Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Cells ; 10(8)2021 07 23.
Article in English | MEDLINE | ID: mdl-34440640

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disorder and results in severe neurodegeneration and progressive cognitive decline. Neurotrophins are growth factors involved in the development and survival of neurons, but also in underlying mechanisms for memory formation such as hippocampal long-term potentiation. Our aim was to identify small molecules with stimulatory effects on the signaling of two neurotrophins, the nerve growth factor (NGF) and the brain derived neurotrophic factor (BDNF). To identify molecules that could potentiate neurotrophin signaling, 25,000 molecules were screened, which led to the identification of the triazinetrione derivatives ACD855 (Ponazuril) and later on ACD856, as positive allosteric modulators of tropomyosin related kinase (Trk) receptors. ACD855 or ACD856 potentiated the cellular signaling of the neurotrophin receptors with EC50 values of 1.9 and 3.2 or 0.38 and 0.30 µM, respectively, for TrkA or TrkB. ACD855 increased acetylcholine levels in the hippocampus by 40% and facilitated long term potentiation in rat brain slices. The compounds acted as cognitive enhancers in a TrkB-dependent manner in several different behavioral models. Finally, the age-induced cognitive dysfunction in 18-month-old mice could be restored to the same level as found in 2-month-old mice after a single treatment of ACD856. We have identified a novel mechanism to modulate the activity of the Trk-receptors. The identification of the positive allosteric modulators of the Trk-receptors might have implications for the treatment of Alzheimer's diseases and other diseases characterized by cognitive impairment.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Cognition/drug effects , Cognitive Dysfunction/drug therapy , Nootropic Agents/pharmacology , Receptors, Nerve Growth Factor/agonists , Age Factors , Animals , Brain/enzymology , Brain/physiopathology , Cell Line, Tumor , Cognitive Dysfunction/enzymology , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/psychology , Disease Models, Animal , Humans , Male , Maze Learning/drug effects , Membrane Glycoproteins , Mice, Inbred C57BL , Motor Activity/drug effects , Protein-Tyrosine Kinases , Rats, Sprague-Dawley , Receptor, trkA/agonists , Receptor, trkA/metabolism , Receptor, trkB/agonists , Receptor, trkB/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Signal Transduction , Small Molecule Libraries , Triazines/pharmacology
2.
Biomolecules ; 10(2)2020 02 01.
Article in English | MEDLINE | ID: mdl-32024191

ABSTRACT

In the last decade, Nerve Growth Factor (NGF)-based clinical approaches have lacked specific and efficient Tyrosine Kinase A (TrkA) agonists for brain delivery. Nowadays, the characterization of novel small peptidomimetic is taking centre stage in preclinical studies, in order to overcome the main size-related limitation in brain delivery of NGF holoprotein for Central Nervous System (CNS) pathologies. Here we investigated the NGF mimetic properties of the human NGF 1-14 sequence (hNGF1-14) and its derivatives, by resorting to primary cholinergic and dorsal root ganglia (DRG) neurons. Briefly, we observed that: 1) hNGF1-14 peptides engage the NGF pathway through TrkA phosphorylation at tyrosine 490 (Y490), and activation of ShcC/PI3K and Plc-γ/MAPK signalling, promoting AKT-dependent survival and CREB-driven neuronal activity, as seen by levels of the immediate early gene c-Fos, of the cholinergic marker Choline Acetyltransferase (ChAT), and of Brain Derived Neurotrophic Factor (BDNF); 2) their NGF mimetic activity is lost upon selective TrkA inhibition by means of GW441756; 3) hNGF1-14 peptides are able to sustain DRG survival and differentiation in absence of NGF. Furthermore, the acetylated derivative Ac-hNGF1-14 demonstrated an optimal NGF mimetic activity in both neuronal paradigms and an electrophysiological profile similar to NGF in cholinergic neurons. Cumulatively, the findings here reported pinpoint the hNGF1-14 peptide, and in particular its acetylated derivative, as novel, specific and low molecular weight TrkA specific agonists in both CNS and PNS primary neurons.


Subject(s)
Cholinergic Neurons/metabolism , Ganglia, Spinal/metabolism , Nerve Growth Factor/chemistry , Receptor, trkA/agonists , Receptor, trkA/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 3/metabolism , Animals , Biological Assay , Cell Differentiation , Cell Survival , Cells, Cultured , Humans , Peptides/chemistry , Phosphorylation , Rats , Signal Transduction , Tyrosine/chemistry
3.
Sci Adv ; 6(4): eaax6646, 2020 01.
Article in English | MEDLINE | ID: mdl-32010781

ABSTRACT

The degeneration of cholinergic neurons is a prominent feature of Alzheimer's disease (AD). In animal models of injury and aging, nerve growth factor (NGF) enhances cholinergic cell survival and function, contributing to improved memory. In the presence of AD pathology, however, NGF-related therapeutics have yet to fulfill their regenerative potential. We propose that stimulating the TrkA receptor, without p75NTR activation, is key for therapeutic efficacy. Supporting this hypothesis, the selective TrkA agonist D3 rescued neurotrophin signaling in TgCRND8 mice, whereas NGF, interacting with both TrkA and p75NTR, did not. D3, delivered intravenously and noninvasively to the basal forebrain using MRI-guided focused ultrasound (MRIgFUS)-mediated blood-brain barrier (BBB) permeability activated TrkA-related signaling cascades and enhanced cholinergic neurotransmission. Recent clinical trials support the safety and feasibility of MRIgFUS BBB modulation in AD patients. Neuroprotective agents targeting TrkA, combined with MRIgFUS BBB modulation, represent a promising strategy to counter neurodegeneration in AD.


Subject(s)
Alzheimer Disease/metabolism , Choline/metabolism , Cholinergic Agents/administration & dosage , Drug Delivery Systems , Receptor, trkA/agonists , Receptor, trkA/metabolism , Ultrasonic Waves , Alzheimer Disease/drug therapy , Alzheimer Disease/etiology , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Disease Models, Animal , Mice , Mice, Transgenic , Nerve Growth Factor/metabolism , Signal Transduction/drug effects
4.
PLoS One ; 14(10): e0224022, 2019.
Article in English | MEDLINE | ID: mdl-31671109

ABSTRACT

Neurotrophins and their mimetics are potential treatments for hearing disorders because of their trophic effects on spiral ganglion neurons (SGNs) whose connections to hair cells may be compromised in many forms of hearing loss. Studies in noise or ototoxin-exposed animals have shown that local delivery of NT-3 or BDNF has beneficial effects on SGNs and hearing. We evaluated several TrkB or TrkC monoclonal antibody agonists and small molecules, along with BDNF and NT-3, in rat cochlea ex vivo models. The TrkB agonists BDNF and a monoclonal antibody, M3, had the greatest effects on SGN survival, neurite outgrowth and branching. In organotypic cochlear explants, BDNF and M3 enhanced synapse formation between SGNs and inner hair cells and restored these connections after excitotoxin-induced synaptopathy. Loss of these synapses has recently been implicated in hidden hearing loss, a condition characterized by difficulty hearing speech in the presence of background noise. The unique profile of M3 revealed here warrants further investigation, and the broad activity profile of BDNF observed underpins its continued development as a hearing loss therapeutic.


Subject(s)
Antibodies, Monoclonal/immunology , Brain-Derived Neurotrophic Factor/pharmacology , Cochlea/cytology , Hearing Loss/pathology , Neurites/metabolism , Receptor, trkA/agonists , Synapses/metabolism , Animals , Cell Line , Cell Survival , Disease Models, Animal , Hearing Loss/immunology , Humans , Neurites/drug effects , Neurites/immunology , Rats , Receptor, trkA/immunology , Synapses/drug effects , Synapses/immunology
5.
PLoS One ; 14(8): e0221757, 2019.
Article in English | MEDLINE | ID: mdl-31465471

ABSTRACT

The human hair follicle is a neuroendocrine mini-organ that can be used to study aging processes in vitro. Neurotrophins maintain homeostasis in hair biology via the Trk-family of receptors. TrkA, the high affinity receptor for nerve growth factor (NGF), is expressed in hair follicle melanocytes and keratinocytes, where it regulates proliferation, differentiation and apoptosis and may thereby play a role in hair pigmentation and growth. We investigated TrkA expression during the human hair cycle and the effects of a selective high affinity TrkA agonist, Gambogic Amide, on hair pigmentation and hair growth in human hair follicles in vitro. In human scalp skin, TrkA expression was strongest in proliferating melanocytes re-establishing the pigmentary unit in the hair bulb during the early hair growth phase, anagen. During high anagen and in the de-composing pigmentary-unit of the regression phase, catagen, bulb-melanocytes lost TrkA expression and only undifferentiated outer root sheath melanocytes maintained it. In cultured human anagen hair follicles, Gambogic Amide was able to prevent gradual pigment loss, while it stimulated hair shaft elongation. This was achieved by increased melanocyte activation, migration and dendricity, highlighted by distinct c-KIT-expression in melanocyte sub-populations. Our results suggest that Gambogic Amide can maintain hair follicle pigmentation by acting on undifferentiated melanocytes residing in the outer root sheath and making them migrate to establish the pigmentary-unit. This suggests that the selective TrkA agonist Gambogic Amide acts as an anti-hair greying and hair growth promoting molecule in vitro.


Subject(s)
Hair Follicle/growth & development , Pigmentation/drug effects , Receptor, trkA/agonists , Xanthones/pharmacology , Adult , Aged , Cell Differentiation/drug effects , Cells, Cultured , Hair Follicle/drug effects , Homeostasis/drug effects , Humans , Melanocytes/cytology , Melanocytes/drug effects , Middle Aged , Nerve Growth Factor/pharmacology , Receptor, trkA/metabolism
6.
PLoS One ; 14(6): e0218036, 2019.
Article in English | MEDLINE | ID: mdl-31233568

ABSTRACT

In the brain, the TrkA receptor for Nerve Growth Factor (NGF) is expressed primarily in the cholinergic system. TrkA/NGF support neuronal health and function, and deficiencies in this axis are associated with progressive cholinergic neuron atrophy and death, and with cognitive deficit in disorders such as Down's syndrome and Alzheimer's disease. These observations led to the hypothesis that TrkA agonists may rescue atrophic cholinergic neurons and benefit cognition. Indeed, a small molecule TrkA partial agonist called D3 normalized TrkA signals and improved memory in cognitive impairment models of ageing and an APP mouse model of Alzheimer's disease. Paradoxically, in young healthy mice chronic delivery of D3 caused impaired memory without impairing learning, a form of anterograde amnesia. Here, we use this as a model to study the mechanisms of impaired memory. In young healthy mice acute or chronic treatment with D3 induces hyperactivation of TrkA-mediated signals in hippocampus, and causes a deficit in hippocampal-dependent memory consolidation proximal to drug exposure, without affecting learning or memory retrieval. The impairment after acute drug exposure is reversible. The impairment after long-term drug exposure is irreversible, likely due to a decrease in hippocampal CA1 neuron basal arborization. These findings support the notion of a homeostatic role for TrkA in memory, and demonstrate the differential outcomes of TrkA (hyper)activation in healthy versus disease states.


Subject(s)
Hippocampus/metabolism , Memory Consolidation , Receptor, trkA/agonists , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Dendrites/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice, Inbred C57BL , Phosphorylation , Receptor, trkA/metabolism , Spatial Memory
7.
J Musculoskelet Neuronal Interact ; 19(1): 94-103, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30839307

ABSTRACT

OBJECTIVES: To study effects of the selective TrkA agonist, gambogic amide (GA), on fracture healing in mice and on an osteoprogenitor cell line in vitro. METHODS: Mice were given bilateral fibular fractures and treated for two weeks with vehicle or 1 mg/kg/day GA and euthanized at 14-, 21-, and 42-days post-fracture. Calluses were analysed by micro-computed tomography (µCT), three-point bending and histology. For RT-PCR analyses, Kusa O cells were treated with 0.5nM of GA or vehicle for 3, 7, and 14 days, while for mineralization assessment, cells were treated for 21 days. RESULTS: µCT analysis found that 21-day GA-treated calluses had both decreased tissue volume (p<0.05) and bone surface (p<0.05) and increased fractional bone volume (p<0.05) compared to controls. Biomechanical analyses of 42-day calluses revealed that GA treatment increased stiffness per unit area by 53% (p<0.01) and load per unit area by 52% (p<0.01). GA treatment increased Kusa O gene expression of alkaline phosphatase and osteocalcin (p<0.05) by 14 days as well as mineralization at 21 days (p<0.05). CONCLUSIONS: GA treatment appeared to have a beneficial effect on fracture healing at 21- and 42-days post-fracture. The exact mechanism is not yet understood but may involve increased osteoblastic differentiation and matrix mineralization.


Subject(s)
Calcification, Physiologic/drug effects , Fracture Healing/drug effects , Osteoblasts/drug effects , Xanthones/pharmacology , Animals , Cell Differentiation/drug effects , Fracture Healing/physiology , Male , Mice , Mice, Inbred C57BL , Osteoblasts/cytology , Receptor, trkA/agonists
8.
Pharmacol Res ; 139: 17-25, 2019 01.
Article in English | MEDLINE | ID: mdl-30391352

ABSTRACT

Nerve Growth Factor (NGF) is a therapeutic candidate for Alzheimer's disease, based on its well known actions on basal forebrain cholinergic neurons. However, because of its pro-nociceptive activity, in current clinical trials NGF has to be administered intraparenchymally into the brain by neurosurgery via cell or gene therapy approaches. To prevent the NGF pain-inducing collateral effects, thus avoiding the necessity for local brain injection, we developed painless NGF (hNGFp), based on the human genetic disease Hereditary Sensory and Autonomic Neuropathy type V (HSAN V). hNGFp has similar neurotrophic activity as wild type human NGF, but its pain sensitizing activity is tenfold lower. Pharmacologically, hNGFp is a biased receptor agonist of NGF TrkA receptor. The results of recent studies shed new light on the neuroprotective mechanism by hNGFp and are highly relevant for the planning of NGF-based clinical trials. The intraparenchymal delivery of hNGFp, as used in clinical trials, was simulated in the 5xFAD mouse model and found to be inefficacious in reducing Aß plaque load. On the contrary, the same dose of hNGFp administered intranasally, which was rather widely biodistributed in the brain and did not induce pain sensitization, blocked APP processing into amyloid and restored synaptic plasticity and memory in this aggressive neurodegeneration model. This potent and broad neuroprotection by hNGFp was found to be mediated by hNGFp actions on glial cells. hNGFp increases inflammatory proteins such as the soluble TNFα receptor II and the chemokine CXCL12. Independent work has shown that NGF has a potent anti-inflammatory action on microglia and steers them towards a neuroprotective phenotype. These studies demonstrate that microglia cells are a new target cell of NGF in the brain and have therapeutic significance: i) they establish that the neuroprotective actions of hNGFp relies on a widespread exposure of the brain, ii) they identify a new anti-neurodegenerative pathway, linking hNGFp to inflammatory chemokines and cytokines via microglia, a common target for new therapeutic opportunities for neurodegenerative diseases, iii) they extend the neuroprotective potential of hNGFp beyond its classical cholinergic target, thereby widening the range of neurological diseases for which this neurotrophic factor might be used therapeutically, iv) they help interpreting the results of current NGF clinical trials in AD and the design of future trials with this new potent therapeutic candidate.


Subject(s)
Microglia/drug effects , Nerve Growth Factor/administration & dosage , Neuroprotective Agents/administration & dosage , Receptor, trkA/agonists , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Chemokine CXCL12/metabolism , Humans , Microglia/metabolism , Nerve Growth Factor/adverse effects , Nerve Growth Factor/metabolism , Neuroprotection , Neuroprotective Agents/adverse effects , Neuroprotective Agents/metabolism , Pain/chemically induced
9.
Diabetes ; 67(2): 321-333, 2018 02.
Article in English | MEDLINE | ID: mdl-29208634

ABSTRACT

BNN27, a C17-spiroepoxy derivative of DHEA, was shown to have antiapoptotic properties via mechanisms involving the nerve growth factor receptors (tropomyosin-related kinase A [TrkA]/neurotrophin receptor p75 [p75NTR]). In this study, we examined the effects of BNN27 on neural/glial cell function, apoptosis, and inflammation in the experimental rat streptozotocin (STZ) model of diabetic retinopathy (DR). The ability of BNN27 to activate the TrkA receptor and regulate p75NTR expression was investigated. BNN27 (2,10, and 50 mg/kg i.p. for 7 days) administration 4 weeks post-STZ injection (paradigm A) reversed the diabetes-induced glial activation and loss of function of amacrine cells (brain nitric oxide synthetase/tyrosine hydroxylase expression) and ganglion cell axons via a TrkA receptor (TrkAR)-dependent mechanism. BNN27 activated/phosphorylated the TrkAY490 residue in the absence but not the presence of TrkAR inhibitor and abolished the diabetes-induced increase in p75NTR expression. However, it had no effect on retinal cell death (TUNEL+ cells). A similar result was observed when BNN27 (10 mg/kg i.p.) was administered at the onset of diabetes, every other day for 4 weeks (paradigm B). However, BNN27 decreased the activation of caspase-3 in both paradigms. Finally, BNN27 reduced the proinflammatory (TNFα and IL-1ß) and increased the anti-inflammatory (IL-10 and IL-4) cytokine levels. These findings suggest that BNN27 has the pharmacological profile of a therapeutic for DR, since it targets both the neurodegenerative and inflammatory components of the disease.


Subject(s)
Amacrine Cells/drug effects , Anti-Inflammatory Agents/therapeutic use , Dehydroepiandrosterone/therapeutic use , Diabetic Retinopathy/prevention & control , Neuroprotective Agents/therapeutic use , Receptor, trkA/agonists , Retina/drug effects , Amacrine Cells/immunology , Amacrine Cells/metabolism , Amacrine Cells/pathology , Animals , Anti-Inflammatory Agents/administration & dosage , Axons/drug effects , Axons/immunology , Axons/metabolism , Axons/pathology , Dehydroepiandrosterone/administration & dosage , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/physiopathology , Diabetic Retinopathy/immunology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Dose-Response Relationship, Drug , Eye Proteins/agonists , Eye Proteins/metabolism , Female , Ganglia, Sensory/drug effects , Ganglia, Sensory/immunology , Ganglia, Sensory/metabolism , Ganglia, Sensory/pathology , Male , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/metabolism , Neuroglia/drug effects , Neuroglia/immunology , Neuroglia/metabolism , Neuroglia/pathology , Neuroprotective Agents/administration & dosage , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor/agonists , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Retina/immunology , Retina/pathology , Retina/physiopathology , Streptozocin
10.
Brain Inj ; 32(2): 257-268, 2018.
Article in English | MEDLINE | ID: mdl-29227174

ABSTRACT

OBJECTIVES: There is evidence that treatment with nerve growth factor (NGF) may reduce neuroinflammation and apoptosis after a traumatic brain injury (TBI). NGF is thought to exert its effects via binding to either TrkA or p75 neurotrophin receptors. This study aimed to investigate the effects of a selective TrkA agonist, gambogic amide (GA), on TBI pathology and outcomes in mice following lateral fluid percussion injury. METHODS: Male C57BL/6 mice were given either a TBI or sham injury, and then received subcutaneous injections of either 2 mg/kg of GA or vehicle at 1, 24, and 48 h post-injury. Following behavioural studies, mice were euthanized at 72 h post-injury for analysis of neuroinflammatory, apoptotic, and neurite outgrowth markers. RESULTS: Behavioural testing revealed that GA did not mitigate motor deficits after TBI. TBI caused an increase in cortical and hippocampal expression of several markers of neuroinflammation and apoptosis compared to sham groups. GA treatment did not attenuate these increases in expression, possibly contributed to by our finding of TrkA receptor down-regulation post-TBI. CONCLUSIONS: These findings suggest that GA treatment may not be suitable for attenuating TBI pathology and improving outcomes.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Receptor, trkA/agonists , Xanthones/therapeutic use , Analysis of Variance , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/physiopathology , Calcium-Binding Proteins/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Exploratory Behavior/drug effects , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptor, trkA/genetics , Receptor, trkA/metabolism , Rotarod Performance Test , Treatment Outcome
11.
Exp Neurol ; 300: 87-99, 2018 02.
Article in English | MEDLINE | ID: mdl-29106982

ABSTRACT

Neurotrophic factors and their corresponding receptors play key roles in the maintenance of different phenotypic dorsal root ganglion (DRG) neurons, the axons of which degenerate in small fiber neuropathy, leading to various neuropathic manifestations. Mechanisms underlying positive and negative symptoms of small fiber neuropathy have not been systematically explored. This study investigated the molecular basis of these seemingly paradoxical neuropathic behaviors according to the profiles of TrkA and Ret with immunohistochemical and pharmacological interventions in a mouse model of resiniferatoxin (RTX)-induced small fiber neuropathy. Mice with RTX neuropathy exhibited thermal hypoalgesia and mechanical allodynia, reduced skin innervation, and altered DRG expression profiles with decreased TrkA(+) neurons and increased Ret(+) neurons. RTX neuropathy induced the expression of activating transcription factor 3 (ATF3), and ATF3(+) neurons were colocalized with Ret but not with TrkA (P<0.001). As a neuroprotectant, 4-Methylcatechol (4MC) promoted skin reinnervation partially with correlated reversal of the neuropathic behaviors and altered neurochemical expression. Gambogic amide, a selective TrkA agonist, normalized thermal hypoalgesia, and GW441756, a TrkA kinase inhibitor, induced thermal hypoalgesia, which was already reversed by 4MC. Mechanical allodynia was reversed by a Ret kinase inhibitor, AST487, which induced thermal hyperalgesia in naïve mice. The activation of Ret signaling by XIB4035 induced mechanical allodynia and thermal hypoalgesia in RTX neuropathy mice in which the neuropathic behaviors were previously normalized by 4MC. Distinct neurotrophic factor receptors, TrkA and Ret, accounted for negative and positive neuropathic behaviors in RTX-induced small fiber neuropathy, respectively: TrkA for thermal hypoalgesia and Ret for mechanical allodynia and thermal hypoalgesia.


Subject(s)
Disease Models, Animal , Diterpenes/toxicity , Proto-Oncogene Proteins c-ret/metabolism , Receptor, trkA/metabolism , Small Fiber Neuropathy/chemically induced , Small Fiber Neuropathy/metabolism , Animals , Mice , Pain Measurement/drug effects , Pain Measurement/methods , Proto-Oncogene Proteins c-ret/agonists , Quinolines/pharmacology , Receptor, trkA/agonists , Small Fiber Neuropathy/pathology , Xanthones/pharmacology
12.
Sci Rep ; 7(1): 1467, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28469194

ABSTRACT

Kai-xin-san (KXS) is a famous Chinese medicinal formula applied for treating stress-related psychiatric diseases with the symptoms such as depression, forgetfulness and dizziness. In clinic, the composition ratio of KXS is always varied and KXS series formulae are created. Here, we aim to compare the anti-depressive effect of different ratios of KXS and reveal its action mechanism on regulation of neurotrophic factor system. Firstly, daily intra-gastric administration of chemically standardized extracts of KXS series formulae for seven days significantly alleviated the depressive symptoms of chronic unpredictable mild stressed mice displayed by enhanced sucrose consumptions and decreased immobile time of forced swimming coupled with increased locomotor activities. KXS might fulfill this effect by up-regulating the expressions of NGF, BDNF and Trk receptors in hippocampus, which were confirmed by the treatment of corresponding blockers tPA-stop and K252a. The ratio with higher amounts of Ginseng Radix et Rhizoma and Polygalae Radix exerted most profound effect on anti-depression and regulation enzymes in metabolic pathway of neurotrophic factors. These findings suggested that KXS was beneficial for enhancing supplies, up-regulating receptors, and restoring the dysfunction of metabolic pathway of neurotrophic factors, which might account for its anti-depression effect.


Subject(s)
Antidepressive Agents/pharmacology , Depression/drug therapy , Drugs, Chinese Herbal/pharmacology , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Stress, Psychological/drug therapy , Animals , Antidepressive Agents/chemistry , Brain-Derived Neurotrophic Factor/agonists , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Carbazoles/pharmacology , Depression/genetics , Depression/metabolism , Depression/physiopathology , Disease Models, Animal , Drugs, Chinese Herbal/chemistry , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Indole Alkaloids/pharmacology , Locomotion/drug effects , Male , Mice , Mice, Inbred ICR , Nerve Growth Factor/agonists , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Receptor, trkA/agonists , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/genetics , Receptor, trkA/metabolism , Stress, Psychological/genetics , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Swimming
13.
Vitam Horm ; 104: 1-18, 2017.
Article in English | MEDLINE | ID: mdl-28215291

ABSTRACT

Neurotrophins and their receptors (Trk) play key roles in the development of the nervous system and in cell survival. Trk receptors are therefore attractive pharmacological targets for brain disorders as well as for cancers. While the druggability of the extracellular domain of the receptors, that specifically binds neurotrophins, is yet to be proven, the intracellular kinase domains are attractive targets for small-molecule binding. The recent crystal structures of the three isoforms of the Trk family, TrkA, TrkB, and TrkC have been described in their apo forms and in complex with potent and selective pan-Trk inhibitors. The description of the kinase domain of each of the isoforms will be discussed in their apo forms or bound to potent inhibitors of interest in cancer therapy. Nononcology indications and selectivity issues will also be discussed.


Subject(s)
Membrane Glycoproteins/metabolism , Models, Molecular , Receptor, trkA/metabolism , Receptor, trkB/metabolism , Receptor, trkC/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoenzymes/chemistry , Apoenzymes/metabolism , Binding Sites , Catalytic Domain , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Ligands , Membrane Glycoproteins/agonists , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/chemistry , Molecular Conformation , Nerve Growth Factors/chemistry , Nerve Growth Factors/metabolism , Phenylalanine/chemistry , Protein Conformation , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor, trkA/agonists , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/chemistry , Receptor, trkB/agonists , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/chemistry , Receptor, trkC/agonists , Receptor, trkC/antagonists & inhibitors , Receptor, trkC/chemistry , Structural Homology, Protein
15.
Neuropharmacology ; 111: 266-282, 2016 12.
Article in English | MEDLINE | ID: mdl-27618740

ABSTRACT

Nerve growth factor (NGF) holds a pivotal role in brain development and maintenance, been also involved in the pathophysiology of neurodegenerative diseases. Here, we provide evidence that a novel C17-spiroepoxy steroid derivative, BNN27, specifically interacts with and activates the TrkA receptor of NGF, inducing phosphorylation of TrkA tyrosine residues and down-stream neuronal survival-related kinase signaling. Additionally, BNN27 potentiates the efficacy of low levels of NGF, by facilitating its binding to the TrkA receptors and differentially inducing fast return of internalized TrkA receptors into neuronal cell membranes. Furthermore, BNN27 synergizes with NGF in promoting axonal outgrowth, effectively rescues from apoptosis NGF-dependent and TrkA positive sympathetic and sensory neurons, in vitro, ex vivo and in vivo in NGF null mice. Interestingly, BNN27 does not possess the hyperalgesic properties of NGF. BNN27 represents a lead molecule for the development of neuroprotective TrkA receptor agonists, with potential therapeutic applications in neurodegenerative diseases and in brain trauma.


Subject(s)
Cell Differentiation/drug effects , Cell Survival/drug effects , Dehydroepiandrosterone/pharmacology , Nerve Growth Factor/pharmacology , Neurons/drug effects , Neurons/metabolism , Receptor, trkA/metabolism , Animals , Apoptosis/drug effects , Axons/drug effects , Axons/metabolism , Binding Sites , CHO Cells , Cricetulus , Dehydroepiandrosterone/chemistry , HEK293 Cells , Humans , Hyperalgesia/chemically induced , Mice , Mice, Knockout , Models, Molecular , Molecular Dynamics Simulation , Nerve Growth Factor/genetics , Neuroglia/drug effects , Neuroglia/metabolism , PC12 Cells , Phosphorylation , Rats , Receptor, trkA/agonists , Recombinant Proteins/pharmacology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Signal Transduction
16.
Article in Russian | MEDLINE | ID: mdl-26356399

ABSTRACT

Authors present an overview of theirs author's works on the design of low-molecular mimetic of the nerve growth factor and studies of mechanisms of action and pharmacological properties of the compound. The original working hypothesis, underlying the design of the compound, posited that different neurotrophin hairpin loops could activate different signaling cascades by interaction with the receptor and so be responsible for different effects. The mimetic bis(N-succinyl-L-glutamyl-L-lysine)hexametylendiamide (GK-2), that was designed on the basis of NGF loop 4 ß-turn sequence, activated TrkA and PI3K/Akt, but not MAPK/Erk. GK-2 showed neuroprotective activity in concentrations up to 10-9М against H(2)O(2) or glutamate or MPTP-induced neurotoxicity in РС12, НТ22 cells and primary rat hippocampal neurons. At that, GK-2 has no differentiating activity. In in vivo experiments, GK-2 exhibited significant anti-ischemic, anti-parkinsonic effect, reversed impaired cognitive functions in models of Alzheimer's disease in doses 0.01 - 5 mg/kg intraperitoneally and 5-10 mg/kg orally, but does not induce side effects accompanying the full-length neurotrophin treatment, which are hyperalgesia and weight loss. It was shown that GK-2 was a low-toxicity compound (LD50=700 mg/kg, intraperitoneally, mice) and capable of crossing the blood-brain barrier. The agent GK-2 is promising for development as a neuroprotective agent and is currently in preclinical studies.


Subject(s)
Dipeptides/chemistry , Dipeptides/pharmacology , Nerve Growth Factor/chemistry , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Animals , Blood-Brain Barrier/metabolism , Dipeptides/pharmacokinetics , Drug Design , Flavonoids/pharmacology , HSP70 Heat-Shock Proteins/biosynthesis , Heme Oxygenase-1/biosynthesis , Humans , Mice , PC12 Cells , Peptidomimetics/pharmacokinetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/agonists , Rats , Receptor, trkA/agonists
17.
Pharmacol Rep ; 67(2): 217-23, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25712642

ABSTRACT

BACKGROUND: Gambogic amide is the first identified small molecular agonist for TrkA receptor. It mimics NGF functions by selectively activating TrkA receptor and preventing neuron death. However, its function different from that of NGF remains unknown. METHODS: In the current study, we detect the effect of gambogic amide on TrkA expression using TrkA-expressing cell lines in vitro and hippocampi from mice treated with gambogic amide. RESULTS: We have confirmed that gambogic amide displays robust neurotrophic activities in provoking neurite outgrowth in vitro. However, gambiogic amide displays a different kinetics from NGF in activating TrkA signals. NGF swiftly provokes TrkA activation and quickly induces TrkA degradation, while gambogic amid selectively upregulates TrkA protein and mRNA levels in a time-dependent manner. Administration of this compound in mice also activates TrkA receptor in hippocampus and promotes TrkA transcription and expression. CONCLUSION: This study provides a novel mechanism of how gambogic amide regulates TrkA receptor, other than mimicking NGF in triggering TrkA activation.


Subject(s)
Receptor, trkA/agonists , Receptor, trkA/biosynthesis , Xanthones/pharmacology , Animals , Cell Line, Tumor , Enzyme Activation/drug effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Nerve Growth Factor/pharmacology , Nerve Growth Factors/pharmacology , Neurites/drug effects , Neurites/metabolism , Primary Cell Culture , Rats , Signal Transduction/drug effects , Substrate Specificity , Time Factors , Up-Regulation/drug effects
18.
Proc Natl Acad Sci U S A ; 111(47): 16943-8, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25385598

ABSTRACT

NGF binding to its protein kinase receptor TrkA is known to induce neurite outgrowth and neural cell differentiation. The plasma membrane expansion, necessary for the process, was shown to be contributed by the VAMP7-dependent exocytosis of endocytic vesicles. Working with wild-type PC12 (wtPC12), a cell model widely used to investigate NGF-induced neurite outgrowth, we found that a few hours of treatment with the neurotrophin (and to a lower extent with basic FGF and EGF) induces the appearance of enlargeosome vesicles competent for VAMP4-dependent exocytosis abundant in high REST-PC12 clones. Both the neurite length assay and the immunocytochemistry of enlargeosomes exocytosis revealed that activation of TrkA is induced not only by NGF, but also by the L1 adhesion protein, L1CAM, whose soluble construct binds the receptor with submicromolar affinity. In the intact wtPC12, the L1CAM construct induced autophosphorylation and internalization of TrkA followed by the activation of the PI3K, MEK, and PKCγ signaling cascades, analogous to the responses induced by NGF. Down-regulation of either VAMP7 or VAMP4 revealed the coparticipation of the two corresponding vesicles to the outgrowth responses induced by NGF and L1CAM. Finally, mixing experiments of wtPC12 cells rich in TrkA with high REST PC12 cells transfected with L1CAM documented the transactivation of the receptor by the adhesion protein surface-exposed in adjacent cells. In view of the known inhomogeneous surface distribution of both L1CAM and TrkA in various neural cells including neurons, their transcellular binding could be restricted to discrete sites, governing local signaling events distinct from those induced by soluble messengers.


Subject(s)
Exocytosis , Nerve Growth Factors/physiology , Neural Cell Adhesion Molecule L1/physiology , Neurites , Receptor, trkA/agonists , Animals , PC12 Cells , Rats
19.
J Cardiovasc Pharmacol ; 62(3): 270-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23644989

ABSTRACT

Nerve growth factor (NGF) has been reported to play an important role in physiological and pathological angiogenesis. Based on these observations, we hypothesized that NGF may induce the formation of functional blood vessels in a hindlimb ischemic rabbit model. Hindlimb ischemia was induced in 34 rabbits bilaterally by endovascular embolization of femoral arteries. On the 7th, 14th, and 20th postembolization days, NGF was injected intramuscularly, in 1 ischemic limb, and vehicle was injected in the contralateral control limb. On the 40th day, newly developed collateral vessels (diameter >500 µm) were quantified by transauricular intraarterial subtraction angiography. Perfusion analysis of an in vivo dynamic computed tomography study was performed to the limbs to investigate the hemodynamic recovery of the distal ischemic tissues. Functional estimation of limb perfusion showed a statistically significant increase of blood flow and blood volume for NGF. However, the increase of the collateral vessels was not detectable angiographically, providing evidence for the existence of a NGF-stimulated capillary angiogenic network but not increase of arteriogenesis. The combination of NGF with either tropomyosin-related kinase type A or vascular endothelial growth factor receptor 2 antagonists abolished the NGF-induced hemodynamic recovery. These findings provide new insights into understanding the involvement of NGF in vascular formation and its applications in therapeutic angiogenesis.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Disease Models, Animal , Ischemia/drug therapy , Muscle, Skeletal/drug effects , Nerve Growth Factor/therapeutic use , Receptor, trkA/agonists , Vascular Endothelial Growth Factor Receptor-2/agonists , Angiogenesis Inducing Agents/administration & dosage , Angiogenesis Inducing Agents/antagonists & inhibitors , Angiogenesis Inducing Agents/isolation & purification , Animals , Capillaries/diagnostic imaging , Capillaries/drug effects , Capillaries/pathology , Hemodynamics/drug effects , Hindlimb , Injections, Intramuscular , Ischemia/chemically induced , Ischemia/diagnostic imaging , Ischemia/pathology , Male , Mice , Muscle, Skeletal/blood supply , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/pathology , Neovascularization, Physiologic/drug effects , Nerve Growth Factor/administration & dosage , Nerve Growth Factor/antagonists & inhibitors , Nerve Growth Factor/isolation & purification , Protein Kinase Inhibitors/adverse effects , Rabbits , Radiography , Random Allocation , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/metabolism , Regional Blood Flow/drug effects , Signal Transduction/drug effects , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
20.
Cell Death Dis ; 3: e389, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22951986

ABSTRACT

Exploitation of the biologic activity of neurotrophins is desirable for medical purposes, but their protein nature intrinsically bears adverse pharmacokinetic properties. Here, we report synthesis and biologic characterization of a novel class of low molecular weight, non-peptidic compounds with NGF (nerve growth factor)-mimetic properties. MT2, a representative compound, bound to Trk (tropomyosin kinase receptor)A chain on NGF-sensitive cells, as well as in cell-free assays, at nanomolar concentrations and induced TrkA autophosphorylation and receptor-mediated internalization. MT2 binding involved at least two amino-acid residues within TrkA molecule. Like NGF, MT2 increased phosphorylation of extracellular signal-regulated kinase 1/2 and Akt proteins and production of MKP-1 phosphatase (dual specificity phosphatase 1), modulated p38 mitogen-activated protein kinase activation,sustained survival of serum-starved PC12 or RDG cells, and promoted their differentiation. However, the intensity of such responses was heterogenous, as the ability of maintaining survival was equally possessed by NGF and MT2, whereas the induction of differentiation was expressed at definitely lower levels by the mimetic. Analysis of TrkA autophosphorylation patterns induced by MT2 revealed a strong tyrosine (Tyr)490 and a limited Tyr785 and Tyr674/675 activation, findings coherent with the observed functional divarication. Consistently, in an NGF-deprived rat hippocampal neuronal model of Alzheimer Disease, MT2 could correct the biochemical abnormalities and sustain cell survival. Thus, NGF mimetics may reveal interesting investigational tools in neurobiology, as well as promising drug candidates.


Subject(s)
Azepines/pharmacology , Nerve Growth Factor/pharmacology , Receptor, trkA/agonists , Animals , Azepines/chemistry , Binding Sites , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Hippocampus/cytology , Hippocampus/metabolism , Humans , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Weight , NIH 3T3 Cells , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , PC12 Cells , Phosphorylation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, trkA/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...