Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 711
Filter
1.
Life Sci ; 286: 120067, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34678261

ABSTRACT

AIMS: Brain derived neurotrophic factor (BDNF) and the related receptors TrkB and p75NTR are expressed in skeletal muscle, yet their functions remain to be fully understood. Skeletal muscle denervation, which occurs in spinal injury, peripheral neuropathies, and aging, negatively affects muscle mass and function. In this study, we wanted to understand the role of BDNF, TrkB, and p75NTR in denervation-induced adverse effects on skeletal muscle. MAIN METHODS: Mice with unilateral sciatic denervation were used. Protein levels of pro- and mature BDNF, TrkB, p75NTR, activations of their downstream signaling pathways, and inflammation in the control and denervated muscle were measured with Western blot and tissue staining. Treatment with a p75NTR inhibitor and BDNF skeletal muscle specific knockout in mice were used to examine the role of p75NTR and pro-BDNF. KEY FINDINGS: In denervated muscle, pro-BDNF and p75NTR were significantly upregulated, and JNK and NF-kB, two major downstream signaling pathways of p75NTR, were activated, along with muscle atrophy and inflammation. Inhibition of p75NTR using LM11A-31 significantly reduced JNK activation and inflammatory cytokines in the denervated muscle. Moreover, skeletal muscle specific knockout of BDNF reduced pro-BDNF level, JNK activation and inflammation in the denervated muscle. SIGNIFICANCE: These results reveal for the first time that the upregulation of pro-BDNF and activation of p75NTR pathway are involved in denervation-induced inflammation in skeletal muscle. The results suggest that inhibition of pro-BDNF-p75NTR pathway can be a new target to treat skeletal muscle inflammation.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Muscle, Skeletal/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Brain-Derived Neurotrophic Factor/physiology , Female , Isoleucine/analogs & derivatives , Isoleucine/pharmacology , Male , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , Morpholines/pharmacology , Muscle Denervation/methods , Muscle, Skeletal/physiology , Muscular Atrophy/metabolism , Peripheral Nervous System Diseases , Protein Precursors/metabolism , Protein Precursors/physiology , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/physiology , Receptors, Nerve Growth Factor/physiology , Signal Transduction/physiology
2.
Behav Brain Res ; 404: 113162, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33549684

ABSTRACT

Neurotrophic factors, which can provide nutritional support to neurons and neuronal cells, also played an important role in their proliferation and survival. As signaling molecules, it also mediated the learning, memory and other activities in the brain. The latest study shows that neurotrophic factors have diametrically opposing effects of the pro- and mature form through distinct receptors. In this review, we summarize the different forms of neurotrophic factors, related receptors, and the corresponding biological effects. More importantly, we expounded the physiology and pathology mechanisms of brain-derived neurotrophic factor(BDNF)in depression. It is hopefully to provide new idea on the relationship of neurotrophic factors and depression.


Subject(s)
Depression/etiology , Nerve Growth Factors/physiology , Animals , Depression/metabolism , Depression/physiopathology , Humans , Nerve Growth Factors/metabolism , Receptors, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/physiology
3.
Cell Biol Int ; 45(2): 432-446, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33200854

ABSTRACT

Neural stem cells (NSCs) or neuronal progenitor cells are cells capable of differentiating into oligodendrocytes, myelin-forming cells that have the potential of remyelination. Brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) are two neurotrophic factors that have been studied to stimulate NSC differentiation thus playing a role in multiple sclerosis pathogenesis and several other demyelinating disorders. While several studies have demonstrated the proliferative and protective capabilities of these neurotrophic factors, their cellular and molecular functions are still not well understood. Thus, in the present study, we focus on understanding the role of these neurotrophins (BDNF and NGF) in oligodendrogenesis from NSCs. Both neurotrophic factors have been shown to promote NSC proliferation and NSC differentiation particularly into oligodendroglial lineage in a dose-dependent fashion. Further, to establish the role of these neurotrophins in NSC differentiation, we have employed pharmacological inhibitors for TrkA and TrkB receptors in NSCs. The use of these inhibitors suppressed NSC differentiation into oligodendrocytes along with the downregulation of phosphorylated ERK suggesting active involvement of ERK in the functioning of these neurotrophins. The morphometric analysis also revealed the important role of both neurotrophins in oligodendrocytes development. These findings highlight the importance of neurotrophic factors in stimulating NSC differentiation and may pave a role for future studies to develop neurotrophic factor replacement therapies to achieve remyelination.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Nerve Growth Factor/physiology , Neural Stem Cells , Receptor, trkB/physiology , Receptors, Nerve Growth Factor/physiology , Animals , Cell Differentiation , Cells, Cultured , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Oligodendroglia/cytology
4.
Sci Rep ; 10(1): 11145, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32636407

ABSTRACT

Osteoarthritis (OA), the most common joint disorder, is characterised by progressive structural changes in both the cartilage and the underlying subchondral bone. In late disease stages, subchondral bone sclerosis has been linked to heightened osteogenic commitment of bone marrow stromal cells (BMSCs). This study utilised cell sorting and immunohistochemistry to identify a phenotypically-distinct, osteogenically-committed BMSC subset in human OA trabecular bone. Femoral head trabecular bone tissue digests were sorted into CD45-CD271+CD56+CD146-, CD45-CD271+CD56-CD146+ and CD45-CD271+CD56-CD146-(termed double-negative, DN) subsets, and CD45+CD271-hematopoietic-lineage cells served as control. Compared to the CD146+ subset, the CD56+ subset possessed a lower-level expression of adipocyte-associated genes and significantly over 100-fold higher-level expression of many osteoblast-related genes including osteopontin and osteocalcin, whilst the DN subset presented a transcriptionally 'intermediate' BMSC population. All subsets were tri-potential following culture-expansion and were present in control non-OA trabecular bone. However, while in non-OA bone CD56+ cells only localised on the bone surface, in OA bone they were additionally present in the areas of new bone formation rich in osteoblasts and newly-embedded osteocytes. In summary, this study reveals a distinct osteogenically-committed CD271+CD56+ BMSC subset and implicates it in subchondral bone sclerosis in hip OA. CD271+CD56+ subset may represent a future therapeutic target for OA and other bone-associated pathologies.


Subject(s)
CD56 Antigen/metabolism , Femur Head/metabolism , Mesenchymal Stem Cells/physiology , Nerve Tissue Proteins/metabolism , Osteoarthritis/metabolism , Osteogenesis , Receptors, Nerve Growth Factor/metabolism , Adult , Aged , Aged, 80 and over , CD56 Antigen/physiology , Cancellous Bone/metabolism , Cancellous Bone/pathology , Case-Control Studies , Female , Femur Head/pathology , Flow Cytometry , Humans , Male , Mesenchymal Stem Cells/pathology , Middle Aged , Nerve Tissue Proteins/physiology , Osteoarthritis/pathology , Osteogenesis/physiology , Receptors, Nerve Growth Factor/physiology
5.
J Neurosci ; 39(48): 9503-9520, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31628183

ABSTRACT

The regressive events associated with trophic deprivation are critical for sculpting a functional nervous system. After nerve growth factor withdrawal, sympathetic axons derived from male and female neonatal mice maintain their structural integrity for ∼18 h (latent phase) followed by a rapid and near unison disassembly of axons over the next 3 h (catastrophic phase). Here we examine the molecular basis by which axons transition from latent to catastrophic phases of degeneration following trophic withdrawal. Before catastrophic degeneration, we observed an increase in intra-axonal calcium. This calcium flux is accompanied by p75 neurotrophic factor receptor-Rho-actin-dependent expansion of calcium-rich axonal spheroids that eventually rupture, releasing their contents to the extracellular space. Conditioned media derived from degenerating axons are capable of hastening transition into the catastrophic phase of degeneration. We also found that death receptor 6, but not p75 neurotrophic factor receptor, is required for transition into the catastrophic phase in response to conditioned media but not for the intra-axonal calcium flux, spheroid formation, or rupture that occur toward the end of latency. Our results support the existence of an interaxonal degenerative signal that promotes catastrophic degeneration among trophically deprived axons.SIGNIFICANCE STATEMENT Developmental pruning shares several morphological similarities to both disease- and injury-induced degeneration, including spheroid formation. The function and underlying mechanisms governing axonal spheroid formation, however, remain unclear. In this study, we report that axons coordinate each other's degeneration during development via axonal spheroid rupture. Before irreversible breakdown of the axon in response to trophic withdrawal, p75 neurotrophic factor receptor-RhoA signaling governs the formation and growth of spheroids. These spheroids then rupture, allowing exchange of contents ≤10 kDa between the intracellular and extracellular space to drive death receptor 6 and calpain-dependent catastrophic degeneration. This finding informs not only our understanding of regressive events during development but may also provide a rationale for designing new treatments toward myriad neurodegenerative disorders.


Subject(s)
Axons/metabolism , Nerve Degeneration/metabolism , Receptors, Nerve Growth Factor/physiology , Receptors, Tumor Necrosis Factor/physiology , Spheroids, Cellular/metabolism , Animals , Axons/pathology , Cells, Cultured , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/pathology , Spheroids, Cellular/pathology
6.
J Neurosci ; 39(46): 9119-9129, 2019 11 13.
Article in English | MEDLINE | ID: mdl-31582529

ABSTRACT

Development of brain circuitry requires precise regulation and timing of proliferation and differentiation of neural progenitor cells. The p75 neurotrophin receptor (p75NTR) is highly expressed in the proliferating granule cell precursors (GCPs) during development of the cerebellum. In a previous paper, we showed that proNT3 promoted GCP cell cycle exit via p75NTR. Here we used genetically modified rats and mice of both sexes to show that p75NTR regulates the duration of the GCP cell cycle, requiring activation of RhoA. Rats and mice lacking p75NTR have dysregulated GCP proliferation, with deleterious effects on cerebellar circuit development and behavioral consequences persisting into adulthood. In the absence of p75NTR, the GCP cell cycle is accelerated, leading to delayed cell cycle exit, prolonged GCP proliferation, increased glutamatergic input to Purkinje cells, and a deficit in delay eyeblink conditioning, a cerebellum-dependent form of learning. These results demonstrate the necessity of appropriate developmental timing of the cell cycle for establishment of proper connectivity and associated behavior.SIGNIFICANCE STATEMENT The cerebellum has been shown to be involved in numerous behaviors in addition to its classic association with motor function. Cerebellar function is disrupted in a variety of psychiatric disorders, including those on the autism spectrum. Here we show that the p75 neurotrophin receptor, which is abundantly expressed in the proliferating cerebellar granule cell progenitors, regulates the cell cycle of these progenitors. In the absence of this receptor, the cell cycle is dysregulated, leading to excessive progenitor proliferation, which alters the balance of inputs to Purkinje cells, disrupting the circuitry and leading to functional deficits that persist into adulthood.


Subject(s)
Cell Cycle/physiology , Cerebellum/growth & development , Neural Stem Cells/physiology , Neurons/physiology , Receptors, Nerve Growth Factor/physiology , Animals , Cell Proliferation , Dendritic Spines/physiology , Dendritic Spines/ultrastructure , Excitatory Postsynaptic Potentials , Female , Male , Mice, Transgenic , Nerve Tissue Proteins , Purkinje Cells/physiology , Purkinje Cells/ultrastructure , Rats, Transgenic , Receptors, Growth Factor
7.
F1000Res ; 82019.
Article in English | MEDLINE | ID: mdl-31583078

ABSTRACT

Complex mechanisms control the signaling of neurotrophins through p75 NTR and Trk receptors, allowing cellular responses that are highly context dependent, particularly in the nervous system and particularly with regard to the neurotrophin brain-derived neurotrophic factor (BDNF). Recent reports describe a variety of sophisticated regulatory mechanisms that contribute to such functional flexibility. Mechanisms described include regulation of trafficking of alternative BDNF transcripts, regulation of post-translational processing and secretion of BDNF, engagement of co-receptors that influence localization and signaling of p75 NTR and Trk receptors, and control of trafficking of receptors in the endocytic pathway and during anterograde and retrograde axonal transport.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Signal Transduction , Axonal Transport , Endocytosis , Humans , Nerve Tissue Proteins/pharmacology , Protein Processing, Post-Translational , Receptors, Nerve Growth Factor/physiology
8.
Acta Neuropathol Commun ; 7(1): 147, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31514753

ABSTRACT

The coordinated movement of organisms relies on efficient nerve-muscle communication at the neuromuscular junction. After peripheral nerve injury or neurodegeneration, motor neurons and Schwann cells increase the expression of the p75NTR pan-neurotrophin receptor. Even though p75NTR targeting has emerged as a promising therapeutic strategy to delay peripheral neuronal damage progression, the effects of long-term p75NTR inhibition at the mature neuromuscular junction have not been elucidated. We performed quantitative neuroanathomical analyses of the neuromuscular junction in p75NTR null mice by laser confocal and electron microscopy, which were complemented with electromyography, locomotor tests, and pharmacological intervention studies. Mature neuromuscular synapses of p75NTR null mice show impaired postsynaptic organization and ultrastructural complexity, which correlate with altered synaptic function at the levels of nerve activity-induced muscle responses, muscle fiber structure, force production, and locomotor performance. Our results on primary myotubes and denervated muscles indicate that muscle-derived p75NTR does not play a major role on postsynaptic organization. In turn, motor axon terminals of p75NTR null mice display a strong reduction in the number of synaptic vesicles and active zones. According to the observed pre and postsynaptic defects, pharmacological acetylcholinesterase inhibition rescued nerve-dependent muscle response and force production in p75NTR null mice. Our findings revealing that p75NTR is required to organize mature neuromuscular junctions contribute to a comprehensive view of the possible effects caused by therapeutic attempts to target p75NTR.


Subject(s)
Motor Neurons/physiology , Neuromuscular Junction/physiology , Receptors, Nerve Growth Factor/physiology , Synaptic Vesicles/physiology , Animals , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Motor Neurons/ultrastructure , Neuromuscular Junction/ultrastructure , Receptors, Nerve Growth Factor/genetics , Synaptic Vesicles/ultrastructure
9.
Sci Rep ; 9(1): 7696, 2019 05 22.
Article in English | MEDLINE | ID: mdl-31118427

ABSTRACT

Dysregulation of signaling networks controlling self-renewal and migration of developmental cell lineages is closely linked to the proliferative and invasive properties of tumors. Identification of such signaling pathways and their critical regulators is vital for successful design of effective targeted therapies against neoplastic tissue growth. The neurotrophin receptor (CD271/NGFR/p75NTR) is a key regulator of the melanocytic cell lineage through its ability to mediate cell growth, survival, and differentiation. Using clinical melanoma samples, normal melanocytes and global gene expression profiling we have investigated the role of CD271 in rewiring signal transduction networks of melanoma cells during neoplastic transformation. Our analysis demonstrates that depending on the cell fate of tumor initiation vs normal development, elevated levels of CD271 can serve as a switch between proliferation/survival and differentiation/cell death. Two divergent arms of neurotrophin signaling hold the balance between positive regulators of tumor growth controlled by E2F, MYC, SREBP1 and AKT3 pathways on the one hand, and differentiation, senescence, and apoptosis controlled by TRAF6/IRAK-dependent activation of AP1 and TP53 mediated processes on the other hand. A molecular network map revealed in this study uncovers CD271 as a context-specific molecular switch between normal development and malignant transformation.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Melanocytes/metabolism , Melanoma/metabolism , Neoplasm Proteins/physiology , Nerve Tissue Proteins/physiology , Receptors, Nerve Growth Factor/physiology , Cell Survival , Cell Transformation, Neoplastic , DNA Repair , Disease Progression , Gene Regulatory Networks , Humans , Melanocytes/cytology , Real-Time Polymerase Chain Reaction , Signal Transduction , Transcriptome
10.
J Neurosci ; 39(23): 4489-4510, 2019 06 05.
Article in English | MEDLINE | ID: mdl-30936240

ABSTRACT

By virtue of their extensive axonal arborization and perisomatic synaptic targeting, cortical inhibitory parvalbumin (PV) cells strongly regulate principal cell output and plasticity and modulate experience-dependent refinement of cortical circuits during development. An interesting aspect of PV cell connectivity is its prolonged maturation time course, which is completed only by end of adolescence. The p75 neurotrophin receptor (p75NTR) regulates numerous cellular functions; however, its role on cortical circuit development and plasticity remains elusive, mainly because localizing p75NTR expression with cellular and temporal resolution has been challenging. By using RNAscope and a modified version of the proximity ligation assay, we found that p75NTR expression in PV cells decreases between the second and fourth postnatal week, at a time when PV cell synapse numbers increase dramatically. Conditional knockout of p75NTR in single PV neurons in vitro and in PV cell networks in vivo causes precocious formation of PV cell perisomatic innervation and perineural nets around PV cell somata, therefore suggesting that p75NTR expression modulates the timing of maturation of PV cell connectivity in the adolescent cortex. Remarkably, we found that PV cells still express p75NTR in adult mouse cortex of both sexes and that its activation is sufficient to destabilize PV cell connectivity and to restore cortical plasticity following monocular deprivation in vivo Together, our results show that p75NTR activation dynamically regulates PV cell connectivity, and represent a novel tool to foster brain plasticity in adults.SIGNIFICANCE STATEMENT In the cortex, inhibitory, GABA-releasing neurons control the output and plasticity of excitatory neurons. Within this diverse group, parvalbumin-expressing (PV) cells form the larger inhibitory system. PV cell connectivity develops slowly, reaching maturity only at the end of adolescence; however, the mechanisms controlling the timing of its maturation are not well understood. We discovered that the expression of the neurotrophin receptor p75NTR in PV cells inhibits the maturation of their connectivity in a cell-autonomous fashion, both in vitro and in vivo, and that p75NTR activation in adult PV cells promotes their remodeling and restores cortical plasticity. These results reveal a new p75NTR function in the regulation of the time course of PV cell maturation and in limiting cortical plasticity.


Subject(s)
Aging/physiology , Interneurons/physiology , Neuronal Plasticity/physiology , Receptors, Nerve Growth Factor/physiology , Sexual Maturation/physiology , Visual Cortex/growth & development , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Connectome , Evoked Potentials, Visual , Female , GABAergic Neurons/cytology , Gene Expression Regulation, Developmental , Interneurons/chemistry , Interneurons/ultrastructure , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Parvalbumins/analysis , Protein Precursors/pharmacology , Random Allocation , Receptors, Nerve Growth Factor/biosynthesis , Receptors, Nerve Growth Factor/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Synapses/physiology , Vision, Monocular/physiology , Visual Cortex/cytology , Visual Cortex/metabolism
11.
s.l; s.n; 2019. 10 p.
Non-conventional in English | CONASS, Sec. Est. Saúde SP, HANSEN, Hanseníase Leprosy, SESSP-ILSLPROD, Sec. Est. Saúde SP, SESSP-ILSLACERVO, Sec. Est. Saúde SP | ID: biblio-1147177

ABSTRACT

Schwann cells (SCs) critically maintain the plasticity of the peripheral nervous system. Peripheral nerve injuries and infections stimulate SCs in order to retrieve homeostasis in neural tissues. Previous studies indicate that Mycobacterium leprae (ML) regulates the expression of key factors related to SC identity, suggesting that alterations in cell phenotype may be involved in the pathogenesis of neural damage in leprosy. To better understand whether ML restricts the plasticity of peripheral nerves, the present study sought to determine the expression of Krox­20, Sox­10, c­Jun and p75NTR in SC culture and mice sciatic nerves, both infected by ML Thai­53 strain. Primary SC cultures were stimulated with two different multiplicities of infection (MOI 100:1; MOI 50:1) and assessed after 7 and 14 days. Sciatic nerves of nude mice (NU­Foxn1nu) infected with ML were evaluated after 6 and 9 months. In vitro results demonstrate downregulation of Krox­20 and Sox­10 along with the increase in p75NTR­immunolabelled cells. Concurrently, sciatic nerves of infected mice showed a significant decrease in Krox­20 and increase in p75NTR. Our results corroborate previous findings on the interference of ML in the expression of factors involved in cell maturation, favouring the maintenance of a non­myelinating phenotype in SCs, with possible implications for the repair of adult peripheral nerves(AU).


Subject(s)
Animals , Mice , Schwann Cells/microbiology , Leprosy/metabolism , Leprosy/microbiology , Mycobacterium leprae/isolation & purification , Peripheral Nerves/microbiology , Schwann Cells/metabolism , In Vitro Techniques , Down-Regulation , Receptors, Nerve Growth Factor/physiology , Early Growth Response Protein 2/biosynthesis , Neuronal Plasticity/physiology
12.
Arch Dermatol Res ; 310(9): 737-750, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30209580

ABSTRACT

CD271, a receptor of nerve growth factor (NGF), affects the biological properties of epidermal stem cells (eSCs) which are essential for skin wound closure. Tropomyosin-receptor kinase A (TrkA), another receptor of NGF, combined with CD271 has been involved with nervous system and skin keratinocytes. However, the exact role of TrkA combined with CD271 in eSCs during skin wound closure is still unclear. This study aimed to reveal the role of TrkA in the promoting wounding-healing effect of CD271 on eSCs. We obtained CD271-vo (over-expression of CD271) eSCs by lentiviral infection. K252a was used to inhibit TrkA expression. Full-thickness skin mouse wound closure model (5 mm in diameter) was used to detect the ability of CD271 over-expressed/TrkA-deficient during wound healing. The biological characteristics of eSCs and their proliferation and apoptosis were detected using immunohistochemistry and western blot. The expressions of protein kinase B (pAkt)/Akt, phosphorylated extracellular-signal-related kinase (pERK)/ERK1/2, and c-Jun N-terminal kinase (pJNK)/JNK were also detected by western blot. We found that over-expression of CD271 promoted the biological functions of eSCs. Interestingly, over-expression of CD271 in the absence of TrkA neither promoted eSCs' migration and proliferation nor promoted wound healing in a mouse model. In addition, we observed the reduced expression of pAkt/Akt and pERK/ERK1/2 following TrkA inhibition in vitro. Our studies demonstrated that the role of TrkA in the promoting wounding-healing effect of CD271 on eSCs.


Subject(s)
Epidermis/metabolism , Nerve Tissue Proteins/physiology , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/physiology , Stem Cells/metabolism , Wound Healing , Wounds, Penetrating/metabolism , Animals , Carbazoles/pharmacology , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Epidermis/drug effects , Epidermis/injuries , Epidermis/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Indole Alkaloids/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/pharmacology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, trkA/antagonists & inhibitors , Signal Transduction , Stem Cell Transplantation , Stem Cells/drug effects , Stem Cells/pathology , Wound Healing/drug effects , Wounds, Penetrating/genetics , Wounds, Penetrating/pathology
13.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 53(2): 133-136, 2018 Feb 09.
Article in Chinese | MEDLINE | ID: mdl-29429237

ABSTRACT

The p75 neurotrophic factor receptor is a low affinity receptor for neurotrophic factors and plays an important role in nerve growth, development and function integrity. It is closely related to dental development, oral and maxillofacial tumor, nerve repair and tissue engineering. It shows good prospect for application. In this paper, the research progress of p75 neurotrophic factor receptor in Stomatology is reviewed.


Subject(s)
Nerve Tissue Proteins/physiology , Receptors, Nerve Growth Factor/physiology , Humans , Nerve Regeneration , Neurons , Oral Medicine , Tissue Engineering
14.
Article in Japanese | MEDLINE | ID: mdl-28539548

ABSTRACT

CD271 is common stem cell marker for the epidermis and dermis. We assessed a kinetic movement of epidermal and dermal CD271+ cells in the wound healing process to elucidate the possible involvement with chronic skin ulcers. Epidermal CD271+ cells were proliferated and migrated from 3 days after wounding. Purified epidermal CD271+ cells expressed higher TGFß2 and VEGFα transcripts than CD271- cells. Delayed wound healing was observed in the aged mice compared with young mice. During the wound healing process, the peak of dermal CD271+ cell accumulation was delayed in aged mice compared with young mice. The expression levels of collagen-1, -3, -5, F4-80, EGF, FGF2, TGFß1, and IL-1α were significantly increased in young mice compared with aged mice. Furthermore, purified dermal CD271+ cells expressed higher FGF2, EGF, PDGFB, and TGFß1 gene transcripts than CD271- cells. These results suggested that epidermal and dermal CD271+ cells were closely associated with wound healing process by producing various growth factors. Epidermal and dermal CD271+ cells in chronic skin ulcer patients were significantly reduced compared with healthy controls. Thus, both epidermal and dermal stem cells can play an important role in wound healing process.


Subject(s)
Receptors, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/physiology , Skin Physiological Phenomena , Skin Ulcer/pathology , Skin Ulcer/physiopathology , Skin/cytology , Stem Cells/cytology , Stem Cells/physiology , Wound Healing/physiology , Aging , Animals , Cell Movement , Cell Proliferation , Chronic Disease , Disease Models, Animal , Humans , Mice , Skin/pathology , Stem Cells/metabolism , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism
15.
J Pathol ; 242(3): 347-357, 2017 07.
Article in English | MEDLINE | ID: mdl-28418072

ABSTRACT

Acute basophilic leukaemia (ABL) is a rare subtype of acute myeloblastic leukaemia. We previously described a recurrent t(X;6)(p11;q23) translocation generating an MYB-GATA1 fusion gene in male infants with ABL. To better understand its role, the chimeric MYB-GATA1 transcription factor was expressed in CD34-positive haematopoietic progenitors, which were transplanted into immunodeficient mice. Cells expressing MYB-GATA1 showed increased expression of markers of immaturity (CD34), of granulocytic lineage (CD33 and CD117), and of basophilic differentiation (CD203c and FcϵRI). UT-7 cells also showed basophilic differentiation after MYB-GATA1 transfection. A transcriptomic study identified nine genes deregulated by both MYB-GATA1 and basophilic differentiation. Induction of three of these genes (CCL23, IL1RL1, and NTRK1) was confirmed in MYB-GATA1-expressing CD34-positive cells by reverse transcription quantitative polymerase chain reaction. Interleukin (IL)-33 and nerve growth factor (NGF), the ligands of IL-1 receptor-like 1 (IL1RL1) and neurotrophic receptor tyrosine kinase 1 (NTRK1), respectively, enhanced the basophilic differentiation of MYB-GATA1-expressing UT-7 cells, thus demonstrating the importance of this pathway in the basophilic differentiation of leukaemic cells and CD34-positive primary cells. Finally, gene reporter assays confirmed that MYB and MYB-GATA1 directly activated NTRK1 and IL1RL1 transcription, leading to basophilic skewing of the blasts. MYB-GATA1 is more efficient than MYB, because of better stability. Our results highlight the role of IL-33 and NGF receptors in the basophilic differentiation of normal and leukaemic cells. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Interleukin-33/physiology , Leukemia, Basophilic, Acute/etiology , Receptors, Nerve Growth Factor/physiology , Animals , Cell Transformation, Neoplastic/genetics , Female , GATA1 Transcription Factor/genetics , Gene Fusion/physiology , Hematopoietic Stem Cells/physiology , Male , Mice, SCID , Neoplasm Transplantation , Oncogene Proteins v-myb/genetics , Receptor, trkA/metabolism , Transcription Factors/metabolism , Transfection , Transplantation, Heterologous
16.
Dev Neurobiol ; 77(4): 405-418, 2017 04.
Article in English | MEDLINE | ID: mdl-27503831

ABSTRACT

Neurons are the largest cells in the body and form subcellular compartments such as axons and dendrites. During both development and adulthood building blocks must be continually trafficked long distances to maintain the different regions of the neuron. Beyond building blocks, signaling complexes are also transported, allowing for example, axons to communicate with the soma. The critical roles of signaling via ligand-receptor complexes is perhaps best illustrated in the context of development, where they are known to regulate polarization, survival, axon outgrowth, dendrite development, and synapse formation. However, knowing 'when' and 'how much' signaling is occurring does not provide the complete story. The location of signaling has a significant impact on the functional outcomes. There are therefore complex and functionally important trafficking mechanisms in place to control the precise spatial and temporal aspects of many signal transduction events. In turn, many of these signaling events affect trafficking mechanisms, setting up an intricate connection between trafficking and signaling. In this review we will use neurotrophin receptors, specifically TrkA and TrkB, to illustrate the cell biology underlying the links between trafficking and signaling. Briefly, we will discuss the concepts of how trafficking and signaling are intimately linked for functional and diverse signaling outputs, and how the same protein can play different roles for the same receptor depending on its localization. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 419-437, 2017.


Subject(s)
Endosomes/physiology , Neurons/physiology , Protein Transport/physiology , Receptors, Nerve Growth Factor/physiology , Signal Transduction/physiology , Animals , Humans
17.
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
18.
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
19.
Mol Hum Reprod ; 22(1): 3-17, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26487421

ABSTRACT

The neurotrophin family consists of nerve growth factor (NGF), neurotrophin 3 (NT3) and neurotrophin 4/5 (NT4/5), in addition to brain-derived neurotrophic factor (BDNF) and the neuronal growth factors, glial cell line-derived neurotrophic factor (GDNF) and vasointestinal peptide (VIP). Although there are a few literature reviews, mainly of animal studies, on the importance of neurotrophins in the ovary, we aimed to provide a complete review of neurotrophins as well as neuronal growth factors and their important roles in normal and pathological processes in the ovary. Follicular assembly is probably stimulated by complementary effects of NGF, NT4/5 and BDNF and their receptors. The neurotrophins, GDNF and VIP and their receptors have all been identified in preantral and antral follicles of mammalian species, including humans. Transgenic mice with mutations in the genes encoding for Ngf, Nt4/5 and Bdnf and their tropomyosin-related kinase ß receptor showed a reduction in preantral follicles and an abnormal ovarian morphology, whereas NGF, NT3, GDNF and VIP increased the in vitro activation of primordial follicles in rats and goats. Additionally, NGF, NT3 and GDNF promoted follicular cell proliferation; NGF, BDNF and VIP were shown to be involved in ovulation; VIP inhibited follicular apoptosis; NT4/5, BDNF and GDNF promoted oocyte maturation and NGF, NT3 and VIP stimulated steroidogenesis. NGF may also exert a stimulatory effect in ovarian cancer and polycystic ovarian syndrome (PCOS). Low levels of NGF and BDNF in follicular fluid may be associated with diminished ovarian reserve and high levels with endometriosis. More knowledge of the roles of neuronal growth factors in the ovary has important implications for the development of new therapeutic drugs (such as anti-NGF agents) for ovarian cancer and PCOS as well as various infertility problems, warranting further research.


Subject(s)
Nerve Growth Factors/physiology , Ovary/physiology , Animals , Apoptosis , Endometriosis/physiopathology , Female , Humans , Infertility, Female/physiopathology , Mice , Mice, Transgenic , Oocytes/physiology , Ovarian Follicle/physiology , Ovarian Neoplasms/physiopathology , Ovulation/physiology , Rats , Receptors, Nerve Growth Factor/physiology , Signal Transduction
20.
Neurobiol Aging ; 36(2): 740-52, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25443284

ABSTRACT

Alzheimer's disease (AD) is an extremely prevalent cause of dementia. It is characterized by progressive memory loss, confusion, and other behavioral and physiological problems. The amyloid-ß (Aß) protein is thought to be involved in the pathogenesis of AD, and there is evidence that Aß may act through the p75 neurotrophin receptor (p75) to mediate its pathogenic effects. This raises the possibility that reducing levels of p75 could be a treatment for AD by preventing the effects of Aß. In this study, we have crossed the transgenic AD model mice, Tg2576, with p75(-/-) mice to generate Tg2576/p75(+/-) mice with reduced levels of p75. These mice are rescued from the deficits in learning and memory and hippocampal function which were found in the Tg2576 mice. These findings suggest that reduction of p75 can ameliorate some of the primary symptoms of AD.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Receptors, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Animals , Disease Models, Animal , Female , Hippocampus/physiopathology , Humans , Learning , Male , Memory , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...