Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Zhen Ci Yan Jiu ; 41(1): 65-9, 2016 Feb.
Article in Chinese | MEDLINE | ID: mdl-27141624

ABSTRACT

OBJECTIVE: To observe the effect of electroacupuncture (EA) intervention combined with medication (Gastrodin) on changes of neurological function and expression of Nogo-A and Nogo-A receptor (NgR) in the frontal lobe cortex around the ischemic loci of focal cerebral ischemia (FCI) rats, so as to explore its mechanism underlying improvement of neuroregeneration of FC. METHODS: Fifty male Sprague-Dawley rats were randomly divided into normal control, model, EA, medication and EA+ medication groups (n = 10 in each group). The FCI model was induced by occlusion of the middle cerebral artery (MCAO) with thread embolus. EA was applied to the left "Quchi" (LI 11) and "Hegu" (Li 4) for 30 min, once daily for 14 days after MCAO. For rats of the medication group, Gastrodin (10 mg/kg) was administrated by intraperitoneal injection, once daily for 14 days. The neurological impairment was assessed by Zea Longa's scoring. The expression of Nogo-A and NgR in the frontal lobe cortex around the ischemic loci was detected by immunohistochemistry. RESULTS: In comparison with the normal control group, cerebro- cortical Nogo-A and NgR expression levels of the model group vere significantly increased (P < 0.05). Compared with the model group, the Zea Longa's score and Nogo-A and NgR expression levels were evidently down-regulated in the EA, medication and EA + medication groups (P < 0.05). The Zea Longa's score and Nogo-A and NgR expression levels were significantly lower in the EA + medication group than in the EA and medication groups (P < 0.05). CONCLUSION: EA intervention and Gastrodin administration can down-regulate cerebro-cortical Nogo-A and NgR protein expression in FCI rats, which may contribute to their action in improving neurological impairment. The effect of EA+ Gastrodin is better than simple EA or Gastrodin treatment.


Subject(s)
Benzyl Alcohols/administration & dosage , Brain Ischemia/therapy , Electroacupuncture , Frontal Lobe/metabolism , Glucosides/administration & dosage , Myelin Proteins/genetics , Myelin Proteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Brain Ischemia/drug therapy , Brain Ischemia/genetics , Brain Ischemia/metabolism , Combined Modality Therapy , Frontal Lobe/drug effects , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression , Humans , Male , Nogo Proteins , Nogo Receptor 1 , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics
2.
Zhen Ci Yan Jiu ; 40(2): 94-8, 2015 Apr.
Article in Chinese | MEDLINE | ID: mdl-26054191

ABSTRACT

OBJECTIVE: To observe the effect of Electroacupuncture (EA) stimulation of "Tianquan"(PC 2), "Quze" (PC 3), "Neiguan" (PC 6), "Daling" (PC 7) of the Pericardium Meridian on cerebral angiogenesis in cerebral ischemia (CI) rats, so as to reveal its mechanisms underlying improvement of stroke. METHODS: A total of 50 SD rats were equally randomized into normal control, sham, model, EA-Pericardium-Meridian acupoints (EA-PCM) and EA-Lung-Meridian acupoint (EA-LUM) groups. The CI model was established by occlusion of the middle cerebral artery. EA (2-4 V, 20 Hz) was applied to PC 2, PC 3, PC 6, PC 7 and "Tianfu"(LU 3), "Chize" (LU 5), "Lieque" (LU 7), "Taiyuan" (LU 9) of the Lung Meridian for 30 min, once at time-points of 0 h, 6 h, 24 h, 48 h and 72 h, respectively after modeling. Serum nerve growth factor (NGF) and Nogo protein-A (Nogo-A) contents were assayed by enzyme linked immunosorbent assay (ELISA), and cerebral NGF and Nogo-A immunoactivity levels in the ischemic cerebral tissue were detected by immunohistochemistry. RESULTS: (1) Compared to the normal control group, serum NGF and Nogo-A contents, and cerebral NGF immunoactivity level in the model group were significantly increased (P < 0.01). Following EA interventions, serum and cerebral NGF levels were further significantly up-regulated in the EA-PCM and EA-LUM groups (P < 0.01), while serum Nogo-A contents were down-regulated in the two EA groups (P < 0.01). The effect of EA-PCM was markedly superior to that of EA-LUM in up-regulating serum and cerebral NGF levels and down-regulating serum No- go-A level (P < 0.01). No significant differences were found between the normal control and sham groups in serum and cerebral NGF and Nogo-A levels (P > 0.05) , and among the 5 groups in cerebral Nogo-A levels (P > 0.05). CONCLUSION: EA stimulation of acupoints of both Pericardium Meridian and Lung Meridian can up-regulate serum NGF, cerebral NGF expression and down-regulate serum Nogo-A in CI rats, and the effect of Pericardium Meridian is markedly superior to that of Lung Meridian, suggesting a possible better nerve repair effect of EA-PCM acupoints on ischemic brain.


Subject(s)
Acupuncture Points , Brain Ischemia/therapy , Electroacupuncture , Myelin Proteins/blood , Nerve Growth Factor/blood , Animals , Brain Ischemia/blood , Brain Ischemia/genetics , Humans , Male , Meridians , Myelin Proteins/genetics , Nerve Growth Factor/genetics , Nogo Proteins , Rats , Rats, Sprague-Dawley
3.
Zhongguo Gu Shang ; 28(3): 235-9, 2015 Mar.
Article in Chinese | MEDLINE | ID: mdl-25936193

ABSTRACT

OBJECTIVE: To study the effects of Jisuikang (Chinese characters) on Nogo-NgR gene expression, and to explore the protective effects and mechanism of Jisuikang (Chinese characters) on spinal cord injury in rats. METHODS: One hundred eighty female rats were randomly assigned to 6 groups(30 rats per group). Sham group: T10 lamina was resected only and spinal cord was untreated. Model group: spine cord injury (SCI) was created with a modified impinger of Allen's by impacting on the T10 spinal cord. Prednisolone group: Prednisolone (0.06 g/kg) was given by intragastric administration at a time interval of 24 hours after operation. The Jisuikang (Chinese characters) high, moderate and low dose groups: Jisuikang (Chinese characters) was supplied with different dose (50 g/kg, 25 g/kg, 12.5 g/kg) by intragastric administration in rats after operation,for the first time at 30 min after surgery. Animals were killed 3, 7, 14 days after surgery. The expression levels of Nogo-A and NgR were observed by Western Blot and Real-time PCR. RESULTS: The expression of Nogo-A and NgR was at the basic level at all time points in sham group. Compared with model group, the protein expression levels of Nogo-A and NgR in sham, prednisolone, Jisuikang (Chinese characters) moderate dose groups were statistically significant at all time points (P < 0.05). No difference was found in Jisuikang (Chinese characters) high and low dose groups (P > 0.05). Three days after surgery, the mRNA levels of Nogo-A and NgR in treatment group were significantly lower than that in model group (P < 0.01); 7 days after surgery,Nogo-A and NgR mRNA expression were dramatically upregulated and peaked; 14 days after operation, the expression was decreased, but still significantly higher than that in other treatment groups (P < 0.01). Prednisolone and Jisuikang (Chinese characters) moderate dose groups showed the most significant effects among all groups,but there was no statistically significant difference between two groups (P > 0.05). CONCLUSION: The decoction Jisuikang (Chinese characters) can promote the nerve cell regeneration by regulating Nogo-A and NgR gene expression, activating Nogo- NgR signaling pathways after acute spinal cord injury.


Subject(s)
Medicine, Chinese Traditional , Myelin Proteins/genetics , Receptors, Cell Surface/genetics , Spinal Cord Injuries/drug therapy , Animals , Female , GPI-Linked Proteins/analysis , GPI-Linked Proteins/genetics , GPI-Linked Proteins/physiology , Myelin Proteins/analysis , Myelin Proteins/physiology , Nerve Regeneration/drug effects , Nogo Proteins , Nogo Receptor 1 , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/analysis , Receptors, Cell Surface/physiology , Signal Transduction/drug effects , Spinal Cord Injuries/metabolism
4.
J Ethnopharmacol ; 158 Pt A: 102-12, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25448504

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Zuo-Gui pills (ZGPs) and You-Gui pills (YGPs) are 2 traditional Chinese herbal formulas used for treating multiple sclerosis (MS) in the clinical setting and have been shown to have neuroprotective effects in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. The aim of this study was to explore the mechanisms underlying the neuroprotective functions of ZGPs and YGPs. MATERIALS AND METHODS: Female Lewis rats were randomly divided into normal control, EAE model, 2g/kg ZGP-treated EAE, 3g/kg YGP-treated EAE, and prednisone acetate-treated groups. EAE model was induced by subcutaneous injection of MBP68-86 antigen. The neurological function scores were estimated. Histological structures of the brains and spinal cords were observed, and myelinated and axons imaged. NogoA, Nogo receptor (NgR), and RhoA transcript and protein levels were measured by real-time quantitative RT-PCR and western blotting on postimmunization (PI) days 14 (acute stage) and 28 (remission stage). RESULTS: ZGPs and YGPs significantly reduced neurological functions scores and abrogated inflammatory infiltrates, demyelination, and axonal damage. Furthermore, treatment with ZGPs and YGPs inhibited NogoA, NgR, and RhoA mRNA and protein expression in rats at both the acute and remission stages. ZGPs exhibited stronger effects on NogoA and RhoA expressions, as well as neurological function, during the acute stage of EAE, while YGPs caused greater reductions in NogoA expression during the remission stage. CONCLUSIONS: Our findings suggested that ZGPs and YGPs exerted neuroprotective effects by downregulation of NogoA, NgR, and RhoA pathways, with differences in response times and targets observed between ZGPs and YGPs.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Neuroprotective Agents/pharmacology , Animals , Disease Models, Animal , Down-Regulation/drug effects , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , GPI-Linked Proteins/genetics , Multiple Sclerosis/drug therapy , Multiple Sclerosis/physiopathology , Myelin Proteins/genetics , Nogo Proteins , Nogo Receptor 1 , Rats , Rats, Inbred Lew , Real-Time Polymerase Chain Reaction , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , rhoA GTP-Binding Protein/genetics
5.
J Neuroimmune Pharmacol ; 8(1): 238-50, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22870853

ABSTRACT

Different experimental autoimmune encephalomyelitis models (EAE) have been developed. However, due to the different experimental conditions applied, observations simultaneously considering different pathological targets are still scarce. Using EAE induced in Dark Agouti rats with syngenic whole spinal cord homogenate suspended in incomplete Freund's adjuvant, we here analyze neurosteroidogenic machinery, cytokine levels, microglial cells, infiltration of inflammatory cells, myelin proteins and Na(+), K(+)-ATPase pump activity in the spinal cord. Data obtained in the acute phase of the disease confirmed that neurological signs were accompanied by the presence of perivascular infiltrating T cells (CD3(+) cells) and activated monocytic/microglial cells (ED1(+) and MHC-II(+)) in the spinal cord. In particular, the number of MHC-II(+) cells was significantly increased in association with increased expression of pro- (i.e., TNF-α, IL-1ß) and anti-inflammatory (i.e., TGF-ß) cytokines as well as with decreased expression of proteolipid protein and myelin basic protein. During the chronic phase of the disease, the number of MHC-II(+) cells was still increased, although less than in the acute phase. Changes in the number of MHC-II(+) cells were associated with decreased Na(+),K(+)-ATPase enzymatic activity. A general decrease in the levels of neuroactive steroids, with the exception of an increase in tetrahydroprogesterone and 17ß-estradiol, was detected in the acute phase. These changes were maintained or reverted in the chronic phase of EAE. In conclusion, we report that modifications in the neuroimmune response in the acute and chronic phases of EAE are associated with specific changes in myelin proteins, Na(+),K(+)-ATPase pump and in the levels of neuroactive steroids.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Acute Disease , Animals , Blood Cell Count , Chronic Disease , Cytokines/metabolism , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Fluorometry , Genes, MHC Class II/genetics , Immunohistochemistry , Male , Mass Spectrometry , Myelin Proteins/biosynthesis , Myelin Proteins/genetics , Neurons/pathology , Neutrophil Infiltration/physiology , Nuclease Protection Assays , Rats , Real-Time Polymerase Chain Reaction , Ribonucleases/metabolism , Signal Transduction/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Steroids/pharmacology , Steroids/therapeutic use
6.
ACS Chem Biol ; 7(7): 1205-13, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22530759

ABSTRACT

The structural integrity of myelin formed by Schwann cells in the peripheral nervous system (PNS) is required for proper nerve conduction and is dependent on adequate expression of myelin genes including peripheral myelin protein 22 (PMP22). Consequently, excess PMP22 resulting from its genetic duplication and overexpression has been directly associated with the peripheral neuropathy called Charcot-Marie-Tooth disease type 1A (CMT1A), the most prevalent type of CMT. Here, in an attempt to identify transcriptional inhibitors with therapeutic value toward CMT1A, we developed a cross-validating pair of orthogonal reporter assays, firefly luciferase (FLuc) and ß-lactamase (ßLac), capable of recapitulating PMP22 expression, utilizing the intronic regulatory element of the human PMP22 gene. Each compound from a collection of approximately 3,000 approved drugs was tested at multiple titration points to achieve a pharmacological end point in a 1536-well plate quantitative high-throughput screen (qHTS) format. In conjunction with an independent counter-screen for cytotoxicity, the design of our orthogonal screen platform effectively contributed to selection and prioritization of active compounds, among which three drugs (fenretinide, olvanil, and bortezomib) exhibited marked reduction of endogenous Pmp22 mRNA and protein. Overall, the findings of this study provide a strategic approach to assay development for gene-dosage diseases such as CMT1A.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Drug Delivery Systems/methods , Gene Dosage/physiology , Gene Targeting/methods , Myelin Proteins/antagonists & inhibitors , Myelin Proteins/genetics , Capsaicin/administration & dosage , Capsaicin/analogs & derivatives , Charcot-Marie-Tooth Disease/drug therapy , Charcot-Marie-Tooth Disease/metabolism , Drug Evaluation, Preclinical/methods , Fenretinide/administration & dosage , Gene Dosage/drug effects , Humans , Myelin Proteins/biosynthesis
7.
J Tradit Chin Med ; 32(4): 664-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23427407

ABSTRACT

OBJECTIVE: To observe the effect of tonifying liver and kidney-essence herbs on expression of a nerve regeneration inhibitor, Nogo for neuron A (Nogo-A), and its associated signaling molecule, low-affinity neurotrophin receptor p75 (p75(NTR)), in rats with cerebral ischemic stroke (CIS), with the aim of exploring the possible mechanism of tonifying liver and kidney-essence herbs in recovery following injury to the central nervous system. METHODS: A cerebral ischemic stroke model in SD rats was established with the suture-occlusion method. Successful model rats were divided into placebo and herb groups at random; sham-operated and control groups were set up simultaneously. Each of these groups was divided into six subgroups at random. Expression of Nogo-A and p75(NTR) was evaluated with immunofluorescence microscopy at days 3, and weeks 1, 2, 3, 4 and 8 after administration. RESULTS: Tonifying liver and kidney-essence herbs suppressed the expression of Nogo-A and p75(NTR) (P < 0.05 and P < 0.01, respectively). CONCLUSION: Suppressing the expression of Nogo-A and p75(NTR) is possibly one of the mechanisms underlying the ability of tonifying liver and kidney-essence herbs to promote recovery of the injured central nervous system.


Subject(s)
Drugs, Chinese Herbal/administration & dosage , Myelin Proteins/genetics , Receptor, Nerve Growth Factor/genetics , Stroke/drug therapy , Animals , Disease Models, Animal , Gene Expression/drug effects , Humans , Male , Myelin Proteins/metabolism , Nogo Proteins , Rats , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor/metabolism , Stroke/genetics , Stroke/metabolism
8.
Behav Brain Res ; 224(1): 73-9, 2011 Oct 10.
Article in English | MEDLINE | ID: mdl-21645550

ABSTRACT

The reticulon-4 receptor, encoded by RTN4R, limits axonal sprouting and neural plasticity by inhibiting the outgrowth of neurites. Human association studies have implicated mutations in RTN4R in the development of schizophrenia, including the identification of several rare nonconservative missense mutations of RTN4R in schizophrenia patients. To investigate the effects of missense mutation of the reticulon-4 receptor on phenotypes relevant to schizophrenia, we behaviourally characterized a novel Rtn4r mutant mouse line with an amino acid substitution (R189H) in the Nogo-66 binding site. Behavioural assays included prepulse inhibition of acoustic startle, locomotor activity, social interaction and spatial cognition. When compared with wildtype littermates, Rtn4r mutant mice exhibited greater social preference, which may reflect a social-anxyolitic effect, and a mild impairment in spatial cognition. Given the mild effect of the R189H mutation of Rtn4r on behavioural phenotypes relevant to schizophrenia, our results do not support missense mutation of RTN4R as a strong risk factor in the pathogenesis of schizophrenia.


Subject(s)
Interpersonal Relations , Memory Disorders/genetics , Mutation, Missense/genetics , Myelin Proteins/genetics , Receptors, Cell Surface/genetics , Acoustic Stimulation/adverse effects , Animals , Arginine/genetics , Behavior, Animal , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/genetics , Histidine/genetics , Inhibition, Psychological , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Myelin Proteins/deficiency , Nogo Receptor 1 , Receptors, Cell Surface/deficiency , Reflex, Acoustic/genetics
9.
Pediatr Neurol ; 41(4): 305-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19748054

ABSTRACT

Curcumin is the newest therapeutic agent for ameliorating the clinical and neuropathologic phenotype of a mouse model of Déjérine-Sottas disease. We undertook a 12-month dose-escalation safety trial of oral curcumin in a 15-year-old Caucasian girl with Déjérine-Sottas disease (point mutation, Ser72Leu) complicated by severe weakness, scoliosis, and respiratory impairment. The patient received 50 mg/kg/day oral curcumin for the first 4 months and 75 mg/kg/day thereafter, to complete a 12-month trial. Outcome measures included muscle strength, pulmonary function, upper/lower extremity disability, neurophysiologic studies, and health-related quality of life. After 12 months, the patient experienced no adverse events, and reported good compliance. There was little improvement in objective outcome measures. Knee flexion and foot strength increased slightly, but hand and elbow strength decreased. Pulmonary function, hand function, and measures of upper/lower extremity disability were stable or reduced. Her neurophysiologic findings were unchanged. Parent-reported quality of life improved for most domains, especially self-esteem, during the 12 months of treatment. Child-reported quality of life, assessed at the final visit, mirrored these results, with overall feelings of happiness and contentment. Further studies are required to explore the efficacy and safety of curcumin for severe demyelinating neuropathies of infancy and early childhood.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Curcumin/therapeutic use , Hereditary Sensory and Motor Neuropathy/drug therapy , Administration, Oral , Adolescent , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Curcumin/administration & dosage , Drug-Related Side Effects and Adverse Reactions , Female , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Myelin Proteins/genetics , Point Mutation , Quality of Life , Sural Nerve/pathology , Treatment Outcome
10.
Otol Neurotol ; 30(5): 668-75, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19546827

ABSTRACT

HYPOTHESIS: Different members of the Nogo system are expressed in the mammalian cochlea. BACKGROUND: The protein Nogo has gained a lot of attention during the last couple of years because it inhibits neurite outgrowth in the adult central nervous system. In contrast to the central nervous system, very little is known regarding the expression and possible function of the Nogo system within the inner ear. METHODS: Using reverse-transcriptase-polymerase chain reaction and immunohistochemistry, we analyzed for the expression of members of the Nogo system within the cochlea. In addition, we determined hearing levels of Nogo A knockout and wild-type mice with auditory brainstem response audiometry. RESULTS: In this study, we demonstrate the expression of Nogo A, B, C, and of Nogo receptor mRNA in the organ of Corti, spiral ganglion, and stria vascularis. Immunohistochemistry revealed that Nogo A and Nogo receptor localize to the spiral ganglion neurons. Interestingly, Nogo A expression was also observed in the outer and inner hair cells of the organ of Corti. As revealed by light microscopy, deletion of Nogo A does not alter cochlear microanatomy. We have assessed hearing levels in 10-month old wild-type and Nogo A knockout mice, and thereby, we could not detect any differences between these 2 groups. CONCLUSION: Different members of the Nogo family are expressed in the mammalian cochlea. Deletion of Nogo A does not alter cochlea microanatomy or hearing levels compared with wild-type mice.


Subject(s)
Cochlea/physiology , Myelin Proteins/genetics , Myelin Proteins/physiology , Acoustic Stimulation , Animals , Audiometry , Auditory Threshold/physiology , Cochlea/anatomy & histology , Fluorescent Antibody Technique , Hair Cells, Vestibular/metabolism , Hearing Loss/genetics , Hearing Loss/physiopathology , Immunohistochemistry , Mammals , Mice , Mice, Inbred C57BL , Mice, Knockout , Nogo Proteins , Organ Culture Techniques , RNA/biosynthesis , RNA/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
11.
Genes Brain Behav ; 8(2): 181-92, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19077178

ABSTRACT

The membrane protein Nogo-A inhibits neurite outgrowth and regeneration in the injured central nervous system, primarily because of its expression in oligodendrocytes. Hence, deletion of Nogo-A enhances regeneration following spinal cord injury. Yet, the effects of Nogo-A deletion on general behavior and cognition have not been explored. The possibility of potential novel functions of Nogo-A beyond growth inhibition is strongly suggested by the presence of subpopulations of neurons also expressing Nogo-A - not only during development but also in adulthood. We evaluated here Nogo-A(-/-) mice in a series of general basic behavioral assays as well as functional analyses related to brain regions with notable expression levels of Nogo-A. The SHIRPA protocol did not show any major basic behavioral changes in Nogo-A(-/-) mice. Anxiety-related behavior, pain sensitivity, startle reactivity, spatial learning, and associative learning also appeared indistinguishable between Nogo-A(-/-) and control Nogo-A(+/+) mice. However, motor co-ordination and balance were enhanced in Nogo-A(-/-) mice. Spontaneous locomotor activity was also elevated in Nogo-A(-/-) mice, but this was specifically observed in the dark (active) phase of the circadian cycle. Enhanced locomotor reaction to systemic amphetamine in Nogo-A(-/-) mice further pointed to an altered dopaminergic tone in these mice. The present study is the first behavioral characterization of mice lacking Nogo-A and provides significant insights into the potential behavioral relevance of Nogo-A in the modulation of dopaminergic and motor functions.


Subject(s)
Behavior, Animal/physiology , Myelin Proteins/genetics , Amphetamine/pharmacology , Animals , Anxiety/genetics , Anxiety/psychology , Association Learning/physiology , Avoidance Learning/physiology , Central Nervous System Stimulants/pharmacology , Cerebellum/cytology , Cerebellum/physiology , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Hypothalamus/cytology , Hypothalamus/physiology , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Motor Activity/physiology , Neurons/physiology , Nogo Proteins , Pain Measurement , Postural Balance/physiology , Psychomotor Performance/physiology , Reflex, Startle/physiology , Retinal Ganglion Cells/physiology
12.
Am J Hum Genet ; 81(3): 438-53, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17701891

ABSTRACT

Mutations in myelin genes cause inherited peripheral neuropathies that range in severity from adult-onset Charcot-Marie-Tooth disease type 1 to childhood-onset Dejerine-Sottas neuropathy and congenital hypomyelinating neuropathy. Many myelin gene mutants that cause severe disease, such as those in the myelin protein zero gene (MPZ) and the peripheral myelin protein 22 gene (PMP22), appear to make aberrant proteins that accumulate primarily within the endoplasmic reticulum (ER), resulting in Schwann cell death by apoptosis and, subsequently, peripheral neuropathy. We previously showed that curcumin supplementation could abrogate ER retention and aggregation-induced apoptosis associated with neuropathy-causing MPZ mutants. We now show reduced apoptosis after curcumin treatment of cells in tissue culture that express PMP22 mutants. Furthermore, we demonstrate that oral administration of curcumin partially mitigates the severe neuropathy phenotype of the Trembler-J mouse model in a dose-dependent manner. Administration of curcumin significantly decreases the percentage of apoptotic Schwann cells and results in increased number and size of myelinated axons in sciatic nerves, leading to improved motor performance. Our findings indicate that curcumin treatment is sufficient to relieve the toxic effect of mutant aggregation-induced apoptosis and improves the neuropathologic phenotype in an animal model of human neuropathy, suggesting a potential therapeutic role in selected forms of inherited peripheral neuropathies.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Curcumin/therapeutic use , Hereditary Sensory and Motor Neuropathy/drug therapy , Myelin Proteins/genetics , Schwann Cells/drug effects , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Apoptosis/drug effects , Curcumin/administration & dosage , Disease Models, Animal , HeLa Cells , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/pathology , Humans , Mice , Mice, Mutant Strains , Sciatic Nerve/drug effects , Sciatic Nerve/pathology
13.
Exp Neurol ; 197(1): 70-83, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16321384

ABSTRACT

Axons show a poor regenerative capacity following traumatic central nervous system (CNS) injury, partly due to the expression of inhibitors of axonal outgrowth, of which Nogo-A is considered the most important. We evaluated the acute expression of Nogo-A, the Nogo-66 receptor (NgR) and the novel small proline-rich repeat protein 1A (SPRR1A, previously undetected in brain), following experimental lateral fluid percussion (FP) brain injury in rats. Immunofluorescence with antibodies against Nogo-A, NgR and SPRR1A was combined with antibodies against the neuronal markers NeuN and microtubule-associated protein (MAP)-2 and the oligodendrocyte marker RIP, while Western blot analysis was performed for Nogo-A and NgR. Brain injury produced a significant increase in Nogo-A expression in injured cortex, ipsilateral external capsule and reticular thalamus from days 1-7 post-injury (P < 0.05) compared to controls. Increased expression of Nogo-A was observed in both RIP- and NeuN positive (+) cells in the ipsilateral cortex, in NeuN (+) cells in the CA3 region of the hippocampus and reticular thalamus and in RIP (+) cells in white matter tracts. Alterations in NgR expression were not observed following traumatic brain injury (TBI). Brain injury increased the extent of SPRR1A expression in the ipsilateral cortex and the CA3 at all post-injury time-points in NeuN (+) cells. The marked increases in Nogo-A and SPRR1A in several important brain regions suggest that although inhibitors of axonal growth may be upregulated, the injured brain is also capable of expressing proteins promoting axonal outgrowth following TBI.


Subject(s)
Brain Injuries/metabolism , Membrane Proteins/genetics , Myelin Proteins/genetics , Receptors, Cell Surface/genetics , Animals , Blotting, Western , Brain/pathology , Brain Injuries/pathology , Cell Count , Cornified Envelope Proline-Rich Proteins , Densitometry , Functional Laterality/physiology , GPI-Linked Proteins , Hippocampus/metabolism , Hippocampus/pathology , Immunohistochemistry , Male , Microtubule-Associated Proteins/metabolism , Nogo Proteins , Nogo Receptor 1 , Oligodendroglia/metabolism , Rats , Rats, Sprague-Dawley , Thalamus/metabolism , Thalamus/pathology
14.
Biochem J ; 385(Pt 1): 125-34, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15350194

ABSTRACT

Over the last few years, the widely distributed family of reticulons (RTNs) is receiving renewed interest because of the implication of RTN4/Nogo in neurite regeneration. Four genes were identified in mammals and are referred to as RTN1, 2, 3 and the neurite outgrowth inhibitor RTN4/Nogo. In the present paper, we describe the existence of five new isoforms of RTN3 that differ in their N-termini, and analysed their tissue distribution and expression in neurons. We redefined the structure of human and murine rtn3 genes, and identified two supplementary exons that may generate up to seven putative isoforms arising by alternative splicing or differential promoter usage. We confirmed the presence of five of these isoforms at the mRNA and protein levels, and showed their preferential expression in the central nervous system. We analysed rtn3 expression in the cerebellum further, and observed increased levels of several of the RTN3 isoforms during cerebellum development and during in vitro maturation of cerebellar granule cells. This pattern of expression paralleled that shown by RTN4/Nogo isoforms. Specifically, RTN3A1 expression was down-regulated upon cell death of cerebellar granule neurons triggered by potassium deprivation. Altogether, our results demonstrate that the rtn3 gene generates multiple isoforms varying in their N-termini, and that their expression is tightly regulated in neurons. These findings suggest that RTN3 isoforms may contribute, by as yet unknown mechanisms, to neuronal survival and plasticity.


Subject(s)
Alternative Splicing/genetics , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Animals , Apoptosis , Base Sequence , Carrier Proteins/genetics , Cell Differentiation , Cells, Cultured , Cerebellum/cytology , Cloning, Molecular , Computational Biology , Exons/genetics , Humans , Introns/genetics , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Molecular Weight , Myelin Proteins/genetics , Myelin Proteins/metabolism , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Nogo Proteins , Organ Specificity , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Protein Transport , RNA Splice Sites/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
Blood ; 103(7): 2753-60, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14656887

ABSTRACT

DNA methylation plays critical roles in the development and differentiation of mammalian cells, and its dysregulation has been implicated in oncogenesis. This study was designed to determine whether DNA hypomethylation-associated aberrant gene expression is involved in adult T-cell leukemia (ATL) leukemogenesis. We isolated hypomethylated DNA regions of ATL cells compared with peripheral blood mononuclear cells from a carrier by a methylated CpG-island amplification/representational difference analysis method. The DNA regions identified contained MEL1, CACNA1H, and Nogo receptor genes. Sequencing using sodium bisulfite-treated genomic DNAs revealed the decreased methylated CpG sites, confirming that this method detected hypomethylated DNA regions. Moreover, these hypomethylated genes were aberrantly transcribed. Among them, MEL1S, an alternatively spliced form of MEL1 lacking the PR (positive regulatory domain I binding factor 1 and retinoblastoma-interacting zinc finger protein) domain, was frequently transcribed in ATL cells, and the transcriptional initiation sites were identified upstream from exons 4 and 6. Transfection of MEL1S into CTLL-2 cells conferred resistance against transforming growth factor beta (TGF-beta), suggesting that aberrant expression of MEL1S was associated with dysregulation of TGF-beta-mediated signaling. Although Tax renders cells resistant to TGF-beta, Tax could not be produced in most fresh ATL cells, in which MEL1S might be responsible for TGF-beta resistance. Our results suggest that aberrant gene expression associated with DNA hypomethylation is implicated in leukemogenesis of ATL.


Subject(s)
Alternative Splicing/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Leukemia-Lymphoma, Adult T-Cell/genetics , Transcription Factors/genetics , Zinc Fingers/genetics , Adult , Calcium Channels, T-Type/genetics , Cell Line, Transformed , Chromosome Mapping , DNA Primers , DNA, Complementary/genetics , DNA-Binding Proteins/chemistry , GPI-Linked Proteins , Human T-lymphotropic virus 1/genetics , Humans , Methylation , Models, Molecular , Myelin Proteins/genetics , Nogo Receptor 1 , Polymerase Chain Reaction , Protein Conformation , Receptors, Cell Surface/genetics , Restriction Mapping , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription Factors/chemistry
16.
Mult Scler ; 8(2): 130-8, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11990870

ABSTRACT

Treatment with paclitaxel by four intraperitoneal injections (20 mg/kg) 1 week apart attenuated clinical signs in a spontaneously demyelinating model, if given with onset of clinical signs. If given at 2 months of age (1 month prior to clinical signs), disease was almost completely prevented The astrogliosis, prominent in our model, was reversed by paditaxel as determined by astrocyte counts and quantitation of GFAP. Electron microscopic examination of affected regions at 2.5 months demonstrated that the myelin was generally normal. By 4 months of age, demyelination was common in the superior cerebellar peduncle, maximal at 6 months, but continued to 8 months. In addition to myelin vacuolation and nude axons, the presence of many thin myelin sheaths suggested remyelination or partial demyelination. Although no evidence of oligodendrocyte loss was seen, nuclear changes were observed. To substantiate that remyelination was occurring, we measured MBP (18.5 kDa), MBP-exon II, Golli-MBP, TP8, Golli-MBP-J37, platelet-derived growth factor alpha (PDGFR alpha) and sonic hedgehog (SHH). Of these TP8, PDGFR alpha and SHH were up-regulated in the untreated transgenic. After paditaxel treatment, MBP-Exon II, TP8, PDGFR alpha and SHH were further up-regulated. We concluded that some of the effects of paditaxel were to stimulate proteins involved in early myelinating events possibly via a signal transduction mechanism.


Subject(s)
Hereditary Central Nervous System Demyelinating Diseases/drug therapy , Multiple Sclerosis , Nervous System Autoimmune Disease, Experimental/drug therapy , Paclitaxel/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/ultrastructure , Biomarkers , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Division/drug effects , Cell Nucleus/ultrastructure , Cerebellum/pathology , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/analysis , Gliosis/drug therapy , Gliosis/genetics , Gliosis/pathology , Hereditary Central Nervous System Demyelinating Diseases/genetics , Hereditary Central Nervous System Demyelinating Diseases/pathology , Injections, Intraperitoneal , Mice , Mice, Neurologic Mutants , Mice, Transgenic , Myelin Proteins/biosynthesis , Myelin Proteins/genetics , Myelin Proteolipid Protein/genetics , Myelin Sheath/drug effects , Myelin Sheath/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nervous System Autoimmune Disease, Experimental/genetics , Nervous System Autoimmune Disease, Experimental/pathology , Oligodendroglia/pathology , Signal Transduction/drug effects
17.
Cell Death Differ ; 5(1): 116-25, 1998 Jan.
Article in English | MEDLINE | ID: mdl-10200452

ABSTRACT

C6.9 rat glioma cells undergo a cell death program when exposed to 1, 25-dihydroxyvitamin D3 (1,25-D3). As a global analytical approach, we have investigated gene expression in C6.9 engaged in this cell death program using differential screening of a rat brain cDNA library with probes derived from control and 1,25-D3-treated cells. Using this methodology we report the isolation of 61 differentially expressed cDNAs. Forty-seven cDNAs correspond to genes already characterized in rat cells or tissues. Seven cDNAs are homologous to yeast, mouse or human genes and seven are not related to known genes. Some of the characterized genes have been reported to be differentially expressed following induction of programmed cell death. These include PMP22/gas3, MGP and beta-tubulin. For the first time, we also show a cell death program induced up-regulation of the c-myc associated primary response gene CRP, and of the proteasome RN3 subunit and TCTP/mortalin genes. Another interesting feature of this 1,25-D3 induced-cell death program is the down-regulated expression of transcripts for the microtubule motor dynein heavy chain/MAP 1C and of the calcium-binding S100beta protein. Finally 15 upregulated cDNAs encode ribosomal proteins suggesting a possible involvement of the translational apparatus in this cell program. Alternatively, these ribosomal protein genes could be up-regulated in response to altered rates of cellular metabolism, as has been demonstrated for most of the other isolated genes which encode proteins involved in metabolic pathways. Thus, this study presents to our knowledge the first characterization of genes which are differentially expressed during a cell death program induced by 1, 25-D3. Therefore, this data provides new information on the fundamental mechanisms which participate in the antineoplastic effects of 1,25-D3 and on the machinery of a cell death program in a glioma cell line.


Subject(s)
Apoptosis/drug effects , Biomarkers, Tumor , Calcitriol/pharmacology , Calcium Channel Agonists/pharmacology , Extracellular Matrix Proteins , Glioma , Vitamin D/pharmacology , Animals , Apoptosis/physiology , Bone and Bones/physiology , Calcium-Binding Proteins/genetics , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cysteine , Cysteine Endopeptidases/genetics , DNA/analysis , DNA, Complementary , Dyneins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Library , HSP70 Heat-Shock Proteins/genetics , Multienzyme Complexes/genetics , Myelin Proteins/genetics , Neoplasm Proteins/genetics , Osteonectin/genetics , Proteasome Endopeptidase Complex , Protein Biosynthesis/physiology , RNA, Messenger/analysis , Rats , Ribosomal Proteins/genetics , Tubulin/genetics , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/physiology , Tumor Protein, Translationally-Controlled 1 , Matrix Gla Protein
18.
Genomics ; 41(1): 40-8, 1997 Apr 01.
Article in English | MEDLINE | ID: mdl-9126480

ABSTRACT

CL-20 is a novel gene encoding a protein that is structurally related to but distinct from the peripheral myelin protein PMP22. Like PMP22, CL-20 is likely to play important roles in the regulation of cell proliferation, differentiation, and cell death. In this study, we describe the cloning and sequencing of a cDNA encoding the human homologue of CL-20 and characterize the genomic structure of this gene. The hCL-20 gene (HGMW-approved symbol EMP1) encodes a protein of 157 amino acids that exhibits 76% identity to the rabbit CL-20 and to the rat EMP-1, which have been described recently, and 39% identity to human PMP22. CL-20 contains four hydrophobic domains, suggesting that it is an integral membrane protein. In particular the second hydrophobic domain encoded within the fourth exon is highly conserved among CL-20, EMP-1, and PMP22, suggesting a functional role for this region. CL-20 mRNA is abundant in squamous-differentiated bronchial epithelial cells; however, low levels of CL-20 mRNA can be detected in several human tissues by Northern analysis. Retinoic acid, which inhibits squamous differentiation, represses CL-20 expression in normal human bronchial epithelial cells. The genomic structure of the hCL-20 gene was analyzed using a P1 vector containing this gene. The hCL-20 gene contains five exons about 0.2, 0.12, 0.1, 0.14, and 2.2 kb and four introns about 15, 1.9, 0.1, and 0.7 kb. We have mapped the hCL-20 gene to chromosome 12p12 by fluorescence in situ hybridization.


Subject(s)
DNA, Complementary/genetics , Membrane Proteins/genetics , Myelin Proteins/genetics , Receptors, Cell Surface , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Chromosomes, Human, Pair 12/genetics , Cloning, Molecular , Conserved Sequence , DNA Primers/genetics , Epithelium/metabolism , Exons , Gene Expression Regulation/drug effects , Humans , Introns , Molecular Sequence Data , Neoplasm Proteins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Rats , Retinoids/pharmacology , Sequence Homology, Amino Acid , Species Specificity , Tissue Distribution
19.
J Biol Chem ; 270(48): 28910-6, 1995 Dec 01.
Article in English | MEDLINE | ID: mdl-7499420

ABSTRACT

In this study, we identify and characterize a novel gene, CL-20, that encodes a 17.8-kDa protein with sequence and structural similarity to the growth arrest-specific gene gas3/peripheral myelin protein gene PMP22. The CL-20 protein exhibits a 43% identity with PMP22. The positions of the four lipophilic domains and the N-glycosylation site of PMP22 are conserved in CL-20, suggesting that it also is an integral membrane glycoprotein. The CL-20 gene is located on human chromosome 12 rather than 17 and encodes a 2.8-kilobase mRNA instead of 1.7-kilobase mRNA. These observations indicate that the CL-20 gene is related to but distinct from PMP22. In contrast to PMP22, CL-20 mRNA and protein are induced during squamous differentiation of rabbit tracheal epithelial cells in vitro, and Northern blot analysis and in situ hybridization demonstrated that CL-20 mRNA is most abundant in squamous epithelia. These results indicate that the high expression of CL-20 is closely correlated with squamous differentiation. The differences in tissue-specific expression and regulation between CL-20 and PMP22 suggest different roles for these two proteins. Retinoids, which inhibit squamous differentiation, repress the induction of CL-20. The retinoic acid receptor-selective retinoid SRI-6751-84 is the most effective in suppressing CL-20, suggesting that the activation of the retinoic acid receptor signaling pathway is important in this suppression.


Subject(s)
Membrane Glycoproteins/genetics , Myelin Proteins/genetics , Receptors, Cell Surface , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation , Cell Line , Chromosome Mapping , Chromosomes, Human, Pair 12 , Cloning, Molecular , DNA, Complementary , Epithelial Cells , Gene Expression Regulation/drug effects , Humans , Membrane Glycoproteins/chemistry , Molecular Sequence Data , Myelin Proteins/chemistry , Neoplasm Proteins , Rabbits , Retinoids/pharmacology , Sequence Homology, Amino Acid
20.
Brain Res Mol Brain Res ; 29(1): 119-30, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7539519

ABSTRACT

In view of the profound effects of thyroid hormone deficiency on the central nervous system (CNS), neuronal genes regulated by thyroid hormone could potentially be involved in the development of the CNS. Expression of the neuronal gene RC3/neurogranin was shown to be induced by thyroid hormone in the rat. No data are available on RC3 expression in mammals with prenatal brain development, like humans. To study RC3 mRNA expression in a genetic in vivo model of congenital hypothyroidism, which also resembles the human situation in the timing of brain development relative to birth, we used an inbred strain of congenitally hypothyroid goats. We isolated a cDNA for the caprine RC3 homolog. The deduced amino acid sequence had 99% similarity with the rat and bovine protein sequence. An analysis of the developmental expression of RC3 mRNA levels showed a 3-fold increase between E90 and P0. In situ hybridization analysis showed that in euthyroid goats, the RC3 expression pattern was region-specific and resembled that in rats. However, in contrast to rats, hypothyroid goats showed only a reduced RC3 mRNA expression in the striatum. Hypothyroidism had no effect on RC3 mRNA expression in all other brain regions. T4-treatment of the hypothyroid fetus increased RC3 mRNA expression in the striatum to euthyroid control levels. These data suggest that thyroid hormone is a regulator of RC3 gene expression in the caprine brain, and that the striatum is highly sensitive to thyroid hormone deficiency.


Subject(s)
Brain/metabolism , Calmodulin-Binding Proteins/chemistry , Calmodulin-Binding Proteins/metabolism , Congenital Hypothyroidism , Goats/metabolism , Hypothyroidism/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Calmodulin-Binding Proteins/genetics , Cattle , DNA, Complementary/genetics , Female , Humans , Molecular Sequence Data , Myelin Proteins/genetics , Myelin Proteolipid Protein , Nerve Tissue Proteins/genetics , Neurogranin , Pregnancy , RNA, Messenger/metabolism , Rats , Thyroxine/blood , Thyroxine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL