Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
2.
Osteoarthritis Cartilage ; 29(10): 1389-1398, 2021 10.
Article in English | MEDLINE | ID: mdl-34284112

ABSTRACT

Osteoarthritis (OA) is a major health problem worldwide that affects the joints and causes severe disability. It is characterized by pain and low-grade inflammation. However, the exact pathogenesis remains unknown and the therapeutic options are limited. In OA articular chondrocytes undergo a phenotypic transition becoming hypertrophic, which leads to cartilage damage, aggravating the disease. Therefore, a therapeutic agent inhibiting hypertrophy would be a promising disease-modifying drug. The therapeutic use of tyrosine kinase inhibitors has been mainly focused on oncology, but the Food and Drug Administration (FDA) approval of the Janus kinase inhibitor Tofacitinib in Rheumatoid Arthritis has broadened the applicability of these compounds to other diseases. Interestingly, tyrosine kinases have been associated with chondrocyte hypertrophy. In this review, we discuss the experimental evidence that implicates specific tyrosine kinases in signaling pathways promoting chondrocyte hypertrophy, highlighting their potential as therapeutic targets for OA.


Subject(s)
Chondrocytes/pathology , Osteoarthritis/drug therapy , Protein Kinase Inhibitors/pharmacology , Discoidin Domain Receptors/physiology , ErbB Receptors/physiology , Focal Adhesion Protein-Tyrosine Kinases/physiology , Humans , Hypertrophy/drug therapy , Janus Kinase 2/physiology , Osteoarthritis/physiopathology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/physiology , Receptor Tyrosine Kinase-like Orphan Receptors/physiology , Receptor, IGF Type 1/physiology , Receptor, trkA/physiology , Receptors, Fibroblast Growth Factor/physiology , Signal Transduction
3.
Neuron ; 103(3): 412-422.e4, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31221560

ABSTRACT

Selective synaptic and axonal degeneration are critical aspects of both brain development and neurodegenerative disease. Inhibition of caspase signaling in neurons is a potential therapeutic strategy for neurodegenerative disease, but no neuron-specific modulators of caspase signaling have been described. Using a mass spectrometry approach, we discovered that RUFY3, a neuronally enriched protein, is essential for caspase-mediated degeneration of TRKA+ sensory axons in vitro and in vivo. Deletion of Rufy3 protects axons from degeneration, even in the presence of activated CASP3 that is competent to cleave endogenous substrates. Dephosphorylation of RUFY3 at residue S34 appears required for axon degeneration, providing a potential mechanism for neurons to locally control caspase-driven degeneration. Neuronally enriched RUFY3 thus provides an entry point for understanding non-apoptotic functions of CASP3 and a potential target to modulate caspase signaling specifically in neurons for neurodegenerative disease.


Subject(s)
Axons/pathology , Nerve Degeneration/pathology , Nerve Tissue Proteins/physiology , Animals , Axons/enzymology , Caspase 3/physiology , Cells, Cultured , Cytoskeletal Proteins , Enzyme Activation , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Mice , Mice, Knockout , Nerve Degeneration/enzymology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/deficiency , Phosphorylation , Protein Processing, Post-Translational , Receptor, trkA/physiology , Sensory Receptor Cells/physiology , Structure-Activity Relationship
4.
Nat Commun ; 10(1): 1745, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988299

ABSTRACT

Early childhood is a critical period for development, and early life stress may increase the risk of gastrointestinal diseases including irritable bowel syndrome (IBS). In rodents, neonatal maternal separation (NMS) induces bowel dysfunctions that resemble IBS. However, the underlying mechanisms remain unclear. Here we show that NMS induces expansion of intestinal stem cells (ISCs) and their differentiation toward secretory lineages including enterochromaffin (EC) and Paneth cells, leading to EC hyperplasia, increased serotonin production, and visceral hyperalgesia. This is reversed by inhibition of nerve growth factor (NGF)-mediated tropomyosin receptor kinase A (TrkA) signalling, and treatment with NGF recapitulates the intestinal phenotype of NMS mice in vivo and in mouse intestinal organoids in vitro. Mechanistically, NGF transactivates Wnt/ß-catenin signalling. NGF and serotonin are positively correlated in the sera of diarrhea-predominant IBS patients. Together, our findings provide mechanistic insights into early life stress-induced intestinal changes that may translate into treatments for gastrointestinal diseases.


Subject(s)
Gastrointestinal Diseases/etiology , Stress, Physiological , Animals , Enterochromaffin Cells/pathology , Humans , Hyperplasia/pathology , Maternal Deprivation , Mice , Nerve Growth Factor/metabolism , Nerve Growth Factor/physiology , Receptor, trkA/genetics , Receptor, trkA/metabolism , Receptor, trkA/physiology , Signal Transduction , Wnt Signaling Pathway
5.
Surg Today ; 49(9): 721-727, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30848386

ABSTRACT

Neuroblastoma is one of the most frequent, yet distinctive and challenging childhood tumors. The uniqueness of this tumor depends on its biological markers, which classify neuroblastomas into favorable and unfavorable, with 5-year survival rates ranging from almost 100-30%. In this review, we focus on some biological factors that play major roles in neuroblastoma: MYCN, Trk, and ALK. The MYCN and Trk family genes have been studied for decades and are known to be crucial for the tumorigenesis and progression of neuroblastoma. ALK gene mutations have been recognized recently to be responsible for familial neuroblastomas. Each factor plays an important role in normal neural development, regulating cell proliferation or differentiation by activating several signaling pathways, and interacting with each other. These factors have been studied not only as prognostic factors, but also as targets of neuroblastoma therapy, and some clinical trials are ongoing. We review the basic aspects of MYCN, Trk, and ALK in both neural development and in neuroblastoma.


Subject(s)
Anaplastic Lymphoma Kinase/physiology , Carcinogenesis/genetics , Membrane Glycoproteins/physiology , N-Myc Proto-Oncogene Protein/physiology , Nervous System/growth & development , Neuroblastoma/genetics , Receptor, trkA/physiology , Receptor, trkB/physiology , Cell Differentiation/genetics , Cell Proliferation/genetics , Child , Disease Progression , Humans , Mutation , Neuroblastoma/pathology , Signal Transduction
6.
Int Immunopharmacol ; 64: 52-59, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30145470

ABSTRACT

Nerve growth factor (NGF) has been shown to protect the viability of kidney cells in acute phase of renal damage. However, since the half-life of NGF is very short, it is too large to pass the blood-brain barrier and rapidly transported to the liver for catabolizing its use in therapy is limited. 4-Methylcatechol (4MC) is a substance that increases NGF synthesis in many tissues. This study aimed to investigate the protective effects of 4MC against acute renal injury induced by streptozotocin (STZ). We have investigated the profibrotic, proinflammatory, oxidative changes in STZ-induced acute renal damage and the possible role of the NGF/TrkA system and Akt/GSK3ß/ß-catenin pathway in this mechanism. Experiment was designed as to be started with injection of 4MC for 10 days as a single dose (10 µg/kg) per day and to be terminated after 4 h of a single dose (75 mg/kg) STZ injection. As the result, 4MC pre-treatment decreased kidney damage, ROS production, the renal levels of TGFß1, CD68, tumor necrosis factor-α and interleukin 1ß. Moreover, 4MC pre-treatment increased levels of NGF and its receptor TrkA, p-Akt (Thr308), p-GSK3ß (Ser9) and nuclear ß-catenin. These data suggest that 4MC prevents the development of STZ-induced renal damage by suppressing ROS production and inflammation via Akt/GSK3ß/ß-catenin pathway which may be stimulated by NGF/TrkA signaling. Therefore, 4MC can be suggested as a potential agent for the prevention of acute renal injury.


Subject(s)
Acute Kidney Injury/prevention & control , Catechols/pharmacology , Glycogen Synthase Kinase 3 beta/physiology , Nerve Growth Factor/physiology , Proto-Oncogene Proteins c-akt/physiology , Reactive Oxygen Species/metabolism , Receptor, trkA/physiology , Streptozocin/toxicity , beta Catenin/physiology , Acute Kidney Injury/chemically induced , Animals , Catechols/therapeutic use , Nerve Growth Factor/analysis , Rats , Rats, Wistar , Receptor, trkA/analysis , Signal Transduction/physiology
7.
Neurosci Biobehav Rev ; 87: 1-16, 2018 04.
Article in English | MEDLINE | ID: mdl-29407522

ABSTRACT

NGF is a well-studied neurotrophic factor, and TrkA is a receptor tyrosine kinase for NGF. The NGF-TrkA system supports the survival and maintenance of NGF-dependent neurons during development. Congenital insensitivity to pain with anhidrosis (CIPA) is an autosomal recessive genetic disorder due to loss-of-function mutations in the NTRK1 gene encoding TrkA. Individuals with CIPA lack NGF-dependent neurons, including NGF-dependent primary afferents and sympathetic postganglionic neurons, in otherwise intact systems. Thus, the pathophysiology of CIPA can provide intriguing findings to elucidate the unique functions that NGF-dependent neurons serve in humans, which might be difficult to evaluate in animal studies. Preceding studies have shown that the NGF-TrkA system plays critical roles in pain, itching and inflammation. This review focuses on the clinical and neurobiological aspects of CIPA and explains that NGF-dependent neurons in the peripheral nervous system play pivotal roles in interoception and homeostasis of our body, as well as in the stress response. Furthermore, these NGF-dependent neurons are likely requisite for neurobiological processes of 'emotions and feelings' in our species.


Subject(s)
Brain/physiopathology , Emotions/physiology , Hypohidrosis/physiopathology , Nerve Growth Factor/physiology , Neurons/physiology , Pain Insensitivity, Congenital/physiopathology , Animals , Humans , Hypohidrosis/complications , Hypohidrosis/psychology , Interoception , Pain Insensitivity, Congenital/complications , Pain Insensitivity, Congenital/psychology , Peripheral Nervous System/physiopathology , Receptor, trkA/physiology
8.
J Zhejiang Univ Sci B ; 17(10): 733-741, 2016.
Article in English | MEDLINE | ID: mdl-27704743

ABSTRACT

Extracellular signal-regulated protein kinase 5 (ERK5), also known as big mitogen-activated protein kinase 1 (MAPK1), is an important member of ERK family, which is a subfamily of the large MAPK family. ERK5 is expressed in many tissues, including the dorsal root ganglion (DRG) neurons and the spinal cord. In this review, we focus on elaborating ERK5-associated pathway in pathological pain, in which the ERK5/CREB (cyclic adenosine monophosphate (cAMP)-response element-binding protein) pathway plays a crucial role in the transduction of pain signal and contributes to pain hypersensitivity. ERK5 activation in the spinal dorsal horn occurs mainly in microglia. The activation of ERK5 can be mediated by N-methyl-D-aspartate (NMDA) receptors. We also elaborate the relationship between ERK5 activation and nerve growth factor-tyrosine kinase A (NGF-TrkA), and the connection between ERK5 activation and brain-derived neurotrophic factor (BDNF) in pathological pain in detail.


Subject(s)
MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 7/physiology , Pain/etiology , Animals , Brain-Derived Neurotrophic Factor/physiology , Cyclic AMP Response Element-Binding Protein/physiology , Humans , Nerve Growth Factor/physiology , Receptor, trkA/physiology , Receptors, N-Methyl-D-Aspartate/physiology
9.
Medicine (Baltimore) ; 95(18): e3559, 2016 May.
Article in English | MEDLINE | ID: mdl-27149473

ABSTRACT

Tricyclic antidepressant amitriptyline (AM) has been shown to exert neurotrophic activity on neurons. We thus explored whether AM may aid the neuronal development and protect anesthesia-induced neuro-injury in young spinal cord dorsal root ganglion (DRG) neurons.The DRG explants were prepared from 1-day-old rats. The effect of AM on aiding DRG neural development was examined by immunohistochemistry at dose-dependent manner. AM-induced changes in gene and protein expressions, and also phosphorylation states of tyrosine kinases receptor A (TrkA) and B (TrkB) in DRG, were examined by quantitative real-time polymerase chain reaction and western blot. The effect of AM on attenuating lidocaine-induced DRG neurodegeneration was examined by immunohistochemistry, and small interfering RNA (siRNA)-mediated TrkA/B down-regulation.Amitriptyline stimulated DRG neuronal development in dose-dependent manner, but exerted toxic effect at concentrations higher than 10 M. AM activated TrkA in DRG through phosphorylation, whereas it had little effect on TrkB-signaling pathway. AM reduced lidocaine-induced DRG neurodegeneration by regenerating neurites and growth cones. Moreover, the neuroprotection of AM on lidocaine-injured neurodegeneration was blocked by siRNA-mediated TrkA down-regulation, but not by TrkB down-regulation.Amitriptyline facilitated neuronal development and had protective effect on lidocaine-induced neurodegeneration, very likely through the activation of TrkA-signaling pathway in DRG.


Subject(s)
Amitriptyline/pharmacology , Anesthetics, Local/adverse effects , Antidepressive Agents, Tricyclic/pharmacology , Ganglia, Spinal/drug effects , Lidocaine/adverse effects , Nerve Degeneration/chemically induced , Receptor, trkA/drug effects , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Ganglia, Spinal/growth & development , Rats , Real-Time Polymerase Chain Reaction , Receptor, trkA/physiology , Receptor, trkB/drug effects , Receptor, trkB/physiology
10.
Implant Dent ; 25(3): 373-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27064695

ABSTRACT

BACKGROUND: Nerve growth factor (NGF) can, through its receptors TrkA and p75NTR, convey signals for cell survival or cell differentiation. These proteins are also involved in inflammation and in bone resorption. The aim of this study is to evaluate, for the first time, the expression of NGF and its receptors TrkA and p75NTR in peri-implantitis lesions. MATERIALS AND METHODS: Fifteen biopsy specimens from patients with chronic peri-implantitis and 4 of healthy oral mucosa were immunostained with antibodies against NGF, TrkA, and p75NTR. The staining intensity and percentage of stained cells were semi-quantitatively evaluated and results were compared between the 2 groups. RESULTS: In the peri-implant pocket epithelium and gingival epithelium, NGF and TrkA expressions were similar to the healthy oral mucosa, however, a decreased expression of p75NTR was observed. In all cases, more than 75% of the inflammatory cells stained positively for NGF and TrkA, and p75NTR was negatively expressed. CONCLUSION: The intense expression of NGF and TrkA in the inflammatory cell infiltrate associated with decreased expression of p75NTR in both gingival and pocket epithelium suggests that these proteins may have a role in peri-implantitis lesions.


Subject(s)
Nerve Growth Factor/metabolism , Nerve Tissue Proteins/metabolism , Peri-Implantitis/metabolism , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/metabolism , Adult , Aged , Aged, 80 and over , Female , Gingiva/metabolism , Gingiva/pathology , Gingiva/physiopathology , Humans , Male , Middle Aged , Mouth Mucosa/metabolism , Mouth Mucosa/pathology , Mouth Mucosa/physiology , Nerve Growth Factor/physiology , Nerve Tissue Proteins/physiology , Peri-Implantitis/pathology , Peri-Implantitis/physiopathology , Receptor, trkA/physiology , Receptors, Nerve Growth Factor/physiology , Retrospective Studies
11.
J Neurosci ; 36(15): 4259-75, 2016 Apr 13.
Article in English | MEDLINE | ID: mdl-27076424

ABSTRACT

Growth of intact axons of noninjured neurons, often termed collateral sprouting, contributes to both adaptive and pathological plasticity in the adult nervous system, but the intracellular factors controlling this growth are largely unknown. An automated functional assay of genes regulated in sensory neurons from the rat in vivo spared dermatome model of collateral sprouting identified the adaptor protein CD2-associated protein (CD2AP; human CMS) as a positive regulator of axon growth. In non-neuronal cells, CD2AP, like other adaptor proteins, functions to selectively control the spatial/temporal assembly of multiprotein complexes that transmit intracellular signals. Although CD2AP polymorphisms are associated with increased risk of late-onset Alzheimer's disease, its role in axon growth is unknown. Assessments of neurite arbor structure in vitro revealed CD2AP overexpression, and siRNA-mediated knockdown, modulated (1) neurite length, (2) neurite complexity, and (3) growth cone filopodia number, in accordance with CD2AP expression levels. We show, for the first time, that CD2AP forms a novel multiprotein complex with the NGF receptor TrkA and the PI3K regulatory subunit p85, with the degree of TrkA:p85 association positively regulated by CD2AP levels. CD2AP also regulates NGF signaling through AKT, but not ERK, and regulates long-range signaling though TrkA(+)/RAB5(+) signaling endosomes. CD2AP mRNA and protein levels were increased in neurons during collateral sprouting but decreased following injury, suggesting that, although typically considered together, these two adult axonal growth processes are fundamentally different. These data position CD2AP as a major intracellular signaling molecule coordinating NGF signaling to regulate collateral sprouting and structural plasticity of intact adult axons. SIGNIFICANCE STATEMENT: Growth of noninjured axons in the adult nervous system contributes to adaptive and maladaptive plasticity, and dysfunction of this process may contribute to neurologic pathologies. Functional screening of genes regulated during growth of noninjured axons revealed CD2AP as a positive regulator of axon outgrowth. A novel association of CD2AP with TrkA and p85 suggests a distinct intracellular signaling pathway regulating growth of noninjured axons. This may also represent a novel mechanism of generating specificity in multifunctional NGF signaling. Divergent regulation of CD2AP in different axon growth conditions suggests that separate mechanisms exist for different modes of axon growth. CD2AP is the first signaling molecule associated with adult sensory axonal collateral sprouting, and this association may offer new insights for NGF/TrkA-related Alzheimer's disease mechanisms.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Axons/physiology , Cytoskeletal Proteins/physiology , Nerve Growth Factors/physiology , Neuronal Plasticity/physiology , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Differentiation/genetics , Class Ia Phosphatidylinositol 3-Kinase/physiology , Cytoskeletal Proteins/genetics , Endosomes/metabolism , Female , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/genetics , Pseudopodia/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Receptor, trkA/physiology , Signal Transduction/genetics
12.
Domest Anim Endocrinol ; 56: 20-8, 2016 07.
Article in English | MEDLINE | ID: mdl-26986844

ABSTRACT

The aim of the present study was to evaluate: (1) the presence of nerve growth factor (NGF), neurotrophic tyrosine kinase receptor 1 (NTRK1), and nerve growth factor receptor (NGFR) in the rabbit uterus; and (2) the in vitro effects of NGF on PGF2α and PGE2 synthesis and on the PGE2-9-ketoreductase (PGE2-9-K) activity by the rabbit uterus. Nerve growth factor, NTRK1, and NGFR were immunolocalized in the luminal and glandular epithelium and stroma cells of the endometrium. reverse transcriptase polymerase chain reaction indicated the presence of messenger RNA for NGF, NTRK1, and NGFR in the uterus. Nerve growth factor increased (P < 0.01) in vitro secretions of PGF2α and PGE2 but coincubation with either NTRK1 or oxide nitric synthase (NOS) inhibitors reduced (P < 0.01) PGF2α production and blocked (P < 0.01) PGE2 secretion. Prostaglandins releases were lower (P < 0.01) than control when uterine samples were treated with NGF plus cyclooxygenase inhibitor. However, addition of NGFR inhibitor reduced (P < 0.01) PGF2α secretion less efficiently than NTRK1 or NOS inhibitors but had no effect on PGE2 yield. Nerve growth factor increased (P < 0.01) the activity of PGE2-9-K, whereas coincubation with NTRK1 or NOS inhibitors abolished (P < 0.01) this increase in PGE2-9-K activity. However, cotreatment with either cyclooxygenase or NGFR inhibitors had no effect on PGE2-9-K activity. This is the first study to document the distribution of NGF/NTRK1 and NGFR systems and their effects on prostaglandin synthesis in the rabbit uterus. NGF/NTRK1 increases PGF2α and PGE2 productions by upregulating NOS and PGE2-9-K activities, whereas NGF/NGFR augments only PGF2α secretion, through an intracellular mechanism that is still unknown.


Subject(s)
Gene Expression , Nerve Growth Factor/genetics , Prostaglandins/biosynthesis , Rabbits/metabolism , Receptors, Nerve Growth Factor/genetics , Uterus/metabolism , Animals , Dinoprost/biosynthesis , Female , Hydroxyprostaglandin Dehydrogenases/metabolism , Immunohistochemistry , Nerve Growth Factor/analysis , Nerve Growth Factor/pharmacology , RNA, Messenger/analysis , Receptor, trkA/analysis , Receptor, trkA/genetics , Receptor, trkA/physiology , Receptors, Nerve Growth Factor/analysis , Receptors, Nerve Growth Factor/physiology , Uterus/chemistry
13.
Ann Rheum Dis ; 75(6): 1246-54, 2016 06.
Article in English | MEDLINE | ID: mdl-26286016

ABSTRACT

OBJECTIVES: Tropomyosin receptor kinase A (TrkA) mediates nociceptor sensitisation by nerve growth factor (NGF), but it is unknown whether selective TrkA inhibition will be an effective strategy for treating osteoarthritis (OA) pain. We determined the effects of a TrkA inhibitor (AR786) on pain behaviour, synovitis and joint pathology in two rat OA models. METHODS: Knee OA was induced in rats by intra-articular monosodium-iodoacetate (MIA) injection or meniscal transection (MNX) and compared with saline-injected or sham-operated controls. Pain behaviour was assessed as weight-bearing asymmetry and paw withdrawal threshold to punctate stimulation. Oral doses (30 mg/kg) of AR786 or vehicle were administered twice daily in either preventive (day -1 to -27) or treatment (day 14-28) protocols. Effect maintenance was evaluated for 2 weeks after treatment discontinuation. Alterations in knee structure (cartilage, subchondral bone and synovium) were examined by macroscopic visualisation of articular surfaces and histopathology. RESULTS: Preventive AR786 treatment inhibited pain behaviour development and therapeutic treatment attenuated established pain behaviour. Weight-bearing asymmetry increased 1 week after treatment discontinuation, but remained less than in vehicle-treated arthritic rats, whereas paw withdrawal thresholds returned to levels of untreated rats within 5 days of treatment discontinuation. AR786 treatment reduced MIA-induced synovitis and did not significantly affect osteochondral pathology in either model. CONCLUSIONS: Blocking NGF activity by inhibiting TrkA reduced pain behaviour in two rat models of OA. Analgesia was observed both using preventive and treatment protocols, and was sustained after treatment discontinuation. Selective inhibitors of TrkA therefore hold potential for OA pain relief.


Subject(s)
Analgesics, Non-Narcotic/therapeutic use , Arthritis, Experimental/drug therapy , Osteoarthritis/drug therapy , Pain/prevention & control , Receptor, trkA/antagonists & inhibitors , Analgesics, Non-Narcotic/pharmacology , Animals , Arthritis, Experimental/complications , Arthritis, Experimental/pathology , Drug Evaluation, Preclinical/methods , Iodoacetic Acid , Male , Meniscus/surgery , Osteoarthritis/complications , Osteoarthritis/pathology , Pain/etiology , Pain Measurement/methods , Pain Threshold/drug effects , Rats, Sprague-Dawley , Receptor, trkA/physiology , Synovitis/pathology , Synovitis/prevention & control , Weight-Bearing
14.
Toxicol In Vitro ; 30(1 Pt B): 231-40, 2015 Dec 25.
Article in English | MEDLINE | ID: mdl-26556726

ABSTRACT

Cannabidiol (CBD) is a non-psychoactive constituent of Cannabis sativa with potential to treat neurodegenerative diseases. Its neuroprotection has been mainly associated with anti-inflammatory and antioxidant events; however, other mechanisms might be involved. We investigated the involvement of neuritogenesis, NGF receptors (trkA), NGF, and neuronal proteins in the mechanism of neuroprotection of CBD against MPP(+) toxicity in PC12 cells. CBD increased cell viability, differentiation, and the expression of axonal (GAP-43) and synaptic (synaptophysin and synapsin I) proteins. Its neuritogenic effect was not dependent or additive to NGF, but it was inhibited by K252a (trkA inhibitor). CBD did not increase the expression of NGF, but protected against its decrease induced by MPP(+), probably by an indirect mechanism. We also evaluated the neuritogenesis in SH-SY5Y cells, which do not express trkA receptors. CBD did not induce neuritogenesis in this cellular model, which supports the involvement of trkA receptors. This is the first study to report the involvement of neuronal proteins and trkA in the neuroprotection of CBD. Our findings suggest that CBD has a neurorestorative potential independent of NGF that might contribute to its neuroprotection against MPP(+), a neurotoxin relevant to Parkinson's disease.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Cannabidiol/pharmacology , Nerve Tissue Proteins/biosynthesis , Neurites/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease/prevention & control , Receptor, trkA/physiology , Animals , Axons/metabolism , Humans , Nerve Growth Factor/physiology , Neurites/physiology , Neuroblastoma/pathology , PC12 Cells , Rats , Synapses/metabolism , Up-Regulation
15.
J Dermatol Sci ; 78(3): 215-23, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25823576

ABSTRACT

BACKGROUND: Nerve growth factor (NGF), a neurotrophin that plays a critical role in developmental neurobiology, is released by proliferating keratinocytes and induces proliferation. OBJECTIVE: The aim of this study was to investigate the role of tyrosine kinase receptor A (TrkA), a high-affinity receptor of NGF, in human keratinocytes. METHODS: Expression of TrkA and NGF in skin diseases was investigated by immunohistochemistry. Expression of TrkA in cells was examined by Western blotting and RT-PCR. Cell proliferation was assessed by BrdU assay. RESULTS: We first determined the expression of TrkA and NGF in skin samples from patients with atopic dermatitis, prurigo nodularis, psoriasis vulgaris, and seborrheic keratosis. TrkA was only expressed in proliferating basal cells, and its expression was enhanced in atopic dermatitis samples. NGF expression was enhanced in atopic dermatitis and prurigo nodularis samples and in some samples from seborrheic keratosis patients. Investigation of the role of TrkA in vitro using normal human epidermal keratinocytes (NHEK) revealed that TrkA was significantly enhanced by the T helper type 2 (Th2) cytokines interleukin (IL)-4 and IL-13 but not by other inflammatory cytokines, such as IL-1ß, tumor necrosis factor α, interferon γ, or epidermal growth factor. On the other hand, expression of NGF was not altered by Th2 cytokines. Notably, inhibition of TrkA significantly reversed the effects of IL-4 on proliferation and differentiation. Furthermore, overexpression of TrkA enhanced proliferation of NHEK. These results indicate that IL-4-induced TrkA expression in keratinocytes modulates proliferation and differentiation of these cells. CONCLUSION: Increased TrkA expression in keratinocytes in atopic dermatitis may contribute to the observed epidermal hyperproliferation in these patients.


Subject(s)
Interleukin-4/pharmacology , Keratinocytes/drug effects , Receptor, trkA/physiology , Th2 Cells/immunology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Humans , Interleukin-13/pharmacology , Keratinocytes/cytology , Nerve Growth Factor/analysis , Receptor, trkA/analysis , Up-Regulation
16.
J Orthop Res ; 33(8): 1235-41, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25876530

ABSTRACT

Abatement of fracture-related pain is important in patient welfare. However, the frequently used non-steroidal anti-inflammatory drugs are considered to impair fracture healing through blockade of cyclooxygenase-2. An alternative for fracture-related pain treatment may be blockade of nerve growth factor (NGF)/neurotrophic tyrosine kinase receptor type 1 (TrkA) signaling. Because the effect of blocking this signal-pathway on bone healing has not been extensively investigated, we addressed this issue by applying neutralizing antibodies that target NGF and TrkA, respectively, in a mouse fracture model. Mice with a knock-in for human TrkA underwent femur osteotomy and were randomly allocated to phosphate-buffered-saline, anti-NGF-antibody, or anti-TrkA-antibody treatment. The analgesic effect of the antibodies was determined from the activity and the ground reaction force of the operated limb. The effect of antibody administration on fracture healing was assessed by histomorphometry, micro-computed tomography, and biomechanics. NGF/TrkA-signaling blockade had no negative effect on fracture healing as callus formation and maturation were not altered. Mice treated with anti-TrkA antibody displayed significantly greater activity on post-operative day 2 compared to PBS treatment indicating effective analgesia. Our data indicate, that blockade of NGF/TrkA signaling via specific neutralizing antibodies for pain reduction during fracture healing does not influence fracture healing.


Subject(s)
Analgesia , Fracture Healing/physiology , Nerve Growth Factors/physiology , Receptor, trkA/physiology , Signal Transduction/physiology , Animals , Fractures, Bone/physiopathology , Male , Mice , Nerve Growth Factors/antagonists & inhibitors , Pain/drug therapy , Pain/physiopathology , Receptor, trkA/antagonists & inhibitors
17.
J Neurosci ; 35(9): 3893-902, 2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25740518

ABSTRACT

Development of a functional peripheral nervous system requires axons to rapidly innervate and arborize into final target organs and then slow but not halt their growth to establish stable connections while keeping pace with organ growth. Here we examine the role of the NGF-TrkA effector protein, Coronin-1, on postganglionic sympathetic neuron final target innervation. In the absence of Coronin-1 we find that NGF-TrkA-PI3K signaling drives robust axon growth and branching in part by suppressing GSK3ß. In contrast, the presence of Coronin-1 (wild-type neurons) suppresses but does not halt NGF-TrkA-dependent growth and branching. This relative suppression in axon growth behaviors is due to Coronin-1-dependent calcium release via PLC-γ1 signaling, which releases PI3K-dependent suppression of GSK3ß. Finally, we demonstrate that Coro1a(-/-) mice display sympathetic axon overgrowth and overbranching phenotypes in the developing heart. Together with previous work demonstrating the Coronin-1 expression is NGF dependent, this work suggests that periods before and after NGF-TrkA-induced Coronin-1 expression (and likely other factors) defines two distinct axon growth states, which are critical for proper circuit formation in the sympathetic nervous system.


Subject(s)
Calcium Signaling/physiology , Microfilament Proteins/physiology , Sympathetic Nervous System/physiology , Animals , Axons/physiology , Cells, Cultured , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/physiology , Glycogen Synthase Kinase 3 beta , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/physiology , Nerve Growth Factor/physiology , Phosphatidylinositol 3-Kinases/physiology , Receptor, trkA/physiology , ras Proteins/physiology
18.
Exp Eye Res ; 132: 34-47, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25584870

ABSTRACT

Müller cells can completely repair retinal injury by acting as endogenous stem/progenitor cells in lower-order vertebrates. However, a safe and effective approach to activate progenitor potential of retinal Müller cells in higher-order vertebrates, which rarely re-enter the cell cycle, is a bottleneck problem. In the present study, Royal College of Surgeon's (RCS) rats were subjected to rat bone marrow mesenchymal stem cells (rBMSCs) subretinal space transplantation. Electroretinography (ERG) recordings showed that the b-wave amplitudes and ONL thicknesses statistically increased after transplantation. The number of Müller cells expressing proliferative, stem/progenitor and neuronal markers significantly increased after rBMSCs transplantation in vivo or after co-culturing with rBMSCs in vitro. The cultured rBMSCs could secrete nerve growth factor (NGF). In addition, we confirmed that NGF or NGF-neutralizing antibody could activate or depress Müller cells dedifferentiation, both in vivo and in vitro. Furthermore, Müller cells expressing high levels of the NGF receptor neurotrophic tyrosine kinase receptor type 1 (TrkA) were observed in the retinas of rats transplanted with rBMSCs. Moreover, the protein expression of downstream elements of NGF/TrkA signaling, such as p-PI3K, p-Akt and p-CREB, increased in Müller cells in the retinas of rBMSCs-treated rats in vivo or in Müller cells co-cultured with rBMSCs in vitro. Blocking TrkA with K-252a reduced the number of dedifferentiated Müller cells and the expression of NGF/TrkA signaling in vitro. Thus, rBMSCs might initiate endogenous regenerative mechanisms, which may constitute a new therapeutic strategy for retinal dystrophic diseases.


Subject(s)
Bone Marrow Cells/cytology , Ependymoglial Cells/physiology , Mesenchymal Stem Cell Transplantation , Nerve Growth Factor/physiology , Receptor, trkA/physiology , Retinitis Pigmentosa/surgery , Signal Transduction/physiology , Analysis of Variance , Animals , Blotting, Western , Cell Differentiation/physiology , Disease Models, Animal , Electroretinography , Enzyme-Linked Immunosorbent Assay , Rats , Real-Time Polymerase Chain Reaction , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/physiopathology , Visual Perception/physiology
19.
Neuron ; 82(3): 587-602, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24811380

ABSTRACT

Primary nociceptors relay painful touch information from the periphery to the spinal cord. Although it is established that signals generated by receptor tyrosine kinases TrkA and Ret coordinate the development of distinct nociceptive circuits, mechanisms modulating TrkA or Ret pathways in developing nociceptors are unknown. We have identified tumor necrosis factor (TNF) receptor 1 (TNFR1) as a critical modifier of TrkA and Ret signaling in peptidergic and nonpeptidergic nociceptors. Specifically, TrkA+ peptidergic nociceptors require TNF-α-TNFR1 forward signaling to suppress nerve growth factor (NGF)-mediated neurite growth, survival, excitability, and differentiation. Conversely, TNFR1-TNF-α reverse signaling augments the neurite growth and excitability of Ret+ nonpeptidergic nociceptors. The developmental and functional nociceptive defects associated with loss of TNFR1 signaling manifest behaviorally as lower pain thresholds caused by increased sensitivity to NGF. Thus, TNFR1 exerts a dual role in nociceptor information processing by suppressing TrkA and enhancing Ret signaling in peptidergic and nonpeptidergic nociceptors, respectively.


Subject(s)
Nociceptors/physiology , Pain Measurement/methods , Receptors, Tumor Necrosis Factor, Type I/physiology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Cell Survival/physiology , Cells, Cultured , Down-Regulation/physiology , Mice , Mice, 129 Strain , Mice, Knockout , Neurons/physiology , Proto-Oncogene Proteins c-ret/physiology , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/physiology , Up-Regulation/physiology
20.
Handb Exp Pharmacol ; 220: 103-19, 2014.
Article in English | MEDLINE | ID: mdl-24668471

ABSTRACT

The tropomyosin-related tyrosine kinase (Trk) receptors were initially described as a family of growth factor receptors required for neuronal survival. They have since been shown to influence many aspects of neuronal development and function, including differentiation, outgrowth, and synaptic plasticity. This chapter will give an overview on the biology of Trk receptors within the nervous system. The structure and downstream signaling pathways of the full-length receptors will be described, as well as the biological functions of their truncated isoforms. Finally, the role of Trk receptors in the nervous system in health and disease will be discussed.


Subject(s)
Receptor, trkA/physiology , Receptor, trkB/physiology , Receptor, trkC/physiology , Animals , Axonal Transport , Humans , Neuronal Plasticity , Receptor, trkA/chemistry , Receptor, trkB/chemistry , Receptor, trkC/chemistry , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...