefrontal cortex. In this study, we examined the effects of N2O on glutamate transmission in the hippocampus and compared its effects to those of ketamine. METHODS Glutamate-mediated synaptic transmission was studied in the CA1 region of hippocampal slices from adult albino rats using standard extracellular recording methods. Effects of N2O and ketamine at subanesthetic concentrations were evaluated by acute administration. RESULTS Akin to 1 μM ketamine, 30% N2O administered for 15-20 minutes resulted in persistent enhancement of synaptic responses mediated by both AMPA receptors and NMDA receptors. Synaptic enhancement by both N2O and ketamine was blocked by co-administration of a competitive NMDA receptor antagonist at saturating concentration, but only ketamine was blocked by an AMPA receptor antagonist. Synaptic enhancement by both agents involved TrkB (tropomyosin receptor kinase B), mTOR (mechanistic target of rapamycin), and NOS (nitric oxide synthase) with some differences between N2O and ketamine. N2O potentiation occluded enhancement by ketamine, and in vivo N2O exposure occluded further potentiation by both N2O and ketamine. CONCLUSIONS These results indicate that N2O has ketamine-like effects on hippocampal synaptic function at a subanesthetic, but therapeutically relevant concentration. These 2 rapid antidepressants have similar, but not identical mechanisms that result in persisting synaptic enhancement, possibly contributing to psychotropic actions.
Collapse
Affiliation(s)
- Yukitoshi Izumi
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri; Center for Brain Research in Mood Disorders, Washington University School of Medicine, St. Louis, Missouri
| | - Fong-Fu Hsu
- Department of Medicine and Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
| | - Charles R Conway
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri; Center for Brain Research in Mood Disorders, Washington University School of Medicine, St. Louis, Missouri
| | - Peter Nagele
- Department of Anesthesia and Critical Care, University of Chicago, Chicago, Illinois
| | - Steven J Mennerick
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri; Center for Brain Research in Mood Disorders, Washington University School of Medicine, St. Louis, Missouri
| | - Charles F Zorumski
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri; Center for Brain Research in Mood Disorders, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
8
|
Van Hook MJ. Brain-derived neurotrophic factor is a regulator of synaptic transmission in the adult visual thalamus. J Neurophysiol 2022; 128:1267-1277. [PMID: 36224192 PMCID: PMC9662800 DOI: 10.1152/jn.00540.2021] [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: 12/16/2021] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 11/22/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is an important regulator of circuit development, neuronal survival, and plasticity throughout the nervous system. In the visual system, BDNF is produced by retinal ganglion cells (RGCs) and transported along their axons to central targets. Within the dorsolateral geniculate nucleus (dLGN), a key RGC projection target for conscious vision, the BDNF receptor tropomyosin receptor kinase B (TrkB) is present on RGC axon terminals and postsynaptic thalamocortical (TC) relay neuron dendrites. Based on this, the goal of this study was to determine how BDNF modulates the conveyance of signals through the retinogeniculate (RG) pathway of adult mice. Application of BDNF to dLGN brain slices increased TC neuron spiking evoked by optogenetic stimulation of RGC axons. There was a modest contribution to this effect from a BDNF-dependent enhancement of TC neuron intrinsic excitability including increased input resistance and membrane depolarization. BDNF also increased evoked vesicle release from RGC axon terminals, as evidenced by increased amplitude of evoked excitatory postsynaptic currents (EPSCs), which was blocked by inhibition of TrkB or phospholipase C. High-frequency stimulation revealed that BDNF increased synaptic vesicle pool size, release probability, and replenishment rate. There was no effect of BDNF on EPSC amplitude or short-term plasticity of corticothalamic feedback synapses. Thus, BDNF regulates RG synapses by both presynaptic and postsynaptic mechanisms. These findings suggest that BNDF influences the flow of visual information through the retinogeniculate pathway.NEW & NOTEWORTHY Brain-derived neurotrophic factor (BDNF) is an important regulator of neuronal development and plasticity. In the visual system, BDNF is transported along retinal ganglion cell (RGC) axons to the dorsolateral geniculate nucleus (dLGN), although it is not known how it influences mature dLGN function. Here, BDNF enhanced thalamocortical relay neuron responses to signals arising from RGC axons in the dLGN, pointing toward an important role for BDNF in processing signals en route to the visual cortex.
Collapse
Affiliation(s)
- Matthew J Van Hook
- Truhlsen Eye Institute, Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
9
|
McGinty JF. BDNF as a therapeutic candidate for cocaine use disorders. ADDICTION NEUROSCIENCE 2022; 2:100006. [PMID: 37206683 PMCID: PMC10195100 DOI: 10.1016/j.addicn.2022.100006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Cocaine self-administration disturbs intracellular signaling in multiple reward circuitry neurons that underlie relapse to drug seeking. Cocaine-induced deficits in prelimbic (PL) prefrontal cortex change during abstinence, resulting in different neuroadaptations during early withdrawal from cocaine self-administration than after one or more weeks of abstinence. Infusion of brain-derived neurotrophic factor (BDNF) into the PL cortex immediately following a final session of cocaine self-administration attenuates relapse to cocaine seeking for an extended period. BDNF affects local (PL) and distal subcortical target areas that mediate cocaine-induced neuroadaptations that lead to cocaine seeking. Blocking synaptic activity selectively in the PL projection to the nucleus accumbens during early withdrawal prevents BDNF from decreasing subsequent relapse. In contrast, blocking synaptic activity selectively in the PL projection to the paraventricular thalamic nucleus by itself decreases subsequent relapse and prior intra-PL BDNF infusion prevents the decrease. Infusion of BDNF into other brain structures at different timepoints after cocaine self administration differentially alters cocaine seeking. Thus, the effects of BDNF on drug seeking are different depending on the brain region, the timepoint of intervention, and the specific pathway that is affected.
Collapse
Affiliation(s)
- Jacqueline F McGinty
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Ave MSC 510, Charleston, SC 29425, USA
| |
Collapse
|
10
|
Gao C, Wu M, Du Q, Deng J, Shen J. Naringin Mediates Adult Hippocampal Neurogenesis for Antidepression via Activating CREB Signaling. Front Cell Dev Biol 2022; 10:731831. [PMID: 35478969 PMCID: PMC9037031 DOI: 10.3389/fcell.2022.731831] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 03/07/2022] [Indexed: 11/17/2022] Open
Abstract
The brain-derived neurotrophic factor/tropomyosin receptor kinase B/cAMP response element-binding protein (BDNF/TrkB/CREB) signaling pathway is a critical therapeutic target for inducing adult hippocampal neurogenesis and antidepressant therapy. In this study, we tested the hypothesis that naringin, a natural medicinal compound, could promote adult hippocampal neurogenesis and improve depression-like behaviors via regulating the BDNF/TrkB/CREB signaling pathway. We first investigated the effects of naringin on promoting adult hippocampal neurogenesis in both normal and chronic corticosterone (CORT)-induced depressive mice. Under physiological condition, naringin treatment enhanced the proliferation of neural stem/progenitor cells (NSPCs) and accelerated neuronal differentiation. In CORT-induced depression mouse model, naringin treatment promoted neuronal differentiation and maturation of NSPCs for hippocampal neurogenesis. Forced swim test, tail suspension test, and open field test confirmed the antidepressant and anxiolytic effects of naringin. Co-treatment of temozolomide (TMZ), a neurogenic inhibitor, abolished these antidepressant and anxiolytic effects. Meanwhile, naringin treatment increased phosphorylation of cAMP response element binding protein (CREB) but had no effect on the expression of brain-derived neurotrophic factor and phosphorylation of TrkB in the hippocampus of CORT-induced depressive mice. Co-treatment of CREB inhibitor 666-15, rather than TrkB inhibitor Cyc-B, abolished the neurogenesis-promoting and antidepressant effects of naringin. Taken together, naringin has antidepressant and anxiolytic effects, and the underlying mechanisms could be attributed to enhance hippocampal neurogenesis via activating CREB signaling.
Collapse
Affiliation(s)
- Chong Gao
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hon Kong SAR, China
- The Institute of Brain and Cognitive Sciences, School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Meiling Wu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hon Kong SAR, China
| | - Qiaohui Du
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hon Kong SAR, China
| | - Jiagang Deng
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
| | - Jiangang Shen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hon Kong SAR, China
- *Correspondence: Jiangang Shen,
| |
Collapse
|
11
|
Ahn JW, Kim S, Ko S, Kim YH, Jeong JH, Chung S. Modified (−)-gallocatechin gallate-enriched green tea extract rescues age-related cognitive deficits by restoring hippocampal synaptic plasticity. Biochem Biophys Rep 2022; 29:101201. [PMID: 35198737 PMCID: PMC8841891 DOI: 10.1016/j.bbrep.2022.101201] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/19/2021] [Accepted: 01/03/2022] [Indexed: 11/17/2022] Open
Affiliation(s)
- Ji-Woong Ahn
- BnH Research Co., LTD., Goyang-si, Gyeonggi-do, 10594, Republic of Korea
| | - Sohyun Kim
- Brain Korea 21 Plus Project for Medical Science, Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sukjin Ko
- BnH Research Co., LTD., Goyang-si, Gyeonggi-do, 10594, Republic of Korea
| | - Young-Hwan Kim
- BnH Research Co., LTD., Goyang-si, Gyeonggi-do, 10594, Republic of Korea
| | - Ji-Hyun Jeong
- BnH Research Co., LTD., Goyang-si, Gyeonggi-do, 10594, Republic of Korea
| | - Seungsoo Chung
- BnH Research Co., LTD., Goyang-si, Gyeonggi-do, 10594, Republic of Korea
- Brain Korea 21 Plus Project for Medical Science, Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Corresponding author. Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| |
Collapse
|
12
|
Mannes M, Martin C, Menet C, Ballet S. Wandering beyond small molecules: peptides as allosteric protein modulators. Trends Pharmacol Sci 2021; 43:406-423. [PMID: 34857409 DOI: 10.1016/j.tips.2021.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 12/28/2022]
Abstract
Recent years have seen the rise of allosteric modulation as an innovative approach for drug design and discovery, efforts which culminated in the development of several clinical candidates. Allosteric modulation of many drug targets, including mainly membrane-embedded receptors, have been vastly explored through small molecule screening campaigns, but much less attention has been paid to peptide-based allosteric modulators. However, peptides have a significant impact on the pharmaceutical industry due to the typically higher potency and selectivity for their targets, as compared with small molecule therapeutics. Therefore, peptides represent one of the most promising classes of molecules that can modulate key biological pathways. Here, we report on the allosteric modulation of proteins (ranging from G protein-coupled receptors to specific protein-protein interactions) by peptides for applications in drug discovery.
Collapse
Affiliation(s)
- Morgane Mannes
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, Brussels, Belgium
| | - Charlotte Martin
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, Brussels, Belgium.
| | - Christel Menet
- Confo Therapeutics N.V., Technologiepark-Zwijnaarde 30, Ghent, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, Brussels, Belgium.
| |
Collapse
|
13
|
Vinel C, Rosser G, Guglielmi L, Constantinou M, Pomella N, Zhang X, Boot JR, Jones TA, Millner TO, Dumas AA, Rakyan V, Rees J, Thompson JL, Vuononvirta J, Nadkarni S, El Assan T, Aley N, Lin YY, Liu P, Nelander S, Sheer D, Merry CLR, Marelli-Berg F, Brandner S, Marino S. Comparative epigenetic analysis of tumour initiating cells and syngeneic EPSC-derived neural stem cells in glioblastoma. Nat Commun 2021; 12:6130. [PMID: 34675201 PMCID: PMC8531305 DOI: 10.1038/s41467-021-26297-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
Epigenetic mechanisms which play an essential role in normal developmental processes, such as self-renewal and fate specification of neural stem cells (NSC) are also responsible for some of the changes in the glioblastoma (GBM) genome. Here we develop a strategy to compare the epigenetic and transcriptional make-up of primary GBM cells (GIC) with patient-matched expanded potential stem cell (EPSC)-derived NSC (iNSC). Using a comparative analysis of the transcriptome of syngeneic GIC/iNSC pairs, we identify a glycosaminoglycan (GAG)-mediated mechanism of recruitment of regulatory T cells (Tregs) in GBM. Integrated analysis of the transcriptome and DNA methylome of GBM cells identifies druggable target genes and patient-specific prediction of drug response in primary GIC cultures, which is validated in 3D and in vivo models. Taken together, we provide a proof of principle that this experimental pipeline has the potential to identify patient-specific disease mechanisms and druggable targets in GBM.
Collapse
Affiliation(s)
- Claire Vinel
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Gabriel Rosser
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Loredana Guglielmi
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Myrianni Constantinou
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Nicola Pomella
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Xinyu Zhang
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - James R Boot
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Tania A Jones
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Thomas O Millner
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Anaelle A Dumas
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Vardhman Rakyan
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Jeremy Rees
- Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London, UK
| | - Jamie L Thompson
- Stem Cell Glycobiology Group, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Juho Vuononvirta
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Suchita Nadkarni
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Tedani El Assan
- Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London, UK
| | - Natasha Aley
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, UK
| | - Yung-Yao Lin
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
- Stem Cell Laboratory, National Bowel Research Centre, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 2 Newark Street, London, UK
| | - Pentao Liu
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, Hong Kong, Hong Kong
| | - Sven Nelander
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Denise Sheer
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Catherine L R Merry
- Stem Cell Glycobiology Group, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Federica Marelli-Berg
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK
| | - Sebastian Brandner
- Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, Queen Square, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, UK
| | - Silvia Marino
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, UK.
| |
Collapse
|
14
|
The brain-derived neurotrophic factor prompts platelet aggregation and secretion. Blood Adv 2021; 5:3568-3580. [PMID: 34546355 DOI: 10.1182/bloodadvances.2020004098] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 06/08/2021] [Indexed: 12/19/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) has both autocrine and paracrine roles in neurons, and its release and signaling mechanisms have been extensively studied in the central nervous system. Large quantities of BDNF have been reported in circulation, essentially stored in platelets with concentrations reaching 100- to 1000-fold those of neurons. Despite this abundance, the function of BDNF in platelet biology has not been explored. At low concentrations, BDNF primed platelets, acting synergistically with classical agonists. At high concentrations, BDNF induced complete biphasic platelet aggregation that in part relied on amplification from secondary mediators. Neurotrophin-4, but not nerve growth factor, and an activating antibody against the canonical BDNF receptor tropomyosin-related kinase B (TrkB) induced similar platelet responses to BDNF, suggesting TrkB could be the mediator. Platelets expressed, both at their surface and in their intracellular compartment, a truncated form of TrkB lacking its tyrosine kinase domain. BDNF-induced platelet aggregation was prevented by inhibitors of Ras-related C3 botulinum toxin substrate 1 (Rac1), protein kinase C, and phosphoinositide 3-kinase. BDNF-stimulated platelets secreted a panel of angiogenic and inflammatory cytokines, which may play a role in maintaining vascular homeostasis. Two families with autism spectrum disorder were found to carry rare missense variants in the BDNF gene. Platelet studies revealed defects in platelet aggregation to low concentrations of collagen, as well as reduced adenosine triphosphate secretion in response to adenosine diphosphate. In summary, circulating BDNF levels appear to regulate platelet activation, aggregation, and secretion through activation of a truncated TrkB receptor and downstream kinase-dependent signaling.
Collapse
|
15
|
Azoulay D, Horowitz NA. Brain-derived neurotrophic factor in hematological malignancies: From detrimental to potentially beneficial. Blood Rev 2021; 51:100871. [PMID: 34344590 DOI: 10.1016/j.blre.2021.100871] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/13/2021] [Accepted: 07/21/2021] [Indexed: 12/20/2022]
Abstract
Emerging studies have highlighted brain-derived neurotrophic factor (BDNF), a neuronal growth factor abundant in the peripheral blood, and its tyrosine kinase receptor TRKB, as onco-genes and proteins that support the survival of malignant hematological cells. In contrast, other researchers reported on a favorable association between BDNF blood levels and prognosis, chemotherapy response and neurological side effects in patients with hematological malignancies. Here, we review the accumulated data regarding the expression of BDNF and its receptors in normal hematopoietic and lymphatic cells and tissue. In addition, in-vitro experiments, animal models and human sample studies that investigated the role of BDNF and its receptors in hematological malignancies are discussed. Finally, directions for future research aimed at revealing the mechanisms underlying the protective effect of BDNF in patients with these diseases are suggested.
Collapse
Affiliation(s)
- David Azoulay
- Hematology Unit and Laboratories, Galilee Medical Center, Naharia, Israel; Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
| | - Netanel A Horowitz
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
16
|
Giraud F, Pereira E, Anizon F, Moreau P. Recent Advances in Pain Management: Relevant Protein Kinases and Their Inhibitors. Molecules 2021; 26:molecules26092696. [PMID: 34064521 PMCID: PMC8124620 DOI: 10.3390/molecules26092696] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/20/2021] [Accepted: 04/30/2021] [Indexed: 12/16/2022] Open
Abstract
The purpose of this review is to underline the protein kinases that have been established, either in fundamental approach or clinical trials, as potential biological targets in pain management. Protein kinases are presented according to their group in the human kinome: TK (Trk, RET, EGFR, JAK, VEGFR, SFK, BCR-Abl), CMGC (p38 MAPK, MEK, ERK, JNK, ASK1, CDK, CLK2, DYRK1A, GSK3, CK2), AGC (PKA, PKB, PKC, PKMζ, PKG, ROCK), CAMK, CK1 and atypical/other protein kinases (IKK, mTOR). Examples of small molecule inhibitors of these biological targets, demonstrating an analgesic effect, are described. Altogether, this review demonstrates the fundamental role that protein kinase inhibitors could play in the development of new pain treatments.
Collapse
|
17
|
Ko S, Jang WS, Jeong JH, Ahn JW, Kim YH, Kim S, Chae HK, Chung S. (-)-Gallocatechin gallate from green tea rescues cognitive impairment through restoring hippocampal silent synapses in post-menopausal depression. Sci Rep 2021; 11:910. [PMID: 33441611 PMCID: PMC7806886 DOI: 10.1038/s41598-020-79287-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 12/03/2020] [Indexed: 01/29/2023] Open
Abstract
Post-menopausal depression (PMD) is a common psychological disorder accompanied by a cognitive deficit, which is caused by a series of uncontrolled emotional disruptions by strong environmental stressors during menopause. To overcome PMD-induced cognitive deficit, Green tea has been suggested as a dietary supplement because of its ameliorating effect on cognitive dysfunction induced by normal aging or neurodegenerative syndromes; however, its clinical use to improve PMD-accompanied cognitive deficit is still limited due to the controversy for the active ingredients and ambiguous mechanism of its action. Here, we developed modified high-temperature-processed green tea extract (HTP-GTE), which showed lower neuronal toxicity than the conventional green tea extract (GTE). We also demonstrated that HTP-GTE administration prevented the development of learned helplessness (LH) in a rat post-menopausal model. Additionally, HTP-GTE improved LH-induced cognitive impairments simultaneously with rescued the long-term synaptic plasticity. This occurred via the restoration of silent synapse formation by increasing the hippocampal BDNF-tyrosine receptor kinase B pathway in the helpless ovariectomized (OVX) rats. Likewise, we also identified that (-)-gallocatechin gallate was the main contributor of the HTP-GTE effect. Our findings suggested that HTP-GTE has a potential as a preventive nutritional supplement to ameliorate cognitive dysfunctions associated with PMD.
Collapse
Affiliation(s)
- Sukjin Ko
- grid.15444.300000 0004 0470 5454Brain Korea 21 Plus Project for Medical Science, Department of Physiology, Yonsei University College of Medicine, Seoul, 03722 Republic of Korea
| | - Won Seuk Jang
- grid.15444.300000 0004 0470 5454Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722 Republic of Korea
| | - Ji-Hyun Jeong
- grid.15444.300000 0004 0470 5454Brain Korea 21 Plus Project for Medical Science, Department of Physiology, Yonsei University College of Medicine, Seoul, 03722 Republic of Korea
| | - Ji Woong Ahn
- grid.15444.300000 0004 0470 5454Brain Korea 21 Plus Project for Medical Science, Department of Physiology, Yonsei University College of Medicine, Seoul, 03722 Republic of Korea
| | - Young-Hwan Kim
- BnH Research Co., LTD., Goyang-si, Gyeonggi-do 10594 Republic of Korea
| | - Sohyun Kim
- grid.15444.300000 0004 0470 5454Brain Korea 21 Plus Project for Medical Science, Department of Physiology, Yonsei University College of Medicine, Seoul, 03722 Republic of Korea
| | - Hyeon Kyeong Chae
- BnH Research Co., LTD., Goyang-si, Gyeonggi-do 10594 Republic of Korea
| | - Seungsoo Chung
- grid.15444.300000 0004 0470 5454Brain Korea 21 Plus Project for Medical Science, Department of Physiology, Yonsei University College of Medicine, Seoul, 03722 Republic of Korea
| |
Collapse
|
18
|
Ferrini F, Salio C, Boggio EM, Merighi A. Interplay of BDNF and GDNF in the Mature Spinal Somatosensory System and Its Potential Therapeutic Relevance. Curr Neuropharmacol 2021; 19:1225-1245. [PMID: 33200712 PMCID: PMC8719296 DOI: 10.2174/1570159x18666201116143422] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/17/2020] [Accepted: 10/05/2020] [Indexed: 11/22/2022] Open
Abstract
The growth factors BDNF and GDNF are gaining more and more attention as modulators of synaptic transmission in the mature central nervous system (CNS). The two molecules undergo a regulated secretion in neurons and may be anterogradely transported to terminals where they can positively or negatively modulate fast synaptic transmission. There is today a wide consensus on the role of BDNF as a pro-nociceptive modulator, as the neurotrophin has an important part in the initiation and maintenance of inflammatory, chronic, and/or neuropathic pain at the peripheral and central level. At the spinal level, BDNF intervenes in the regulation of chloride equilibrium potential, decreases the excitatory synaptic drive to inhibitory neurons, with complex changes in GABAergic/glycinergic synaptic transmission, and increases excitatory transmission in the superficial dorsal horn. Differently from BDNF, the role of GDNF still remains to be unraveled in full. This review resumes the current literature on the interplay between BDNF and GDNF in the regulation of nociceptive neurotransmission in the superficial dorsal horn of the spinal cord. We will first discuss the circuitries involved in such a regulation, as well as the reciprocal interactions between the two factors in nociceptive pathways. The development of small molecules specifically targeting BDNF, GDNF and/or downstream effectors is opening new perspectives for investigating these neurotrophic factors as modulators of nociceptive transmission and chronic pain. Therefore, we will finally consider the molecules of (potential) pharmacological relevance for tackling normal and pathological pain.
Collapse
Affiliation(s)
- Francesco Ferrini
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
- Department of Psychiatry & Neuroscience, Université Laval, Québec, Canada
| | - Chiara Salio
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Elena M. Boggio
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
- National Institute of Neuroscience, Grugliasco, Italy
| |
Collapse
|
19
|
Emili M, Guidi S, Uguagliati B, Giacomini A, Bartesaghi R, Stagni F. Treatment with the flavonoid 7,8-Dihydroxyflavone: a promising strategy for a constellation of body and brain disorders. Crit Rev Food Sci Nutr 2020; 62:13-50. [DOI: 10.1080/10408398.2020.1810625] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Marco Emili
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Beatrice Uguagliati
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Andrea Giacomini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| |
Collapse
|
20
|
Frick C, Fink S, Schmidbauer D, Rousset F, Eickhoff H, Tropitzsch A, Kramer B, Senn P, Glueckert R, Rask-Andersen H, Wiesmüller KH, Löwenheim H, Müller M. Age-Dependency of Neurite Outgrowth in Postnatal Mouse Cochlear Spiral Ganglion Explants. Brain Sci 2020; 10:E580. [PMID: 32839381 PMCID: PMC7564056 DOI: 10.3390/brainsci10090580] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The spatial gap between cochlear implants (CIs) and the auditory nerve limits frequency selectivity as large populations of spiral ganglion neurons (SGNs) are electrically stimulated synchronously. To improve CI performance, a possible strategy is to promote neurite outgrowth toward the CI, thereby allowing a discrete stimulation of small SGN subpopulations. Brain-derived neurotrophic factor (BDNF) is effective to stimulate neurite outgrowth from SGNs. METHOD TrkB (tropomyosin receptor kinase B) agonists, BDNF, and five known small-molecule BDNF mimetics were tested for their efficacy in stimulating neurite outgrowth in postnatal SGN explants. To modulate Trk receptor-mediated effects, TrkB and TrkC ligands were scavenged by an excess of recombinant receptor proteins. The pan-Trk inhibitor K252a was used to block Trk receptor actions. RESULTS THF (7,8,3'-trihydroxyflavone) partly reproduced the BDNF effect in postnatal day 7 (P7) mouse cochlear spiral ganglion explants (SGEs), but failed to show effectiveness in P4 SGEs. During the same postnatal period, spontaneous and BDNF-stimulated neurite outgrowth increased. The increased neurite outgrowth in P7 SGEs was not caused by the TrkB/TrkC ligands, BDNF and neurotrophin-3 (NT-3). CONCLUSIONS The age-dependency of induction of neurite outgrowth in SGEs was very likely dependent on presently unidentified factors and/or molecular mechanisms which may also be decisive for the age-dependent efficacy of the small-molecule TrkB receptor agonist THF.
Collapse
Affiliation(s)
- Claudia Frick
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
- Department of Microbiome Science, Max Planck Institute for Developmental Biology, 72076 Tübingen, Germany
| | - Stefan Fink
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
| | - Dominik Schmidbauer
- Inner Ear Laboratory Innsbruck, Medical University Innsbruck, 6020 Innsbruck, Austria; (D.S.); (R.G.)
| | - Francis Rousset
- The Inner Ear & Olfaction Lab, Department of Clinical Neurosciences, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland; (F.R.); (P.S.)
| | - Holger Eickhoff
- EMC Microcollections GmbH, 72070 Tübingen, Germany; (H.E.); (K.-H.W.)
| | - Anke Tropitzsch
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
| | - Benedikt Kramer
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Mannheim, 68167 Mannheim, Germany
| | - Pascal Senn
- The Inner Ear & Olfaction Lab, Department of Clinical Neurosciences, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland; (F.R.); (P.S.)
| | - Rudolf Glueckert
- Inner Ear Laboratory Innsbruck, Medical University Innsbruck, 6020 Innsbruck, Austria; (D.S.); (R.G.)
- Tirol Kliniken Innsbruck, University Clinic of Otolaryngology, 6020 Innsbruck, Austria
| | - Helge Rask-Andersen
- Department of Surgical Sciences, Otorhinolaryngology and Head and Neck Surgery, University of Uppsala, 751 85 Uppsala, Sweden;
| | | | - Hubert Löwenheim
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
| | - Marcus Müller
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
| |
Collapse
|
21
|
NTRK2 methylation is related to reduced PTSD risk in two African cohorts of trauma survivors. Proc Natl Acad Sci U S A 2020; 117:21667-21672. [PMID: 32817534 DOI: 10.1073/pnas.2008415117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Extensive pharmacologic, genetic, and epigenetic research has linked the glucocorticoid receptor (GR) to memory processes, and to risk and symptoms of posttraumatic stress disorder (PTSD). In the present study we investigated the epigenetic pattern of 12 genes involved in the regulation of GR signaling in two African populations of heavily traumatized individuals: Survivors of the rebel war in northern Uganda (n = 463) and survivors of the Rwandan genocide (n = 350). The strongest link between regional methylation and PTSD risk and symptoms was observed for NTRK2, which encodes the transmembrane receptor tropomyosin-related kinase B, binds the brain-derived neurotrophic factor, and has been shown to play an important role in memory formation. NTRK2 methylation was not related to trauma load, suggesting that methylation differences preexisted the trauma. Because NTRK2 methylation differences were predominantly associated with memory-related PTSD symptoms, and because they seem to precede traumatic events, we next investigated the relationship between NTRK2 methylation and memory in a sample of nontraumatized individuals (n = 568). We found that NTRK2 methylation was negatively associated with recognition memory performance. Furthermore, fMRI analyses revealed NTRK2 methylation-dependent differences in brain network activity related to recognition memory. The present study demonstrates that NTRK2 is epigenetically linked to memory functions in nontraumatized subjects and to PTSD risk and symptoms in traumatized populations.
Collapse
|
22
|
Ma T, Zhang H, Xu ZP, Lu Y, Fu Q, Wang W, Li GH, Wang YY, Yang YT, Mi WD. Activation of brain-derived neurotrophic factor signaling in the basal forebrain reverses acute sleep deprivation-induced fear memory impairments. Brain Behav 2020; 10:e01592. [PMID: 32157827 PMCID: PMC7177564 DOI: 10.1002/brb3.1592] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 01/15/2020] [Accepted: 02/15/2020] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION The mechanisms underlying sleep deprivation-induced memory impairments and relevant compensatory signaling pathways remain elusive. We tested the hypothesis that increased brain-derived neurotrophic factor (BDNF) expression in the basal forebrain following acute sleep deprivation was a compensatory mechanism to maintain fear memory performance. METHODS Adult male Wistar rats were deprived of 6-hr total sleep from the beginning of the light cycle. The effects of sleep deprivation on BDNF protein expression and activation of downstream tropomyosin receptor kinase B (TrkB)/phospholipase C-γ1 (PLCγ1) signaling in the basal forebrain and fear memory consolidation were examined. BDNF or selective downstream TrkB receptor antagonist ANA-12 was further injected into the basal forebrain bilaterally to observe the changes in fear memory consolidation in response to modulation of the BDNF/TrkB signaling. RESULTS Six hours of sleep deprivation-induced both short- and long-term fear memory impairments. Increased BDNF protein expression and TrkB and PLCγ1 phosphorylation in the basal forebrain were observed after sleep deprivation. Microinjection of BDNF into the basal forebrain partly reversed fear memory deficits caused by sleep deprivation, which were accompanied by increased BDNF protein levels and TrkB/PLCγ1 activation. After ANA-12 microinjection, sleep deprivation-induced activation of the BDNF/TrkB pathway was inhibited and impairments of fear memory consolidation were further aggravated. CONCLUSIONS Acute sleep deprivation induces compensatory increase of BDNF expression in the basal forebrain. Microinjection of BDNF into the basal forebrain mitigates the fear memory impairments caused by sleep deprivation by activating TrkB/PLCγ1 signaling.
Collapse
Affiliation(s)
- Tao Ma
- Anesthesia and Operation Center, Chinese PLA Medical School, Beijing, China.,Department of Anesthesiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Hao Zhang
- Department of Anesthesiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Zhi-Peng Xu
- Anesthesia and Operation Center, Chinese PLA Medical School, Beijing, China
| | - Yan Lu
- Department of Neurology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Qiang Fu
- Anesthesia and Operation Center, Chinese PLA Medical School, Beijing, China
| | - Wei Wang
- Department of Anesthesiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Guan-Hua Li
- Department of Anesthesiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Ying-Ying Wang
- Department of Anesthesiology, PLA Rocket Force Characteristic Medical Center, Beijing, China.,PLA Rocket Force Characteristic Medical Center, Postgraduate Training Base of Jinzhou Medical University, Beijing, China
| | - Yi-Tian Yang
- Anesthesia and Operation Center, Chinese PLA Medical School, Beijing, China
| | - Wei-Dong Mi
- Anesthesia and Operation Center, Chinese PLA Medical School, Beijing, China
| |
Collapse
|
23
|
Medlej Y, Salah H, Wadi L, Saad S, Bashir B, Allam J, Atoui Z, Darwish N, Karnib N, Darwish H, Kobeissy F, Wang KKW, Hamade E, Obeid M. Lestaurtinib (CEP-701) modulates the effects of early life hypoxic seizures on cognitive and emotional behaviors in immature rats. Epilepsy Behav 2019; 92:332-340. [PMID: 30769278 DOI: 10.1016/j.yebeh.2019.01.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 11/30/2022]
Abstract
Hypoxic encephalopathy of the newborn is a major cause of long-term neurological sequelae. We have previously shown that CEP-701 (lestaurtinib), a drug with an established safety profile in children, attenuates short-term hyperexcitability and tropomyosin-related kinase B (TrkB) receptor activation in a well-established rat model of early life hypoxic seizures (HS). Here, we investigated the potential long-term neuroprotective effects of a post-HS transient CEP-701 treatment. Following exposure to global hypoxia, 10 day old male Sprague-Dawley pups received CEP-701 or its vehicle and were sequentially subjected to the light-dark box test (LDT), forced swim test (FST), open field test (OFT), Morris water maze (MWM), and the modified active avoidance (MAAV) test between postnatal days 24 and 44 (P24-44). Spontaneous seizure activity was assessed by epidural cortical electroencephalography (EEG) between P50 and 100. Neuronal density and glial fibrillary acidic protein (GFAP) levels were evaluated on histological sections in the hippocampus, amygdala, and prefrontal cortex at P100. Vehicle-treated hypoxic rats exhibited significantly increased immobility in the FST compared with controls, and post-HS CEP-701 administration reversed this HS-induced depressive-like behavior (p < 0.05). In the MAAV test, CEP-701-treated hypoxic rats were slower at learning both context-cued and tone-signaled shock-avoidance behaviors (p < 0.05). All other behavioral outcomes were comparable, and no recurrent seizures, neuronal loss, or increase in GFAP levels were detected in any of the groups. We showed that early life HS predispose to long-lasting depressive-like behaviors, and that these are prevented by CEP-701, likely via TrkB modulation. Future mechanistically more specific studies will further investigate the potential role of TrkB signaling pathway modulation in achieving neuroprotection against neonatal HS, without causing neurodevelopmental adverse effects.
Collapse
Affiliation(s)
- Yasser Medlej
- Department of Anatomy, Cell biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Houssein Salah
- Department of Anatomy, Cell biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Lara Wadi
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Sarah Saad
- Faculty of Arts and Sciences, American University of Beirut, Lebanon
| | - Bashir Bashir
- Faculty of Arts and Sciences, American University of Beirut, Lebanon
| | - Jad Allam
- Faculty of Arts and Sciences, American University of Beirut, Lebanon
| | - Zahraa Atoui
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nora Darwish
- Faculty of Arts and Sciences, American University of Beirut, Lebanon
| | - Nabil Karnib
- Department of Anatomy, Cell biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Lebanon
| | - Hala Darwish
- Department of Anatomy, Cell biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Rafic Hariri School of Nursing, American University of Beirut, Beirut, Lebanon
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Program for Neurotrauma, Neuroproteomics, Department of Emergency Medicine, Department of Chemistry, Department of Neuroscience, and Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics, Department of Emergency Medicine, Department of Chemistry, Department of Neuroscience, and Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Eva Hamade
- Department of Biochemistry, Faculty of Science, Lebanese University, Lebanon
| | - Makram Obeid
- Department of Anatomy, Cell biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Division of Child Neurology, Department of Pediatric and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon.
| |
Collapse
|
24
|
Meng L, Liu B, Ji R, Jiang X, Yan X, Xin Y. Targeting the BDNF/TrkB pathway for the treatment of tumors. Oncol Lett 2018; 17:2031-2039. [PMID: 30675270 DOI: 10.3892/ol.2018.9854] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 12/12/2018] [Indexed: 12/12/2022] Open
Abstract
Neurotrophins are a family of growth factors that regulate neural survival, development, function and plasticity in the central and the peripheral nervous system. There are four neurotrophins: nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3) and NT-4. Among them, BDNF is the most studied due to its high expression in the brain. Over the past two decades, BDNF and its receptor tropomyosin receptor kinase B (TrkB) have been reported to be upregulated in a wide range of tumors. This activated signal stimulates a series of downstream pathways, including phosphoinositide 3-kinase/protein kinase B, Ras-Raf-mitogen activated protein kinase kinase-extracellular signal-regulated kinases, the phospholipase-C-γ pathway and the transactivation of epidermal growth factor receptor. Activation of these signaling pathways induces oncogenic effects by increasing cancer cell growth, proliferation, survival, migration and epithelial to mesenchymal transition, and decreasing anoikis, relapse and chemotherapeutic sensitivity. The present review summarizes recent findings to discuss the role of BDNF in tumors, the underlying molecular mechanism, targeting Trk receptors for treatment of cancers and its potential risk.
Collapse
Affiliation(s)
- Lingbin Meng
- Department of Internal Medicine, Florida Hospital, Orlando, FL 32803, USA
| | - Baoqiong Liu
- Department of Internal Medicine, Florida Hospital, Orlando, FL 32803, USA
| | - Rui Ji
- Department of Biology, Valencia College, Orlando, FL 32825, USA
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xuebo Yan
- Department of Respiratory Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
25
|
Okamura R, Boichard A, Kato S, Sicklick JK, Bazhenova L, Kurzrock R. Analysis of NTRK Alterations in Pan-Cancer Adult and Pediatric Malignancies: Implications for NTRK-Targeted Therapeutics. JCO Precis Oncol 2018; 2018. [PMID: 30637364 DOI: 10.1200/po.18.00183] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose Fusions that involve neurotrophic-tropomyosin receptor kinase (NTRK) genes are known drivers of oncogenesis. Therapies that target these ultra-rare, constitutionally active NTRK fusions have been remarkably effective. Herein, we analyze the prevalence of the full array of NTRK alterations-fusions, mutations, copy number alterations, and increased transcript expression-in diverse adult and pediatric tumor types to understand the landscape of NTRK aberrations in cancer. Methods We assessed 13,467 samples available from The Cancer Genome Atlas (adult tumors) and the St Jude PeCan database (pediatric tumors) for the prevalence of NTRK fusions, as well as associated genomic and transcriptomic co-aberrations in different tumor types. Results NTRK fusions were observed in 0.31% of adult tumors and in 0.34% of pediatric tumors. The most common gene partners were NTRK3 (0.16% of adult tumors) followed by NTRK1 (0.14% of pediatric tumors). NTRK fusions were found more commonly in pediatric melanoma (11.1% of samples), pediatric glioma (3.97%), and adult thyroid cancers (2.34%). Additional genomic and transcriptomic NTRK alterations- mutation, amplification, and mRNA overexpression-occurred in 14.2% of samples, whereas the frequency of alterations that implicated NTRK ligands and the NTRK co-receptor (p75NTR) ranged from 3.8% to 5.4%. Among 31 adult samples carrying NTRK fusions, co-alterations occurred often and usually involved the downstream phosphoinositide-3-kinase signaling pathway, cell-cycle machinery, other tyrosine-kinase receptors, and mitogen-activated protein kinase signals. Conclusion Whereas NTRK fusions are exceedingly rare, other NTRK abnormalities affect 14% of patients with cancer. Affecting these alterations has not yet been achievable in cancer. Genomic co-alterations occur frequently with NTRK fusions, but it is not known if co-targeting them can attenuate primary or secondary resistance to NTRK inhibitors.
Collapse
Affiliation(s)
- Ryosuke Okamura
- University of California, San Diego, Moores Cancer Center, La Jolla, CA
| | - Amélie Boichard
- University of California, San Diego, Moores Cancer Center, La Jolla, CA
| | - Shumei Kato
- University of California, San Diego, Moores Cancer Center, La Jolla, CA
| | - Jason K Sicklick
- University of California, San Diego, Moores Cancer Center, La Jolla, CA
| | | | - Razelle Kurzrock
- University of California, San Diego, Moores Cancer Center, La Jolla, CA
| |
Collapse
|
26
|
Zhang H, Zhang C, Vincent J, Zala D, Benstaali C, Sainlos M, Grillo-Bosch D, Daburon S, Coussen F, Cho Y, David DJ, Saudou F, Humeau Y, Choquet D. Modulation of AMPA receptor surface diffusion restores hippocampal plasticity and memory in Huntington's disease models. Nat Commun 2018; 9:4272. [PMID: 30323233 PMCID: PMC6189172 DOI: 10.1038/s41467-018-06675-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/19/2018] [Indexed: 11/10/2022] Open
Abstract
Impaired hippocampal synaptic plasticity contributes to cognitive impairment in Huntington’s disease (HD). However, the molecular basis of such synaptic plasticity defects is not fully understood. Combining live-cell nanoparticle tracking and super-resolution imaging, we show that AMPAR surface diffusion, a key player in synaptic plasticity, is disturbed in various rodent models of HD. We demonstrate that defects in the brain-derived neurotrophic factor (BDNF)–tyrosine receptor kinase B (TrkB) signaling pathway contribute to the deregulated AMPAR trafficking by reducing the interaction between transmembrane AMPA receptor regulatory proteins (TARPs) and the PDZ-domain scaffold protein PSD95. The disturbed AMPAR surface diffusion is rescued by the antidepressant drug tianeptine via the BDNF signaling pathway. Tianeptine also restores the impaired LTP and hippocampus-dependent memory in different HD mouse models. These findings unravel a mechanism underlying hippocampal synaptic and memory dysfunction in HD, and highlight AMPAR surface diffusion as a promising therapeutic target. Cognitive decline in Huntington’s disease (HD) may be due to impaired hippocampal synaptic plasticity. In this study the authors show that AMPA receptor surface diffusion, a key player in synaptic plasticity, is deregulated in multiple HD mouse models as a result of impaired BDNF signalling that underlies the memory deficits, and can be pharmacologically rescued.
Collapse
Affiliation(s)
- Hongyu Zhang
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, 33076, France. .,Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique (CNRS) UMR 5297, Bordeaux, 33076, France. .,Department of Biomedicine, KG Jebsen Centre for Research on Neuropsychiatric Disorders, University of Bergen, Jonas Lies vei 91, N-5009, Bergen, Norway.
| | - Chunlei Zhang
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, 33076, France.,Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique (CNRS) UMR 5297, Bordeaux, 33076, France
| | - Jean Vincent
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, 33076, France.,Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique (CNRS) UMR 5297, Bordeaux, 33076, France
| | - Diana Zala
- Institut Curie, CNRS, UMR3306, Inserm, U1005, F-91405, Orsay, France.,INSERM U894, Center of Psychiatry and Neuroscience, Paris, France, University Paris-Descartes, Paris, 75006, France
| | - Caroline Benstaali
- Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, F-38000, Grenoble, France.,INSERM, U1216, F-38000, Grenoble, France
| | - Matthieu Sainlos
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, 33076, France.,Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique (CNRS) UMR 5297, Bordeaux, 33076, France
| | - Dolors Grillo-Bosch
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, 33076, France.,Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique (CNRS) UMR 5297, Bordeaux, 33076, France
| | - Sophie Daburon
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, 33076, France.,Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique (CNRS) UMR 5297, Bordeaux, 33076, France
| | - Françoise Coussen
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, 33076, France.,Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique (CNRS) UMR 5297, Bordeaux, 33076, France
| | - Yoon Cho
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, University of Bordeaux, Bordeaux, 33000, France
| | - Denis J David
- Université Paris-Saclay, Univ. Paris-Sud, Faculté de Pharmacie, CESP, INSERM UMRS1178, Chatenay-Malabry, 92296, France
| | - Frederic Saudou
- Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, F-38000, Grenoble, France. .,INSERM, U1216, F-38000, Grenoble, France. .,CHU Grenoble Alpes, F-38000, Grenoble, France.
| | - Yann Humeau
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, 33076, France.,Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique (CNRS) UMR 5297, Bordeaux, 33076, France
| | - Daniel Choquet
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, 33076, France. .,Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique (CNRS) UMR 5297, Bordeaux, 33076, France. .,Bordeaux Imaging Center, CNRS UMS 3420, University of Bordeaux, INSERM US04, 33076, Bordeaux, France.
| |
Collapse
|
27
|
Retamal J, Reyes A, Ramirez P, Bravo D, Hernandez A, Pelissier T, Villanueva L, Constandil L. Burst-Like Subcutaneous Electrical Stimulation Induces BDNF-Mediated, Cyclotraxin B-Sensitive Central Sensitization in Rat Spinal Cord. Front Pharmacol 2018; 9:1143. [PMID: 30364099 PMCID: PMC6191473 DOI: 10.3389/fphar.2018.01143] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/21/2018] [Indexed: 11/18/2022] Open
Abstract
Intrathecal administration of brain derived neurotrophic factor (BDNF) induces long-term potentiation (LTP) and generates long-lasting central sensitization in spinal cord thus mimicking chronic pain, but the relevance of these observations to chronic pain mechanisms is uncertain. Since C-fiber activation by a high-frequency subcutaneous electrical stimulation (SES) protocol causes spinal release of BDNF and induces spinal cord LTP, we propose that application of such protocol would be a sufficient condition for generating long-lasting BDNF-mediated central sensitization. Results showed that application of burst-like SES to rat toes produced (i) rapid induction of hyperalgesia that lasted for more than 3 weeks, (ii) early increase of C-reflex activity followed by increased wind-up scores lasting for more than 1 week, and (iii) early increase followed by late decrease in BDNF protein levels and phosphorylated TrkB that lasted for more than 1 week. These changes were prevented by the TrkB antagonist cyclotraxin-B administered shortly before SES, while hyperalgesia was reversed by cyclotraxin-B administered 3 days after SES. Results suggest that mechanisms underlying central sensitization first involve BDNF release of probably neuronal origin, followed by brief increased expression of likely glial BDNF and pTrkB that could switch early phase sensitization into late one.
Collapse
Affiliation(s)
- Jeffri Retamal
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile.,Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Santiago, Chile
| | - Andrea Reyes
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Paulina Ramirez
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile.,Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Santiago, Chile
| | - David Bravo
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile.,Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Santiago, Chile
| | - Alejandro Hernandez
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Teresa Pelissier
- Program of Molecular and Clinical Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Luis Villanueva
- Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Paris, France
| | - Luis Constandil
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile.,Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Santiago, Chile
| |
Collapse
|
28
|
Shaib AH, Staudt A, Harb A, Klose M, Shaaban A, Schirra C, Mohrmann R, Rettig J, Becherer U. Paralogs of the Calcium-Dependent Activator Protein for Secretion Differentially Regulate Synaptic Transmission and Peptide Secretion in Sensory Neurons. Front Cell Neurosci 2018; 12:304. [PMID: 30254567 PMCID: PMC6141663 DOI: 10.3389/fncel.2018.00304] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/21/2018] [Indexed: 12/29/2022] Open
Abstract
The two paralogs of the calcium-dependent activator protein for secretion (CAPS) are priming factors for synaptic vesicles (SVs) and neuropeptide containing large dense-core vesicles (LDCVs). Yet, it is unclear whether CAPS1 and CAPS2 regulate exocytosis of these two vesicle types differentially in dorsal root ganglion (DRG) neurons, wherein synaptic transmission and neuropeptide release are of equal importance. These sensory neurons transfer information from the periphery to the spinal cord (SC), releasing glutamate as the primary neurotransmitter, with co-transmission via neuropeptides in a subset of so called peptidergic neurons. Neuropeptides are key components of the information-processing machinery of pain perception and neuropathic pain generation. Here, we compared the ability of CAPS1 and CAPS2 to support priming of both vesicle types in single and double knock-out mouse (DRG) neurons using a variety of high-resolution live cell imaging methods. While CAPS1 was localized to synapses of all DRG neurons and promoted synaptic transmission, CAPS2 was found exclusively in peptidergic neurons and mediated LDCV exocytosis. Intriguingly, ectopic expression of CAPS2 empowered non-peptidergic neurons to drive LDCV fusion, thereby identifying CAPS2 as an essential molecular determinant for peptidergic signaling. Our results reveal that these distinct functions of both CAPS paralogs are based on their differential subcellular localization in DRG neurons. Our data suggest a major role for CAPS2 in neuropathic pain via control of neuropeptide release.
Collapse
Affiliation(s)
- Ali H. Shaib
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Angelina Staudt
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Ali Harb
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
- ZHMB Junior Group, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Margarete Klose
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Ahmed Shaaban
- ZHMB Junior Group, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Claudia Schirra
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Ralf Mohrmann
- ZHMB Junior Group, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Jens Rettig
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Ute Becherer
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| |
Collapse
|
29
|
Levy MJF, Boulle F, Steinbusch HW, van den Hove DLA, Kenis G, Lanfumey L. Neurotrophic factors and neuroplasticity pathways in the pathophysiology and treatment of depression. Psychopharmacology (Berl) 2018; 235:2195-2220. [PMID: 29961124 PMCID: PMC6061771 DOI: 10.1007/s00213-018-4950-4] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/18/2018] [Indexed: 02/06/2023]
Abstract
Depression is a major health problem with a high prevalence and a heavy socioeconomic burden in western societies. It is associated with atrophy and impaired functioning of cortico-limbic regions involved in mood and emotion regulation. It has been suggested that alterations in neurotrophins underlie impaired neuroplasticity, which may be causally related to the development and course of depression. Accordingly, mounting evidence suggests that antidepressant treatment may exert its beneficial effects by enhancing trophic signaling on neuronal and synaptic plasticity. However, current antidepressants still show a delayed onset of action, as well as lack of efficacy. Hence, a deeper understanding of the molecular and cellular mechanisms involved in the pathophysiology of depression, as well as in the action of antidepressants, might provide further insight to drive the development of novel fast-acting and more effective therapies. Here, we summarize the current literature on the involvement of neurotrophic factors in the pathophysiology and treatment of depression. Further, we advocate that future development of antidepressants should be based on the neurotrophin theory.
Collapse
Affiliation(s)
- Marion J F Levy
- Centre de Psychiatrie et Neurosciences (Inserm U894), Université Paris Descartes, 102-108 rue de la santé, 75014, Paris, France
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Fabien Boulle
- Centre de Psychiatrie et Neurosciences (Inserm U894), Université Paris Descartes, 102-108 rue de la santé, 75014, Paris, France
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Harry W Steinbusch
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Daniël L A van den Hove
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Gunter Kenis
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Laurence Lanfumey
- Centre de Psychiatrie et Neurosciences (Inserm U894), Université Paris Descartes, 102-108 rue de la santé, 75014, Paris, France.
- EURON-European Graduate School of Neuroscience, Maastricht, The Netherlands.
| |
Collapse
|
30
|
Dheer Y, Chitranshi N, Gupta V, Abbasi M, Mirzaei M, You Y, Chung R, Graham SL, Gupta V. Bexarotene Modulates Retinoid-X-Receptor Expression and Is Protective Against Neurotoxic Endoplasmic Reticulum Stress Response and Apoptotic Pathway Activation. Mol Neurobiol 2018; 55:9043-9056. [PMID: 29637440 DOI: 10.1007/s12035-018-1041-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 03/27/2018] [Indexed: 12/21/2022]
Abstract
Retinoid X-receptors (RXRs) are members of the ligand-dependent transcription factor family of nuclear receptors that have gained recent research focus as potential targets for neurodegenerative disorders. Bexarotene is an RXR pharmacological agonist that is shown to be neuroprotective through its effects in promoting amyloid beta (Aβ) uptake by the glial cells in the brain. This study aimed to evaluate the dose-dependent effects of bexarotene on RXR expression in SH-SY5Y neuroblastoma cells and validate the drug effects in the brain in vivo. The protein expression studies were carried out using a combination of various drug treatment paradigms followed by expression analysis using Western blotting and immunofluorescence. Our study demonstrated that bexarotene promoted the expression of RXR α, β and γ isoforms at optimal concentrations in the cells and in the mice brain. Interestingly, a decreased RXR expression was identified in Alzheimer's disease mouse model and in the cells that were treated with Aβ. Bexarotene treatment not only rescued the RXR expression loss caused by Aβ treatment (p < 0.05) but also protected the cells against Aβ-induced ER stress (p < 0.05) and pro-apoptotic BAD protein activation (p < 0.05). In contrast, higher concentrations of bexarotene upregulated the ER stress proteins and led to BAD activation. Our study revealed that these downstream neurotoxic effects of high drug concentrations could be prevented by pharmacological targeting of the TrkB receptor. The ER stress and BAD activation induced by high concentrations of bexarotene were rescued by the TrkB agonist, 7,8 dihydroxyflavone (p < 0.05) while TrkB inhibitor CTX-B treatment further exacerbated these effects. Together, these findings suggest a cross-talk of TrkB signalling with downstream effects of bexarotene toxicity and indicate that therapeutic targeting of RXRs could prevent the Aβ-induced molecular neurotoxic effects.
Collapse
Affiliation(s)
- Yogita Dheer
- Faculty of Medicine and Health Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia.
| | - Nitin Chitranshi
- Faculty of Medicine and Health Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Veer Gupta
- School of Medical Sciences, Edith Cowan University, Perth, Australia
| | - Mojdeh Abbasi
- Faculty of Medicine and Health Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Mehdi Mirzaei
- Department of Chemistry and Biomolecular Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Yuyi You
- Save Sight Institute, Sydney University, Sydney, NSW, 2000, Australia
| | - Roger Chung
- Faculty of Medicine and Health Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| | - Stuart L Graham
- Faculty of Medicine and Health Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia.,Save Sight Institute, Sydney University, Sydney, NSW, 2000, Australia
| | - Vivek Gupta
- Faculty of Medicine and Health Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW, 2109, Australia
| |
Collapse
|
31
|
Merk DJ, Ohli J, Merk ND, Thatikonda V, Morrissy S, Schoof M, Schmid SN, Harrison L, Filser S, Ahlfeld J, Erkek S, Raithatha K, Andreska T, Weißhaar M, Launspach M, Neumann JE, Shakarami M, Plenker D, Marra MA, Li Y, Mungall AJ, Moore RA, Ma Y, Jones SJM, Lutz B, Ertl-Wagner B, Rossi A, Wagener R, Siebert R, Jung A, Eberhart CG, Lach B, Sendtner M, Pfister SM, Taylor MD, Chavez L, Kool M, Schüller U. Opposing Effects of CREBBP Mutations Govern the Phenotype of Rubinstein-Taybi Syndrome and Adult SHH Medulloblastoma. Dev Cell 2018; 44:709-724.e6. [PMID: 29551561 DOI: 10.1016/j.devcel.2018.02.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/08/2018] [Accepted: 02/12/2018] [Indexed: 10/17/2022]
Abstract
Recurrent mutations in chromatin modifiers are specifically prevalent in adolescent or adult patients with Sonic hedgehog-associated medulloblastoma (SHH MB). Here, we report that mutations in the acetyltransferase CREBBP have opposing effects during the development of the cerebellum, the primary site of origin of SHH MB. Our data reveal that loss of Crebbp in cerebellar granule neuron progenitors (GNPs) during embryonic development of mice compromises GNP development, in part by downregulation of brain-derived neurotrophic factor (Bdnf). Interestingly, concomitant cerebellar hypoplasia was also observed in patients with Rubinstein-Taybi syndrome, a congenital disorder caused by germline mutations of CREBBP. By contrast, loss of Crebbp in GNPs during postnatal development synergizes with oncogenic activation of SHH signaling to drive MB growth, thereby explaining the enrichment of somatic CREBBP mutations in SHH MB of adult patients. Together, our data provide insights into time-sensitive consequences of CREBBP mutations and corresponding associations with human diseases.
Collapse
Affiliation(s)
- Daniel J Merk
- Center for Neuropathology, Ludwig-Maximilians-University, 81377 Munich, Germany; Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Neurobiology, Harvard Medical School, Boston, MA 02215, USA
| | - Jasmin Ohli
- Center for Neuropathology, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Natalie D Merk
- Munich Center for Integrated Protein Science at the Chemistry Department, Technical University, 85747 Munich, Germany
| | - Venu Thatikonda
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sorana Morrissy
- Arthur and Sonia Labatt Brain Tumour Research Centre and Division of Neurosurgery, Hospital for Sick Children (HSC), Toronto, ON M5G 1L7, Canada; Program in Developmental and Stem Cell Biology, HSC, Toronto, ON M5G 1X8, Canada
| | - Melanie Schoof
- Research Institute Children's Cancer Center Hamburg, Martinistrasse 52, N63 (HPI), Hamburg 20251, Germany
| | - Susanne N Schmid
- Center for Neuropathology, Ludwig-Maximilians-University, 81377 Munich, Germany; Department of Neuropathology, University Medical Center Göttingen, 37099 Göttingen, Germany
| | - Luke Harrison
- Center for Neuropathology, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Severin Filser
- German Center for Neurodegenerative Diseases (DZNE), Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Julia Ahlfeld
- Center for Neuropathology, Ludwig-Maximilians-University, 81377 Munich, Germany; Division of Clinical Pharmacology, Department of Internal Medicine IV, Ludwig-Maximilians-University, 80337 Munich, Germany
| | - Serap Erkek
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; European Molecular Biology Laboratory (EMBL), Genome Biology Unit, 69117 Heidelberg, Germany; German Cancer Consortium (DKTK), Core Center Heidelberg, 69120 Heidelberg, Germany
| | - Kaamini Raithatha
- Microarray and Deep-Sequencing Core Facility, University Medical Center Göttingen, 37077 Göttingen, Germany
| | - Thomas Andreska
- Institute for Clinical Neurobiology, University of Würzburg, 97078 Würzburg, Germany
| | - Marc Weißhaar
- Center for Neuropathology, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Michael Launspach
- Center for Neuropathology, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Julia E Neumann
- Center for Neuropathology, Ludwig-Maximilians-University, 81377 Munich, Germany; Institute of Neuropathology, University Medical Center, Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Mehdi Shakarami
- Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Dennis Plenker
- Department of Translational Genomics, University of Cologne, 50931 Cologne, Germany
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC VSZ 4S6, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Yisu Li
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC VSZ 4S6, Canada
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC VSZ 4S6, Canada
| | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC VSZ 4S6, Canada
| | - Yussanne Ma
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC VSZ 4S6, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC VSZ 4S6, Canada
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, 55128 Mainz, Germany
| | - Birgit Ertl-Wagner
- Institute of Clinical Radiology, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Andrea Rossi
- Department of Pediatric Neuroradiology, Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Rabea Wagener
- Institute of Human Genetics, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany; Institute for Human Genetics, Ulm University and Ulm University Medical Center, 89081 Ulm, Germany
| | - Reiner Siebert
- Institute of Human Genetics, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany; Institute for Human Genetics, Ulm University and Ulm University Medical Center, 89081 Ulm, Germany
| | - Andreas Jung
- Institute of Pathology, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Charles G Eberhart
- Division of Neuropathology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Boleslaw Lach
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Michael Sendtner
- Institute for Clinical Neurobiology, University of Würzburg, 97078 Würzburg, Germany
| | - Stefan M Pfister
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; German Cancer Consortium (DKTK), Core Center Heidelberg, 69120 Heidelberg, Germany; Department of Pediatric Hematology and Oncology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Michael D Taylor
- Arthur and Sonia Labatt Brain Tumour Research Centre and Division of Neurosurgery, Hospital for Sick Children (HSC), Toronto, ON M5G 1L7, Canada; Program in Developmental and Stem Cell Biology, HSC, Toronto, ON M5G 1X8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Lukas Chavez
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Marcel Kool
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; German Cancer Consortium (DKTK), Core Center Heidelberg, 69120 Heidelberg, Germany
| | - Ulrich Schüller
- Center for Neuropathology, Ludwig-Maximilians-University, 81377 Munich, Germany; Research Institute Children's Cancer Center Hamburg, Martinistrasse 52, N63 (HPI), Hamburg 20251, Germany; Institute of Neuropathology, University Medical Center, Hamburg-Eppendorf, 20246 Hamburg, Germany; Department of Pediatric Hematology and Oncology, University Medical Center, Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
32
|
Totoson P, Pedard M, Marie C, Demougeot C. Activation of endothelial TrkB receptors induces relaxation of resistance arteries. Vascul Pharmacol 2018; 106:46-53. [PMID: 29471140 DOI: 10.1016/j.vph.2018.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 02/07/2018] [Accepted: 02/17/2018] [Indexed: 12/18/2022]
Abstract
While brain-derived neurotrophic factor (BDNF) was previously reported to induce relaxation of conduit artery, whether the BDNF/TrkB (tropomyosin-related kinase) pathway is involved in the tone control of resistance arteries is not known. This study investigated TrkB receptors levels/localization and the vasomotor effect of the TrkB receptor agonist LM22A-4 in isolated third-order mesenteric arteries from rats. Immunostaining revealed the presence of both full-length and truncated TrkB receptors, especially at the endothelial level. By using wire myography, LM22A-4 induced vascular relaxation that was significantly decreased by cyclotraxin B as a non-competitive TrkB antagonist and fully prevented by endothelium removal. Inhibitors of NO, EDHF, PGI2 production and the PI3K/Akt pathways separately reduced LM22A-4 induced-relaxation. By contrast, inhibition of Raf/MEK, PLCγ and CaM/CaMKII pathways did not change the relaxant effect of LM22A-4. Interestingly, BDNF also induced an endothelium and TrkB-dependent relaxation. These results indicate that endothelial TrkB activation results in the relaxation of resistance vessels via PI3K/Akt-induced eNOS phosphorylation and production of EDHF and PGI2. These data are consistent with the contribution of the endothelial BDNF/TrkB pathway to the regulation of peripheral vascular tone. They also validate the use of LM22A-4 as a reliable pharmacological agent for studying the vascular effect of BDNF.
Collapse
Affiliation(s)
- P Totoson
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25030 Besançon, France
| | - M Pedard
- INSERM UMR 1093-CAPS, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France
| | - C Marie
- INSERM UMR 1093-CAPS, Univ. Bourgogne Franche-Comté, F-21000 Dijon, France.
| | - C Demougeot
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25030 Besançon, France
| |
Collapse
|
33
|
Zhang Y, Ji F, Wang G, He D, Yang L, Zhang M. BDNF Activates mTOR to Upregulate NR2B Expression in the Rostral Anterior Cingulate Cortex Required for Inflammatory Pain-Related Aversion in Rats. Neurochem Res 2018; 43:681-691. [PMID: 29353374 DOI: 10.1007/s11064-018-2470-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/02/2018] [Accepted: 01/08/2018] [Indexed: 12/11/2022]
Abstract
The mechanistic target of rapamycin (mTOR) has been demonstrated to mediate pain-related aversion induced by formalin in the rostral anterior cingulate cortex (rACC). However, it remains unclear the signaling pathways and regulatory proteins involved. In the rACC, brain-derived neurotrophic factor (BDNF), an activity-dependent neuromodulator, has been shown to play a role in the development and persistence of chronic pain. In this study, we used a rat formalin-induced inflammatory pain model to demonstrate BDNF up-regulation in the rACC. Stimulation with exogenous BDNF up-regulated mTOR, whilst cyclotraxin B (CTX-B), a tropomyosin receptor kinase B (TrkB) antagonist, down-regulated mTOR. Our results suggest BDNF could activate an mTOR signaling pathway. Subsequently, we used formalin-induced conditioned place avoidance (F-CPA) training in rat models to investigate if mTOR activation was required for pain-related aversion. We demonstrated that BDNF/mTOR signaling could activate the NMDA receptor subunit episilon-2 (NR2B), which is required for F-CPA. Our results reveal that BDNF activates mTOR to up-regulate NR2B expression, which is required for inflammatory pain-related aversion in the rACC of rats.
Collapse
Affiliation(s)
- Yuangui Zhang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
- Department of Anesthesiology, Weifang People's Hospital, Weifang, China
| | - Fanceng Ji
- Department of Anesthesiology, Weifang People's Hospital, Weifang, China
| | - Gongming Wang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Dong He
- Shandong University, Jinan, China
| | - Le Yang
- Shandong University, Jinan, China
| | - Mengyuan Zhang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.
| |
Collapse
|
34
|
Wang X, Zhang L, Zhan Y, Li D, Zhang Y, Wang G, Zhang M. Contribution of BDNF/TrkB signalling in the rACC to the development of pain-related aversion via activation of ERK in rats with spared nerve injury. Brain Res 2017; 1671:111-120. [DOI: 10.1016/j.brainres.2017.07.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 07/12/2017] [Accepted: 07/14/2017] [Indexed: 01/02/2023]
|
35
|
de Moraes JK, Wagner VP, Fonseca FP, Vargas PA, de Farias CB, Roesler R, Martins MD. Uncovering the role of brain-derived neurotrophic factor/tyrosine kinase receptor B signaling in head and neck malignancies. J Oral Pathol Med 2017. [PMID: 28650560 DOI: 10.1111/jop.12611] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin family of growth factors that was first known as responsible for sustain the growth, function, and plasticity of neural cells. BDNF exerts its effects by binding to the tyrosine kinase receptor B (TrkB). The BDNF/TrkB axis has been reported to be overexpressed in several neurogenic and non-neurogenic tumors. Its higher expression was associated with a poor prognosis to patients affected by different human malignancies, tumor growth, invasion, and metastasis; epithelial-mesenchymal transition and resistance to chemotherapy. BDNF/TrkB represent promising targets to the development of novel anticancer therapies. Some clinical trials are currently evaluating the efficacy of Trk protein-target drugs in different types of solid tumors. To date, few groups have evaluated the DNF/TrkB pathway in head and neck malignancies. The aims of this study were to review the literature concerning the role of BDNF/TrkB activation in head and neck squamous cell carcinoma and malignant salivary gland tumors and to discuss future perspectives of BDNF/TrkB-target therapy.
Collapse
Affiliation(s)
- Juliana Kern de Moraes
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil
| | - Vivian Petersen Wagner
- Department of Pathology, School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Felipe Paiva Fonseca
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Pablo Agustin Vargas
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil
| | - Caroline Brunetto de Farias
- Cancer and Neurobiology Laboratory, Experimental Research Center, Porto Alegre Clinical Hospital, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,Children's Cancer Institute, Porto Alegre, Brazil
| | - Rafael Roesler
- Cancer and Neurobiology Laboratory, Experimental Research Center, Porto Alegre Clinical Hospital, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,Department of Pharmacology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Manoela Domingues Martins
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil.,Department of Pathology, School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,Experimental Pathology Unit, Hospital de Clinicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
36
|
Systemic Delivery of a Brain-Penetrant TrkB Antagonist Reduces Cocaine Self-Administration and Normalizes TrkB Signaling in the Nucleus Accumbens and Prefrontal Cortex. J Neurosci 2017; 36:8149-59. [PMID: 27488635 DOI: 10.1523/jneurosci.2711-14.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 06/10/2016] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED Cocaine exposure alters brain-derived neurotrophic factor (BDNF) expression in the brain. BDNF signaling through TrkB receptors differentially modulates cocaine self-administration, depending on the brain regions involved. In the present study, we determined how brain-wide inhibition of TrkB signaling affects cocaine intake, the motivation for the drug, and reinstatement of drug taking after extinction. To overcome the inability of TrkB ligands to cross the blood-brain barrier, the TrkB antagonist cyclotraxin-B was fused to the nontoxic transduction domain of the tat protein from human immunodeficiency virus type 1 (tat-cyclotraxin-B). Intravenous injection of tat-cyclotraxin-B dose-dependently reduced cocaine intake, motivation for cocaine (as measured under a progressive ratio schedule of reinforcement), and reinstatement of cocaine taking in rats allowed either short or long access to cocaine self-administration. In contrast, the treatment did not affect operant responding for a highly palatable sweet solution, demonstrating that the effects of tat-cyclotraxin-B are specific for cocaine reinforcement. Cocaine self-administration increased TrkB signaling and activated the downstream Akt pathway in the nucleus accumbens, and had opposite effects in the prefrontal cortex. Pretreatment with tat-cyclotraxin-B normalized protein levels in these two dopamine-innervated brain regions. Cocaine self-administration also increased TrkB signaling in the ventral tegmental area, where the dopaminergic projections originate, but pretreatment with tat-cyclotraxin-B did not alter this effect. Altogether, our data show that systemic administration of a brain-penetrant TrkB antagonist leads to brain region-specific effects and may be a potential pharmacological strategy for the treatment of cocaine addiction. SIGNIFICANCE STATEMENT Brain-derived neurotrophic factor (BDNF) signaling through TrkB receptors plays a well established role in cocaine reinforcement. However, local manipulation of BDNF signaling yields divergent effects, depending on the brain region, thereby questioning the viability of systemic TrkB targeting for the treatment of cocaine use disorders. Our study provides first-time evidence that systemic administration of a brain-penetrant TrkB antagonist (tat-cyclotraxin-B) reduces several behavioral measures of cocaine dependence, without altering motor performance or reinforcement by a sweet palatable solution. In addition, although cocaine self-administration produced opposite effects on TrkB signaling in the nucleus accumbens and prefrontal cortex, tat-cyclotraxin-B administration normalized these cocaine-induced changes in both brain regions.
Collapse
|
37
|
Prince CS, Maloyan A, Myatt L. Tropomyosin Receptor Kinase B Agonist, 7,8-Dihydroxyflavone, Improves Mitochondrial Respiration in Placentas From Obese Women. Reprod Sci 2017; 25:452-462. [PMID: 28677406 DOI: 10.1177/1933719117716776] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Maternal obesity negatively impacts the placenta, being associated with increased inflammation, decreased mitochondrial respiration, decreased expression of brain-derived neurotrophic factor (BDNF), and its receptor, tropomyosin receptor kinase B (TRKB). TRKB induction by 7,8-dihydroxyflavone (7,8-DHF) improves energy expenditure in an obesity animal model. We hypothesized that TRKB activation would improve mitochondrial respiration in trophoblasts from placentas of obese women. Placentas were collected from lean (pre-pregnancy BMI < 25) and obese (pre-pregnancy BMI > 30) women at term following cesarean section delivery without labor. Cytotrophoblasts were isolated and plated, permitting syncytialization. At 72 hours, syncytiotrophoblasts (STs) were treated for 1 hour with 7,8-DHF (10 nM-10 M), TRKB antagonists (ANA-12 (10 nM-1 M), Cyclotraxin B (1 nM-1M)), or vehicle. Mitochondrial respiration was measured using the XF24 Extracellular Flux Analyzer. TRKB, MAPK, and PGC1α were measured using Western blotting. Maternal obesity was associated with decreased mitochondrial respiration in STs; however, 7,8-DHF increased basal, ATP-coupled, maximal, spare capacity, and nonmitochondrial respiration. A 10 μM dose of 7,8-DHF reduced spare capacity in STs from lean women, with no effect on other respiration parameters. 7,8-DHF had no effect on TRKB phosphorylation; however, there was a concentration-dependent decrease of p38 MAPK phosphorylation and increase of PGC1α in STs from obese, but not in lean women. TRKB antagonism attenuated ATP-coupled respiration, maximal respiration, and spare capacity in STs from lean and obese women. 7,8-DHF improves mitochondrial respiration in STs from obese women, suggesting that the obese phenotype in the placenta can be rescued by TRKB activation.
Collapse
Affiliation(s)
- Calais S Prince
- 1 Center for Pregnancy and Newborn Research, Department of Obstetrics and Gynecology, University of Texas Health Science Center San Antonio, TX, USA
| | - Alina Maloyan
- 1 Center for Pregnancy and Newborn Research, Department of Obstetrics and Gynecology, University of Texas Health Science Center San Antonio, TX, USA.,2 Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Leslie Myatt
- 1 Center for Pregnancy and Newborn Research, Department of Obstetrics and Gynecology, University of Texas Health Science Center San Antonio, TX, USA.,3 Deparment of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
38
|
Rudnitskaya EA, Kolosova NG, Stefanova NA. Impact of changes in neurotrophic supplementation on development of Alzheimer’s disease-like pathology in OXYS rats. BIOCHEMISTRY (MOSCOW) 2017; 82:318-329. [DOI: 10.1134/s0006297917030105] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
39
|
Roles of the serotonin 5-HT4 receptor in dendrite formation of the rat hippocampal neurons in vitro. Brain Res 2016; 1655:114-121. [PMID: 27894797 DOI: 10.1016/j.brainres.2016.11.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/09/2016] [Accepted: 11/19/2016] [Indexed: 02/04/2023]
Abstract
Serotonin (5-HT) is involved in various aspects of hippocampal development, although the specific roles of 5-HT receptors are poorly understood. We investigated the roles of 5-HT receptors in the dendrite formation of hippocampal neurons. We focused on the 5-HT4 receptor, which is coupled with Gs protein, and compared the effects with those of the Gi-coupled 5-HT1A receptor. Neurons from rat hippocampi at embryonic day 18 were dissociated and treated for 4 days with the 5-HT4 receptor agonist BIMU8 or the 5-HT1A receptor agonist 8-OH DPAT. The formation of primary dendrites and dendrite branching were promoted by BIMU8, whereas the dendrite branching was inhibited by 8-OH DPAT. BIMU8-induced promotion of dendrite formation was neutralized by concomitant treatment with the 5-HT4 receptor antagonist, confirming the specific actions of the 5-HT4 receptor. We then examined the signaling mechanisms underlying the actions of the 5-HT4 receptor by using a protein kinase A (PKA) inhibitor. The BIMU8-induced promotion of dendrite formation was reversed partially by the PKA inhibitor, suggesting involvement of PKA signaling downstream of the 5-HT4 receptor. Finally, we examined the contribution of brain-derived neurotrophic factor (BDNF) to the promotion of dendrite formation by BIMU8. Quantitative RT-PCR analysis showed that BIMU8 increased the BDNF mRNA expression and that treatment of cultured neurons with the TrkB antagonist reversed the BIMU8-induced increase in dendrite formation. In summary, the present study suggests a novel role for the 5-HT4 receptor in facilitation of dendrite formation in which intracellular signaling of PKA and the BDNF-TrkB system may be involved.
Collapse
|
40
|
Ji LL, Peng JB, Fu CH, Cao D, Li D, Tong L, Wang ZY. Activation of Sigma-1 receptor ameliorates anxiety-like behavior and cognitive impairments in a rat model of post-traumatic stress disorder. Behav Brain Res 2016; 311:408-415. [PMID: 27275520 DOI: 10.1016/j.bbr.2016.05.056] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 05/21/2016] [Accepted: 05/26/2016] [Indexed: 11/25/2022]
Abstract
Among learning and memory processes, fear memories are crucial in some psychiatric disorders like post-traumatic stress disorder (PTSD). Accumulating evidence shows that the sigma-1 receptor (Sig-1R) has comprehensive involvement in cognitive impairment and neuroprotective effects. It has also been reported that BDNF appears to enhance extinction of fear in anxiety disorders via the MAPK signaling cascade. However, it remains unclear whether BDNF-TrkB-MAPK pathway may be mechanistically involved in the therapeutic effect of sigma-1 receptor in the development of PTSD. To address this question, rats were subjected to a classical single-prolonged stress procedure (SPS) and kept undisturbed for 7 days. After that, rats were re-stressed by re-exposure to the forced swim component of SPS (RSPS). Behavior tests were subsequently performed to assess anxiety and cognitive impairments. Furthermore, we analyzed the expression of BDNF and the phosphorylation of TrkB and three MAPK pathways, namely, the ERK, JNK and p38. We found that the levels of BDNF and p-TrkB were increased following the RSPS procedure, which were reversed by the administration of PRE-084. Meanwhile, among the three MAPK signaling pathways, only the p-ERK expression was increased following the RSPS procedure. Collectively, our results indicate that BDNF-TrkB-ERK signaling pathway may be involved in the activation of sigma-1 receptor to yield therapeutic benefits for PTSD.
Collapse
Affiliation(s)
- Li-Li Ji
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Jun-Bo Peng
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Chang-Hai Fu
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Dong Cao
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Dan Li
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China
| | - Lei Tong
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China.
| | - Zhen-Yu Wang
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, People's Republic of China.
| |
Collapse
|
41
|
Abstract
The drug discovery landscape has been transformed over the past decade by the discovery of allosteric modulators of all major mammalian receptor superfamilies. Allosteric ligands are a rich potential source of drugs and drug targets with clear therapeutic advantages. G protein-coupled receptors, ligand-gated ion channels and intracellular nuclear hormone receptors have all been targeted by allosteric modulators. More recently, a receptor tyrosine kinase (RTK) has been targeted by an extracellular small-molecule allosteric modulator. Allosteric mechanisms of structurally distinct molecules that target the various receptor families are more alike than originally anticipated and include selectivity, orthosteric probe dependence and pathway-biased signaling.
Collapse
|
42
|
Transcriptional analysis reveals distinct subtypes in amyotrophic lateral sclerosis: implications for personalized therapy. Future Med Chem 2016; 7:1335-59. [PMID: 26144267 DOI: 10.4155/fmc.15.60] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable disease, caused by the loss of the upper and lower motor neurons. The lack of therapeutic progress is mainly due to the insufficient understanding of complexity and heterogeneity underlying the pathogenic mechanisms of ALS. Recently, we analyzed whole-genome expression profiles of motor cortex of sporadic ALS patients, classifying them into two subgroups characterized by differentially expressed genes and pathways. Some of the deregulated genes encode proteins, which are primary targets of drugs currently in preclinical or clinical studies for several clinical conditions, including neurodegenerative diseases. In this review, we discuss in-depth the potential role of these candidate targets in ALS pathogenesis, highlighting their possible relevance for personalized ALS treatments.
Collapse
|
43
|
Computational Design of TrkB Peptide Inhibitors and Their Biological Effects on Ovarian Cancer Cell Lines. Int J Pept Res Ther 2016. [DOI: 10.1007/s10989-015-9510-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
44
|
M'Dahoma S, Barthélemy S, Tromilin C, Jeanson T, Viguier F, Michot B, Pezet S, Hamon M, Bourgoin S. Respective pharmacological features of neuropathic-like pain evoked by intrathecal BDNF versus sciatic nerve ligation in rats. Eur Neuropsychopharmacol 2015; 25:2118-30. [PMID: 26343858 DOI: 10.1016/j.euroneuro.2015.07.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 07/27/2015] [Accepted: 07/30/2015] [Indexed: 12/11/2022]
Abstract
Numerous reported data support the idea that Brain Derived Neurotrophic Factor (BDNF) is critically involved in both depression and comorbid pain. The possible direct effect of BDNF on pain mechanisms was assessed here and compared with behavioral/neurobiological features of neuropathic pain caused by chronic constriction injury to the sciatic nerve (CCI-SN). Sprague-Dawley male rats were either injected intrathecally with BDNF (3.0 ng i.t.) or subjected to unilateral CCI-SN. Their respective responses to anti-hyperalgesic drugs were assessed using the Randall-Selitto test and both immunohistochemical and RT-qPCR approaches were used to investigate molecular/cellular mechanisms underlying hyperalgesia in both models. Long lasting hyperalgesia and allodynia were induced by i.t. BDNF in intact healthy rats like those found after CCI-SN. Acute treatment with the BDNF-TrkB receptor antagonist cyclotraxin B completely prevented i.t. BDNF-induced hyperalgesia and partially reversed this symptom in both BDNF-pretreated and CCI-SN lesioned rats. Acute administration of the anticonvulsant pregabalin, the NMDA receptor antagonist ketamine, the opioid analgesics morphine and tapentadol or the antidepressant agomelatine also transiently reversed hyperalgesia in both i.t. BDNF injected- and CCI-SN lesioned-rats. Marked induction of microglia activation markers (OX42, Iba1, P-p38), proinflammatory cytokine IL-6, NMDA receptor subunit NR2B and BDNF was found in spinal cord and/or dorsal root ganglia of CCI-SN rats. A long lasting spinal BDNF overexpression was also observed in BDNF i.t. rats, indicating an autocrine self-induction, with downstream long lasting TrkB-mediated neuropathic-like pain. Accordingly, TrkB blockade appeared as a relevant approach to alleviate not only i.t. BDNF- but also nerve lesion-evoked neuropathic pain.
Collapse
Affiliation(s)
- Saïd M'Dahoma
- Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Paris F-75014, France; Université Pierre et Marie Curie - Paris 6, Faculté de Médecine Pierre et Marie Curie, Site Pitié-Salpêtrière, Paris F-75013, France
| | - Sandrine Barthélemy
- Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Paris F-75014, France; Université Pierre et Marie Curie - Paris 6, Faculté de Médecine Pierre et Marie Curie, Site Pitié-Salpêtrière, Paris F-75013, France
| | - Claire Tromilin
- Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Paris F-75014, France; Université Pierre et Marie Curie - Paris 6, Faculté de Médecine Pierre et Marie Curie, Site Pitié-Salpêtrière, Paris F-75013, France
| | - Tiffany Jeanson
- Université Pierre et Marie Curie - Paris 6, Faculté de Médecine Pierre et Marie Curie, Site Pitié-Salpêtrière, Paris F-75013, France; Theranexus, 91400 Orsay, France
| | - Florent Viguier
- Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Paris F-75014, France; Université Pierre et Marie Curie - Paris 6, Faculté de Médecine Pierre et Marie Curie, Site Pitié-Salpêtrière, Paris F-75013, France
| | - Benoit Michot
- Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Paris F-75014, France; Université Pierre et Marie Curie - Paris 6, Faculté de Médecine Pierre et Marie Curie, Site Pitié-Salpêtrière, Paris F-75013, France
| | - Sophie Pezet
- UMR 8249 CNRS - Brain Plasticity Unit, ESCPI-ParisTech, Paris F-75005, France
| | - Michel Hamon
- Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Paris F-75014, France; Université Pierre et Marie Curie - Paris 6, Faculté de Médecine Pierre et Marie Curie, Site Pitié-Salpêtrière, Paris F-75013, France; Université Paris Descartes, Sorbonne Paris Cité - Paris 5, France
| | - Sylvie Bourgoin
- Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Paris F-75014, France; Université Pierre et Marie Curie - Paris 6, Faculté de Médecine Pierre et Marie Curie, Site Pitié-Salpêtrière, Paris F-75013, France; Université Paris Descartes, Sorbonne Paris Cité - Paris 5, France.
| |
Collapse
|
45
|
Brain-derived neurotrophic factor inhibits osmotic swelling of rat retinal glial (Müller) and bipolar cells by activation of basic fibroblast growth factor signaling. Neuroscience 2015; 295:175-86. [DOI: 10.1016/j.neuroscience.2015.03.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 03/17/2015] [Accepted: 03/18/2015] [Indexed: 12/19/2022]
|
46
|
Fabbro D, Cowan-Jacob SW, Moebitz H. Ten things you should know about protein kinases: IUPHAR Review 14. Br J Pharmacol 2015; 172:2675-700. [PMID: 25630872 DOI: 10.1111/bph.13096] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 12/31/2014] [Accepted: 01/20/2015] [Indexed: 12/12/2022] Open
Abstract
Many human malignancies are associated with aberrant regulation of protein or lipid kinases due to mutations, chromosomal rearrangements and/or gene amplification. Protein and lipid kinases represent an important target class for treating human disorders. This review focus on 'the 10 things you should know about protein kinases and their inhibitors', including a short introduction on the history of protein kinases and their inhibitors and ending with a perspective on kinase drug discovery. Although the '10 things' have been, to a certain extent, chosen arbitrarily, they cover in a comprehensive way the past and present efforts in kinase drug discovery and summarize the status quo of the current kinase inhibitors as well as knowledge about kinase structure and binding modes. Besides describing the potentials of protein kinase inhibitors as drugs, this review also focus on their limitations, particularly on how to circumvent emerging resistance against kinase inhibitors in oncological indications.
Collapse
Affiliation(s)
| | | | - Henrik Moebitz
- Novartis Institutes of Biomedical Research, Basel, Switzerland
| |
Collapse
|
47
|
Activity-dependent upregulation of presynaptic kainate receptors at immature CA3-CA1 synapses. J Neurosci 2015; 34:16902-16. [PMID: 25505341 DOI: 10.1523/jneurosci.1842-14.2014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Presynaptic kainate-type glutamate receptors (KARs) regulate glutamate release probability and short-term plasticity in various areas of the brain. Here we show that long-term depression (LTD) in the area CA1 of neonatal rodent hippocampus is associated with an upregulation of tonic inhibitory KAR activity, which contributes to synaptic depression and causes a pronounced increase in short-term facilitation of transmission. This increased KAR function was mediated by high-affinity receptors and required activation of NMDA receptors, nitric oxide (NO) synthetase, and postsynaptic calcium signaling. In contrast, KAR activity was irreversibly downregulated in response to induction of long-term potentiation in a manner that depended on activation of the TrkB-receptor of BDNF. Both tonic KAR activity and its plasticity were restricted to early stages of synapse development and were lost in parallel with maturation of the network due to ongoing BDNF-TrkB signaling. These data show that presynaptic KARs are targets for activity-dependent modulation via diffusible messengers NO and BDNF, which enhance and depress tonic KAR activity at immature synapses, respectively. The plasticity of presynaptic KARs in the developing network allows nascent synapses to shape their response to incoming activity. In particular, upregulation of KAR function after LTD allows the synapse to preferentially pass high-frequency afferent activity. This can provide a potential rescue from synapse elimination by uncorrelated activity and also increase the computational dynamics of the developing CA3-CA1 circuitry.
Collapse
|
48
|
BDNF-dependent plasticity induced by peripheral inflammation in the primary sensory and the cingulate cortex triggers cold allodynia and reveals a major role for endogenous BDNF as a tuner of the affective aspect of pain. J Neurosci 2015; 34:14739-51. [PMID: 25355226 DOI: 10.1523/jneurosci.0860-14.2014] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Painful experiences are multilayered, composed of sensory, affective, cognitive and behavioral facets. Whereas it is well accepted that the development of chronic pain is due to maladaptive neuronal changes, the underlying molecular mechanisms, their relationship to the different pain modalities, and indeed the localization of these changes are still unknown. Brain-derived neurotrophic factor (BDNF) is an activity-dependent neuromodulator in the adult brain, which enhances neuronal excitability. In the spinal cord, BDNF underlies the development and maintenance of inflammatory and neuropathic pain. Here, we hypothesized that BDNF could be a trigger of some of these plastic changes. Our results demonstrate that BDNF is upregulated in the anterior cingulate cortex (ACC) and the primary sensory cortex (S1) in rats with inflammatory pain. Injections of recombinant BDNF (into the ACC) or a viral vector synthesizing BDNF (into the ACC or S1) triggered both neuronal hyperexcitability, as shown by elevated long-term potentiation, and sustained pain hypersensitivity. Finally, pharmacological blockade of BDNF-tropomyosin receptor kinase B (TrkB) signaling in the ACC, through local injection of cyclotraxin-B (a novel, highly potent, and selective TrkB antagonist) prevented neuronal hyperexcitability, the emergence of cold hypersensitivity, and passive avoidance behavior. These findings show that BDNF-dependent neuronal plasticity in the ACC, a structure known to be involved in the affective-emotional aspect of pain, is a key mechanism in the development and maintenance of the emotional aspect of chronic pain.
Collapse
|
49
|
Fabbro D. 25 Years of Small Molecular Weight Kinase Inhibitors: Potentials and Limitations. Mol Pharmacol 2014; 87:766-75. [DOI: 10.1124/mol.114.095489] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
50
|
Christopoulos A, Changeux JP, Catterall WA, Fabbro D, Burris TP, Cidlowski JA, Olsen RW, Peters JA, Neubig RR, Pin JP, Sexton PM, Kenakin TP, Ehlert FJ, Spedding M, Langmead CJ. International Union of Basic and Clinical Pharmacology. XC. multisite pharmacology: recommendations for the nomenclature of receptor allosterism and allosteric ligands. Pharmacol Rev 2014; 66:918-47. [PMID: 25026896 PMCID: PMC11060431 DOI: 10.1124/pr.114.008862] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Allosteric interactions play vital roles in metabolic processes and signal transduction and, more recently, have become the focus of numerous pharmacological studies because of the potential for discovering more target-selective chemical probes and therapeutic agents. In addition to classic early studies on enzymes, there are now examples of small molecule allosteric modulators for all superfamilies of receptors encoded by the genome, including ligand- and voltage-gated ion channels, G protein-coupled receptors, nuclear hormone receptors, and receptor tyrosine kinases. As a consequence, a vast array of pharmacologic behaviors has been ascribed to allosteric ligands that can vary in a target-, ligand-, and cell-/tissue-dependent manner. The current article presents an overview of allostery as applied to receptor families and approaches for detecting and validating allosteric interactions and gives recommendations for the nomenclature of allosteric ligands and their properties.
Collapse
Affiliation(s)
- Arthur Christopoulos
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Jean-Pierre Changeux
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - William A Catterall
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Doriano Fabbro
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Thomas P Burris
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - John A Cidlowski
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Richard W Olsen
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - John A Peters
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Richard R Neubig
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Jean-Philippe Pin
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Patrick M Sexton
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Terry P Kenakin
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Frederick J Ehlert
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Michael Spedding
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| | - Christopher J Langmead
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (A.C., P.M.S., C.J.L.); Collège de France and CNRS URA 2182, Institut Pasteur, Paris, France (J.-P.C.); Department of Pharmacology, School of Medicine, University of Washington, Seattle, Washington (W.A.C.); PIQUR Therapeutics AG, Basel, Switzerland (D.F.); Department of Pharmacological & Physiological Science, Saint Louis University School of Medicine, St. Louis, Louisiana (T.P.B.); Signal Transduction Laboratory, Molecular Endocrinology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (J.A.C.); Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California (R.W.O.); Division of Neuroscience, School of Medicine, University of Dundee, Scotland, United Kingdom (J.A.P.); Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (R.R.N.); Institut de Genomique Fonctionelle, CNRS, Montpellier, France (J.-P.P.); Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina (T.P.K.); Department of Pharmacology, University of California, Irvine, California (F.J.E.); and Research Solutions SARL, Paris, France (M.S.)
| |
Collapse
|