Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Elife ; 102021 06 29.
Article in English | MEDLINE | ID: mdl-34184986

ABSTRACT

The congenital intellectual disability (ID)-causing gene mutations remain largely unclear, although many genetic variations might relate to ID. We screened gene mutations in Chinese Han children suffering from severe ID and found a single-nucleotide polymorphism (SNP) in the 5'-untranslated region (5'-UTR) of fibroblast growth factor 13 (FGF13) mRNA (NM_001139500.1:c.-32c>G) shared by three male children. In both HEK293 cells and patient-derived induced pluripotent stem cells, this SNP reduced the translation of FGF13, which stabilizes microtubules in developing neurons. Mice carrying the homologous point mutation in 5'-UTR of Fgf13 showed delayed neuronal migration during cortical development, and weakened learning and memory. Furthermore, this SNP reduced the interaction between FGF13 5'-UTR and polypyrimidine-tract-binding protein 2 (PTBP2), which was required for FGF13 translation in cortical neurons. Thus, this 5'-UTR SNP of FGF13 interferes with the translational process of FGF13 and causes deficits in brain development and cognitive functions.


Subject(s)
5' Untranslated Regions/genetics , Fibroblast Growth Factors/genetics , Intellectual Disability/genetics , Point Mutation , Polymorphism, Single Nucleotide , Adolescent , Animals , Child , Child, Preschool , Fibroblast Growth Factors/metabolism , HEK293 Cells , Humans , Learning , Male , Memory , Mice , Mice, Inbred C57BL
2.
Mol Pain ; 14: 1744806918769492, 2018.
Article in English | MEDLINE | ID: mdl-29587571

ABSTRACT

Opioid receptors play an important role in mediating the spinal analgesia. The µ-opioid receptor is the major target of opioid drugs widely used in clinics. However, the regulatory mechanisms of analgesic effect and tolerance for clinical µ-opioid receptor-targeting opioids remain to be fully investigated. Previous studies showed the interaction of δ-opioid receptor with µ-opioid receptor to form the µ-opioid receptor/δ-opioid receptor heteromers that could be processed in the degradation pathway after δ-opioid receptor agonist treatment. Here, we showed that clinical µ-opioid receptor-targeting opioids, morphine, fentanyl, and methadone, but not tramadol, caused µ-opioid receptor co-internalization with δ-opioid receptors in both transfected human embryonic kidney 293 cells and primary sensory neurons. Prolonged treatment of morphine led to µ-opioid receptor co-degradation with δ-opioid receptors. Furthermore, fentanyl and methadone, but not tramadol, induced the drug tolerance similar to morphine. Thus, the clinical µ-opioid receptor-targeting opioids including morphine, fentanyl, and methadone induce µ-opioid receptor co-internalization with δ-opioid receptors, which may be involved in the analgesic tolerance of these opioids.


Subject(s)
Analgesics, Opioid/pharmacology , Endocytosis , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Analgesics/pharmacology , Animals , Cells, Cultured , Drug Tolerance , HEK293 Cells , Humans , Mice , Morphine/pharmacology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism
3.
Neuron ; 93(4): 806-821.e9, 2017 Feb 22.
Article in English | MEDLINE | ID: mdl-28162808

ABSTRACT

The current knowledge about heat nociception is mainly confined to the thermosensors, including the transient receptor potential cation channel V1 expressed in the nociceptive neurons of dorsal root ganglion (DRG). However, the loss of thermosensors only partially impairs heat nociception, suggesting the existence of undiscovered mechanisms. We found that the loss of an intracellular fibroblast growth factor (FGF), FGF13, in the mouse DRG neurons selectively abolished heat nociception. The noxious heat stimuli could not evoke the sustained action potential firing in FGF13-deficient DRG neurons. Furthermore, FGF13 interacted with the sodium channel Nav1.7 in a heat-facilitated manner. FGF13 increased Nav1.7 sodium currents and maintained the membrane localization of Nav1.7 during noxious heat stimulation, enabling the sustained firing of action potentials. Disrupting the FGF13/Nav1.7 interaction reduced the heat-evoked action potential firing and nociceptive behavior. Thus, beyond the thermosensors, the FGF13/Nav1.7 complex is essential for sustaining the transmission of noxious heat signals.


Subject(s)
Fibroblast Growth Factors/metabolism , Ganglia, Spinal/metabolism , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Neurons/physiology , Action Potentials/physiology , Animals , Cells, Cultured , Fibroblast Growth Factors/genetics , Hot Temperature , Humans , Mice, Transgenic
5.
Sheng Li Xue Bao ; 68(2): 179-84, 2016 Apr 25.
Article in Chinese | MEDLINE | ID: mdl-27108905

ABSTRACT

The aim of the present study was to develop three-dimensional (3D) culture model, a more pathologically relevant model, of human breast cancer for drug resistance study. MCF-7 cells were embedded within collagen gel to establish 3D culture model. Cellular morphology was observed using Carmine and HE staining. Cell proliferation was evaluated by CCK-8 assay, and cell activity was detected by Live/Dead staining kit. Drug sensitivities of the 3D culture to doxorubicin, carboplatin, 5-fluorouracil were assayed and compared with those of monolayer (2D) culture. In addition, the levels of drug resistance-related genes P-glycoprotein (P-gp), mrp2 mRNA expressions were detected by real time RT-PCR. Expression level of P-gp protein was detected by Western blot. The results showed that MCF-7 cells in 3D culture formed a number of cell aggregates, and most of them displayed good cell viability. The IC50 values of doxorubicin, carboplatin, 5-fluorouracil were all increased significantly in 3D culture compared with those in 2D culture. Moreover, compared with MCF-7 cells in 2D culture, the cells in 3D culture showed increased mRNA levels of P-gp and mrp2, as well as up-regulated protein expression of P-gp. These results suggest that in vitro collagen-embedded culture system of human breast cancer cells represents an improved pathologically relevant 3D microenvironment for breast cancer cells, providing a robust tool to explore the mechanism of drug resistance of cancer cells.


Subject(s)
Breast Neoplasms , Cell Culture Techniques , Drug Resistance, Neoplasm , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Cell Proliferation , Cell Survival , Doxorubicin , Humans , MCF-7 Cells
6.
Cell Res ; 26(1): 83-102, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26691752

ABSTRACT

Sensory neurons are distinguished by distinct signaling networks and receptive characteristics. Thus, sensory neuron types can be defined by linking transcriptome-based neuron typing with the sensory phenotypes. Here we classify somatosensory neurons of the mouse dorsal root ganglion (DRG) by high-coverage single-cell RNA-sequencing (10 950 ± 1 218 genes per neuron) and neuron size-based hierarchical clustering. Moreover, single DRG neurons responding to cutaneous stimuli are recorded using an in vivo whole-cell patch clamp technique and classified by neuron-type genetic markers. Small diameter DRG neurons are classified into one type of low-threshold mechanoreceptor and five types of mechanoheat nociceptors (MHNs). Each of the MHN types is further categorized into two subtypes. Large DRG neurons are categorized into four types, including neurexophilin 1-expressing MHNs and mechanical nociceptors (MNs) expressing BAI1-associated protein 2-like 1 (Baiap2l1). Mechanoreceptors expressing trafficking protein particle complex 3-like and Baiap2l1-marked MNs are subdivided into two subtypes each. These results provide a new system for cataloging somatosensory neurons and their transcriptome databases.


Subject(s)
Ganglia, Spinal/cytology , Gene Regulatory Networks , Sensory Receptor Cells/cytology , Transcriptome , Animals , Base Sequence , Cells, Cultured , Ganglia, Spinal/metabolism , Male , Mechanoreceptors/cytology , Mechanoreceptors/metabolism , Mice , Mice, Inbred C57BL , Multigene Family , Nociceptors/cytology , Nociceptors/metabolism , Patch-Clamp Techniques , Sensory Receptor Cells/metabolism , Sequence Analysis, RNA
7.
Cell Res ; 25(3): 318-34, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25633594

ABSTRACT

Na⁺, K⁺-ATPase (NKA) is required to generate the resting membrane potential in neurons. Nociceptive afferent neurons express not only the α and ß subunits of NKA but also the γ subunit FXYD2. However, the neural function of FXYD2 is unknown. The present study shows that FXYD2 in nociceptive neurons is necessary for maintaining the mechanical allodynia induced by peripheral inflammation. FXYD2 interacted with α1NKA and negatively regulated the NKA activity, depolarizing the membrane potential of nociceptive neurons. Mechanical allodynia initiated in FXYD2-deficient mice was abolished 4 days after inflammation, whereas it persisted for at least 3 weeks in wild-type mice. Importantly, the FXYD2/α1NKA interaction gradually increased after inflammation and peaked on day 4 post inflammation, resulting in reduction of NKA activity, depolarization of neuron membrane and facilitation of excitatory afferent neurotransmission. Thus, the increased FXYD2 activity may be a fundamental mechanism underlying the persistent hypersensitivity to pain induced by inflammation.


Subject(s)
Hyperalgesia/physiopathology , Inflammation/physiopathology , Nociceptors/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Male , Membrane Potentials/physiology , Mice , Mice, Knockout , Nociceptors/metabolism , Pain/physiopathology , RNA, Messenger/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Synaptic Transmission/physiology
8.
Brain ; 135(Pt 2): 391-403, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22275428

ABSTRACT

Emerging evidence suggests that the suppressive modulators released from nociceptive afferent neurons contribute to pain regulation. However, the suppressive modulators expressed in small-diameter neurons of the dorsal root ganglion remain to be further identified. The present study shows that the activin C expressed in small dorsal root ganglion neurons is required for suppressing inflammation-induced nociceptive responses. The expression of activin C in small dorsal root ganglion neurons of rats was markedly downregulated during the early days of peripheral inflammation induced by intraplantar injection of the complete Freund's adjuvant. Intrathecal treatment with the small interfering RNA targeting activin ßC or the antibodies against activin C could enhance the formalin-induced nociceptive responses, and impair the recovery from the complete Freund's adjuvant-induced thermal hyperalgesia. Intrathecally applied activin C could reduce nociceptive responses induced by formalin or complete Freund's adjuvant. Moreover, activin C was found to inhibit the inflammation-induced phosphorylation of extracellular signal-regulated kinase in the dorsal root ganglia and the dorsal spinal cord. Thus, activin C functions as an endogenous suppressor of inflammatory nociceptive transmission and may have a therapeutic potential for treatment of inflammatory pain.


Subject(s)
Activins/metabolism , Ganglia, Spinal/metabolism , Hyperalgesia/metabolism , Inflammation/metabolism , Inhibin-beta Subunits/metabolism , Nociceptors/metabolism , Animals , Behavior, Animal , Cell Count , Chronic Pain/chemically induced , Chronic Pain/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Hyperalgesia/chemically induced , Inflammation/chemically induced , Phosphorylation , Rats , Rats, Sprague-Dawley
9.
Neuron ; 69(5): 974-87, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21382556

ABSTRACT

Excitatory synaptic transmission is modulated by inhibitory neurotransmitters and neuromodulators. We found that the synaptic transmission of somatic sensory afferents can be rapidly regulated by a presynaptically secreted protein, follistatin-like 1 (FSTL1), which serves as a direct activator of Na(+),K(+)-ATPase (NKA). The FSTL1 protein is highly expressed in small-diameter neurons of the dorsal root ganglion (DRG). It is transported to axon terminals via small translucent vesicles and secreted in both spontaneous and depolarization-induced manners. Biochemical assays showed that FSTL1 binds to the α1 subunit of NKA and elevates NKA activity. Extracellular FSTL1 induced membrane hyperpolarization in cultured cells and inhibited afferent synaptic transmission in spinal cord slices by activating NKA. Genetic deletion of FSTL1 in small DRG neurons of mice resulted in enhanced afferent synaptic transmission and sensory hypersensitivity, which could be reduced by intrathecally applied FSTL1 protein. Thus, FSTL1-dependent activation of NKA regulates the threshold of somatic sensation.


Subject(s)
Follistatin-Related Proteins/metabolism , Sensory Receptor Cells/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Synaptic Transmission/physiology , Analysis of Variance , Animals , Blotting, Northern , Blotting, Western , COS Cells , Cells, Cultured , Chlorocebus aethiops , Follistatin-Related Proteins/genetics , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Immunohistochemistry , Mice , Mice, Knockout , Patch-Clamp Techniques , Presynaptic Terminals/metabolism , Rats
11.
Cell Res ; 21(5): 741-53, 2011 May.
Article in English | MEDLINE | ID: mdl-21321602

ABSTRACT

Stimulus-induced exocytosis of large dense-core vesicles (LDCVs) leads to discharge of neuropeptides and fusion of LDCV membranes with the plasma membrane. However, the contribution of LDCVs to the properties of the neuronal membrane remains largely unclear. The present study found that LDCVs were associated with multiple receptors, channels and signaling molecules, suggesting that neuronal sensitivity is modulated by an LDCV-mediated mechanism. Liquid chromatography-mass spectrometry combined with immunoblotting of subcellular fractions identified 298 proteins in LDCV membranes purified from the dorsal spinal cord, including G-protein-coupled receptors, G-proteins and other signaling molecules, ion channels and trafficking-related proteins. Morphological assays showed that δ-opioid receptor 1 (DOR1), ß2 adrenergic receptor (AR), G(αi2), voltage-gated calcium channel α2δ1 subunit and P2X purinoceptor 2 were localized in substance P (SP)-positive LDCVs in small-diameter dorsal root ganglion neurons, whereas ß1 AR, Wnt receptor frizzled 8 and dishevelled 1 were present in SP-negative LDCVs. Furthermore, DOR1/G(αi2)/G(ß1γ5)/phospholipase C ß2 complexes were associated with LDCVs. Blockade of the DOR1/G(αi2) interaction largely abolished the LDCV localization of G(αi2) and impaired stimulation-induced surface expression of G(αi2). Thus, LDCVs serve as carriers of receptors, ion channels and preassembled receptor signaling complexes, enabling a rapid, activity-dependent modulation of neuronal sensitivity.


Subject(s)
Ion Channels/metabolism , Neuropeptides/metabolism , Receptors, G-Protein-Coupled/metabolism , Secretory Vesicles/metabolism , Signal Transduction , Animals , Biological Transport , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein beta Subunits/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/ultrastructure , Mice , PC12 Cells , Phospholipase C beta/metabolism , Rats
12.
Neuron ; 69(1): 120-31, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21220103

ABSTRACT

δ-opioid receptors (DORs) form heteromers with µ-opioid receptors (MORs) and negatively regulate MOR-mediated spinal analgesia. However, the underlying mechanism remains largely unclear. The present study shows that the activity of MORs can be enhanced by preventing MORs from DOR-mediated codegradation. Treatment with DOR-specific agonists led to endocytosis of both DORs and MORs. These receptors were further processed for ubiquitination and lysosomal degradation, resulting in a reduction of surface MORs. Such effects were attenuated by treatment with an interfering peptide containing the first transmembrane domain of MOR (MOR(TM1)), which interacted with DORs and disrupted the MOR/DOR interaction. Furthermore, the systemically applied fusion protein consisting of MOR(TM1) and TAT at the C terminus could disrupt the MOR/DOR interaction in the mouse spinal cord, enhance the morphine analgesia, and reduce the antinociceptive tolerance to morphine. Thus, dissociation of MORs from DORs in the cell membrane is a potential strategy to improve opioid analgesic therapies.


Subject(s)
Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Signal Transduction/drug effects , Spinal Cord/metabolism , Analgesia/methods , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Animals , Disease Models, Animal , Endocytosis , HEK293 Cells , Humans , Immunoblotting , In Situ Hybridization , Lysosomes/metabolism , Male , Mice , Mice, Inbred Strains , Microscopy, Electron , Morphine/administration & dosage , Morphine/pharmacology , Pain/drug therapy , Pain Measurement/methods , Peptides/pharmacology , Plasmids , Spinal Cord/drug effects , Transfection , Ubiquitination
13.
J Neurosci ; 30(32): 10927-38, 2010 Aug 11.
Article in English | MEDLINE | ID: mdl-20702721

ABSTRACT

B-type natriuretic peptide (BNP) has been known to be secreted from cardiac myocytes and activate its receptor, natriuretic peptide receptor-A (NPR-A), to reduce ventricular fibrosis. However, the function of BNP/NPR-A pathway in the somatic sensory system has been unknown. In the present study, we report a novel function of BNP in pain modulation. Using microarray and immunoblot analyses, we found that BNP and NPR-A were expressed in the dorsal root ganglion (DRG) of rats and upregulated after intraplantar injection of complete Freund's adjuvant (CFA). Immunohistochemistry showed that BNP was expressed in calcitonin gene-related peptide (CGRP)-containing small neurons and IB4 (isolectin B4)-positive neurons, whereas NPR-A was present in CGRP-containing neurons. Application of BNP reduced the firing frequency of small DRG neurons in the presence of glutamate through opening large-conductance Ca2+-activated K+ channels (BKCa channels). Furthermore, intrathecal injection of BNP yielded inhibitory effects on formalin-induced flinching behavior and CFA-induced thermal hyperalgesia in rats. Blockade of BNP signaling by BNP antibodies or cGMP-dependent protein kinase (PKG) inhibitor KT5823 [(9S,10R,12R)-2,3,9,10,11,12-hexahydro-10-methoxy-2,9-dimethyl-1-oxo-9,12-epoxy-1H-diindolo[1,2,3-fg:3',2',1'-kl]pyrrolo[3,4-i][1,6]benzodiazocine-10-carboxylic acid methyl ester] impaired the recovery from CFA-induced thermal hyperalgesia. Thus, BNP negatively regulates nociceptive transmission through presynaptic receptor NPR-A, and activation of the BNP/NPR-A/PKG/BKCa channel pathway in nociceptive afferent neurons could be a potential strategy for inflammatory pain therapy.


Subject(s)
Gene Expression Regulation/physiology , Natriuretic Peptide, Brain/metabolism , Pain/metabolism , Sensory Receptor Cells/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Antibodies/pharmacology , Antibodies/therapeutic use , Biophysical Phenomena/drug effects , Biophysical Phenomena/physiology , Calcitonin Gene-Related Peptide/metabolism , Carbazoles/pharmacology , Carbazoles/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Double-Blind Method , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Excitatory Postsynaptic Potentials/drug effects , Freund's Adjuvant , Ganglia, Spinal/pathology , Gene Expression Regulation/drug effects , Glutamic Acid/pharmacology , Hyperalgesia/complications , Hyperalgesia/drug therapy , Inflammation/chemically induced , Inflammation/complications , Lectins/metabolism , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Natriuretic Peptide, Brain/immunology , Pain/drug therapy , Pain/etiology , Pain Measurement/methods , Patch-Clamp Techniques/methods , Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Atrial Natriuretic Factor/metabolism , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects , Time Factors
14.
Eur J Neurosci ; 25(4): 1087-96, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17331205

ABSTRACT

Peripheral nerve injury-induced structural and chemical modifications of the sensory circuits in the dorsal horn of the spinal cord contribute to the mechanism of neuropathic pain. In contrast to the topographic projection of primary afferents in laminae I-IV in the rat spinal cord, the primary afferents of Macaca mulatta monkeys almost exclusively project into laminae I-II of the spinal cord. After peripheral nerve injury, up-regulation of galanin has been found in sensory neurons in both monkey and rat dorsal root ganglia. However, the nerve injury-induced ultrastructural modification of galanin-containing afferents in the monkey spinal cord remains unknown. Using immunoelectron microscopy, we found that 3 weeks after unilateral sciatic nerve transection, the number of galanin-containing afferents was increased in ipsilateral lamina II of monkey spinal cord. Branching of these galanin-positive afferents was often observed. The afferent terminals contained a large number of synaptic vesicles, peptidergic vesicles and mitochondria, whereas the number of synapses was markedly reduced. Some of the afferents-enriched microtubules were often packed into bundles. Moreover, galanin-labeling could be associated with endosomal structures in many dendrites and axonal terminals of dorsal horn neurons. These results suggest that peripheral nerve injury induces an expansion of the central projection of galanin-containing afferents in lamina II of the monkey spinal cord, not only by increasing galanin levels in primary afferents but also by triggering afferent branching.


Subject(s)
Galanin/metabolism , Posterior Horn Cells/metabolism , Posterior Horn Cells/physiopathology , Sciatic Neuropathy/physiopathology , Spinal Cord/pathology , Animals , Functional Laterality , Immunohistochemistry , Macaca mulatta , Male , Microscopy, Immunoelectron/methods , Posterior Horn Cells/ultrastructure , Sciatic Neuropathy/pathology , Spinal Cord/ultrastructure
15.
J Neurosci ; 23(11): 4775-84, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12805317

ABSTRACT

Opiate abuse causes adaptive changes in several processes of synaptic transmission in which the glutamatergic system appears a critical element involved in opiate tolerance and dependence, but the underlying mechanisms remain unclear. In the present study, we found that glutamate uptake in hippocampal synaptosomes was significantly increased (by 70% in chronic morphine-treated rats) during the morphine withdrawal period, likely attributable to an increase in the number of functional glutamate transporters. Immunoblot analysis showed that expression of GLT1 (glutamate transporter subtype 1) was identified to be upregulated in synaptosomes but not in total tissues, suggesting a redistribution of glutamate transporter expression. Moreover, the increase in glutamate uptake was reproduced in cultured neurons during morphine withdrawal, and the increase of uptake in neurons could be blocked by dihydrokainate, a specific inhibitor of GLT1. Cell surface biotinylation and immunoblot analysis showed that morphine withdrawal produced an increase in GLT1 expression rather than EAAC1 (excitatory amino acids carrier 1), a neuronal subtype, at the cultured neuronal cell surface, whereas no significant change was observed in that of cultured astrocytes. Electron microscopy also revealed that GLT1 expression was markedly increased in the nerve terminals of hippocampus and associated with the plasma membrane in vivo. These results suggest that GLT1 in hippocampal neurons can be induced to translocate to the nerve terminals and express on the cell surface during morphine withdrawal. The translocation of GLT1 at synapses during morphine withdrawal provides a neuronal mechanism for modulation of excitatory neurotransmission during opiate abuse.


Subject(s)
Excitatory Amino Acid Transporter 2/metabolism , Glutamic Acid/pharmacokinetics , Hippocampus/metabolism , Morphine/adverse effects , Substance Withdrawal Syndrome/metabolism , Synapses/metabolism , Amino Acid Transport System X-AG/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 3 , Glutamate Plasma Membrane Transport Proteins , Hippocampus/cytology , Male , Morphine Dependence/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Presynaptic Terminals/metabolism , Protein Transport/drug effects , Protein Transport/physiology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Symporters/metabolism , Synaptosomes/chemistry , Synaptosomes/metabolism , Up-Regulation/drug effects
16.
Neuroreport ; 13(15): 1903-7, 2002 Oct 28.
Article in English | MEDLINE | ID: mdl-12395088

ABSTRACT

Using cDNA array, we observed the expression of eight members of the fibroblast growth factor (FGF) family, FGF 2, 5, 7, 9, 10, 13 and 14, in rat lumbar 4 and 5 dorsal root ganglia (DRGs). Over a period of 28 days after sciatic nerve transection, the array signals for FGF 2 and 7 were significantly increased in the DRGs, while FGF 13 decreased. Using the reverse transcription polymerase chain reaction (RT-PCR), we confirmed the axotomy-induced changes in the expression of FGF 7 and 13. hybridization showed that FGF 13 was expressed in 60% of DRG neurons under normal circumstance. Seven days after axotomy the number of FGF 13-positive neurons was decreased to 18%, but partially recovered to 40% after 28 days. FGF 13 immunoreactivity was also decreased. These data indicate that FGFs are important for DRG neurons under normal circumstance and after nerve injury.


Subject(s)
Fibroblast Growth Factors/genetics , Ganglia, Spinal/metabolism , Neuralgia/metabolism , Neurons, Afferent/metabolism , Peripheral Nerve Injuries , Peripheral Nervous System Diseases/metabolism , Up-Regulation/genetics , Animals , Disease Models, Animal , Down-Regulation/genetics , Fibroblast Growth Factors/metabolism , Ganglia, Spinal/physiopathology , Gene Expression/physiology , Immunohistochemistry , Male , Neuralgia/physiopathology , Peripheral Nervous System Diseases/physiopathology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reaction Time/physiology , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Sciatic Nerve/surgery
17.
Eur J Neurosci ; 16(2): 175-85, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12169100

ABSTRACT

Peripheral axotomy-induced sprouting of thick myelinated afferents (A-fibers) from laminae III-IV into laminae I-II of the spinal cord is a well-established hypothesis for the structural basis of neuropathic pain. However, we show here that the cholera toxin B subunit (CTB), a neuronal tracer used to demonstrate the sprouting of A-fibers in several earlier studies, also labels unmyelinated afferents (C-fibers) in lamina II and thin myelinated afferents in lamina I, when applied after peripheral nerve transection. The lamina II afferents also contained vasoactive intestinal polypeptide and galanin, two neuropeptides mainly expressed in small dorsal root ganglion (DRG) neurons and C-fibers. In an attempt to label large DRG neurons and A-fibers selectively, CTB was applied four days before axotomy (pre-injury-labelling), and sprouting was monitored after axotomy. We found that only a small number of A-fibers sprouted into inner lamina II, a region normally innervated by C-fibers, but not into outer lamina II or lamina I. Such sprouts made synaptic contact with dendrites in inner lamina II. Neuropeptide Y (NPY) was found in these sprouts in inner lamina II, an area very rich in Y1 receptor-positive processes. These results suggest that axotomy-induced sprouting from deeper to superficial layers is much less pronounced than previously assumed, in fact it is only marginal. This limited reorganization involves large NPY immunoreactive DRG neurons sprouting into the Y1 receptor-rich inner lamina II. Even if quantitatively small, it cannot be excluded that this represents a functional circuitry involved in neuropathic pain.


Subject(s)
Afferent Pathways/physiology , Nerve Fibers, Myelinated/physiology , Nerve Regeneration/physiology , Neuronal Plasticity/physiology , Posterior Horn Cells/physiology , Spinal Nerve Roots/physiology , Afferent Pathways/ultrastructure , Animals , Cells, Cultured , Cholera Toxin/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Immunohistochemistry , Male , Microscopy, Electron , Nerve Crush , Nerve Fibers, Myelinated/ultrastructure , Nerve Fibers, Unmyelinated/physiology , Nerve Fibers, Unmyelinated/ultrastructure , Neurofilament Proteins/metabolism , Neuropeptide Y/metabolism , Posterior Horn Cells/ultrastructure , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide Y/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/physiology , Sciatic Nerve/surgery , Spinal Nerve Roots/ultrastructure
18.
Proc Natl Acad Sci U S A ; 99(12): 8360-5, 2002 Jun 11.
Article in English | MEDLINE | ID: mdl-12060780

ABSTRACT

Phenotypic modification of dorsal root ganglion (DRG) neurons represents an important mechanism underlying neuropathic pain. However, the nerve injury-induced molecular changes are not fully identified. To determine the molecular alterations in a broader way, we have carried out cDNA array on the genes mainly made from the cDNA libraries of lumbar DRGs of normal rats and of rats 14 days after peripheral axotomy. Of the 7,523 examined genes and expressed sequence tags (ESTs), the expression of 122 genes and 51 expressed sequence tags is strongly changed. These genes encompass a large number of members of distinct families, including neuropeptides, receptors, ion channels, signal transduction molecules, synaptic vesicle proteins, and others. Of particular interest is the up-regulation of gamma-aminobutyric acid(A) receptor alpha5 subunit, peripheral benzodiazepine receptor, nicotinic acetylcholine receptor alpha7 subunit, P2Y1 purinoceptor, Na(+) channel beta2 subunit, and L-type Ca(2+) channel alpha2delta-1 subunit. Our findings therefore reveal dynamic and complex changes in molecular diversity among DRG neurons after axotomy. Sequences reported in this paper have been deposited in the GenBank database (accession numbers BG 662484-BG 673712)


Subject(s)
Ganglia, Spinal/metabolism , Gene Expression Regulation , Neuritis/physiopathology , Sciatic Nerve/physiology , Animals , Axotomy , Disease Models, Animal , Expressed Sequence Tags , Ganglia, Spinal/physiopathology , Gene Expression Profiling , Molecular Sequence Data , Neuritis/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sciatic Nerve/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...