Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
Add more filters

Country/Region as subject
Publication year range
2.
Gene Ther ; 22(7): 546-52, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25809464

ABSTRACT

We have previously demonstrated that both age-related and noise-induced hearing loss are reduced in transgenic mice that ubiquitously overexpress X-linked inhibitor of apoptosis protein (XIAP). In view of the therapeutic implications of these findings, we have developed a minimally invasive surgical method to deliver adenoid-associated virus (AAV) across the round window membrane (RWM) of the cochlea, enabling efficient gene transfer to hair cells and sensory neurons in this enclosed structure. This RWM approach was used in the present study to evaluate the effectiveness of AAV-mediated XIAP overexpression in protecting against cisplatin-induced ototoxicity. Two weeks following surgery, AAV-derived XIAP was detected in the majority of inner and outer hair cells, resulting in a threefold elevation of this antiapoptotic protein in the cochlea. The protection of AAV-mediated XIAP overexpression was evaluated in animals treated with cisplatin at a dose of 4 mg kg(-1) per day for 4-7 consecutive days. The XIAP overexpression was found to attenuate cisplatin-induced hearing loss by ~22 dB. This was accompanied by a reduction of the loss of vulnerable hair cells and sensory neurons in the cochlea by 13%.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Cochlea/drug effects , Dependovirus/metabolism , Hearing Loss/chemically induced , Hearing Loss/prevention & control , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , Cochlea/metabolism , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors , Guinea Pigs , Hair Cells, Auditory/metabolism , Round Window, Ear/metabolism , Sensory Receptor Cells/metabolism , Transfection , X-Linked Inhibitor of Apoptosis Protein/genetics
3.
Gene Ther ; 19(3): 255-63, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21697953

ABSTRACT

The auditory portion of the inner ear, the cochlea, is an ideal organ for local gene transfection owing to its relative isolation. Various carriers have been tested for cochlear gene transfection. To date, viral vectors appear to have much higher transfection efficacy than non-viral mechanisms. Among these vectors, recombinant adeno-associated virus (rAAV) vectors have several advantages such as being non-pathogenic and the ability to produce prolonged gene expression in various cell types. However, rAAV vectors cannot pass through the intact round window membrane (RWM), otherwise a very attractive approach to access the human inner ear. In this study, performed in guinea-pigs, we describe a method to increase the permeability of RWM to rAAV vectors by partial digestion with collagenase solution. Elevated delivery of rAAV across the partially digested RWM increased transfection efficacy to a satisfactory level, even though it was still lower than that achieved by direct cochleostomy injection. Functional tests (auditory brainstem responses) showed that this enzymatic manipulation did not cause permanent hearing loss if applied appropriately. Morphological observations suggested that the damage to RWM caused by partial digestion healed within four weeks. Taken together, these findings suggest that partial digestion of the RWM is a safe and effective method for increasing the transfection of cochlear sensory cells with rAAV.


Subject(s)
Cochlea/metabolism , Dependovirus/metabolism , Genetic Vectors/metabolism , Round Window, Ear/metabolism , Transfection , Animals , Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , Guinea Pigs , Hair Cells, Auditory/metabolism , Humans , Male
4.
Gene Ther ; 18(6): 560-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21228883

ABSTRACT

Apoptosis is responsible for cochlear cell death induced by noise. Here, we show that transgenic (TG) mice that overexpress X-linked inhibitor of apoptosis protein (XIAP) under control of the ubiquitin promoter display reduced hearing loss and cochlear damage induced by acoustic overstimulation (125 dB sound pressure level, 6 h) compared with wild-type (WT) littermates. Hearing status was evaluated using the auditory brainstem response (ABR), whereas cochlear damage was assessed by counts of surviving hair cells (HCs) and spiral ganglion neurons (SGNs) as well as their fibers to HCs. Significantly smaller threshold shifts were found for TG mice than WT littermates. Correspondingly, the TG mice also showed a reduced loss of HCs, SGNs and their fibers to HCs. HC loss was limited to the basal end of the cochlea that detects high frequency sound. In contrast, the ABRs demonstrated a loss of hearing sensitivity across the entire frequency range tested (2-32 kHz) indicating that the hearing loss could not be fully attributed to HC loss alone. The TG mice displayed superior hearing sensitivity over this whole range, suggesting that XIAP overexpression reduces noise-induced hearing loss not only by protecting HCs but also other components of the cochlea.


Subject(s)
Hearing Loss, Noise-Induced/genetics , X-Linked Inhibitor of Apoptosis Protein/genetics , Animals , Auditory Threshold , Cochlea/injuries , Evoked Potentials, Auditory, Brain Stem , Hair Cells, Auditory/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Spiral Ganglion/innervation , Ubiquitin/genetics
5.
Nat Med ; 3(9): 997-1004, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9288726

ABSTRACT

We show here that transient forebrain ischemia selectively elevates levels of neuronal apoptosis inhibitory protein (NAIP) in rat neurons that are resistant to the injurious effects of this treatment. This observation suggests that increasing NAIP levels may confer protection against ischemic cell death. Consistent with this proposal, we demonstrate that two other treatments that increase neuronal NAIP levels, systemic administration of the bacterial alkaloid K252a and intracerebral injection of an adenovirus vector capable of overexpressing NAIP in vivo, reduce ischemic damage in the rat hippocampus. Taken together, these findings suggest that NAIP may play a key role in conferring resistance to ischemic damage and that treatments that elevate neuronal levels of this antiapoptotic protein may have utility in the treatment of stroke.


Subject(s)
Hippocampus/injuries , Hippocampus/metabolism , Ischemic Attack, Transient/metabolism , Nerve Tissue Proteins/metabolism , Adenoviridae/genetics , Animals , Apoptosis/drug effects , Apoptosis/physiology , Carbazoles/administration & dosage , Carbazoles/therapeutic use , Gene Expression/drug effects , Genetic Therapy , Genetic Vectors , Hippocampus/blood supply , Indole Alkaloids , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/therapy , Male , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/therapeutic use , Nerve Tissue Proteins/genetics , Neuronal Apoptosis-Inhibitory Protein , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Rats , Rats, Wistar
6.
Neuroscience ; 155(3): 864-75, 2008 Aug 26.
Article in English | MEDLINE | ID: mdl-18640243

ABSTRACT

We compared the neuroprotective efficacy of a potent and CNS-penetrant cyclin dependent kinase (CDK) and glycogen synthase kinase 3 beta (GSK3beta) inhibitor (Compound 1) in juvenile (postnatal day 21; P21) and adult C57Bl/6 mice (postnatal day 60; P60) using a model of hypoxic-ischemic brain injury (HI). Neuronal cell counts and density measures from brain sections stained with Cresyl Violet revealed that exposure of P21 mice to 60 min of HI resulted in extensive damage to the ipsilateral cornu ammonis 1 (CA1) region of the hippocampus (40% cell loss) and striatum (30% cell loss) 7 days later. Exposure of P60 mice to 40 min of HI produced a similar pattern of cell loss. Intraperitoneal administration of Compound 1 (3 mg/kg) 1, 5 and 9 h after 60 min of HI did not reduce brain injury in P21 mice relative to vehicle controls. By contrast, in P60 mice, this treatment significantly decreased cell loss in the ipsilateral hippocampus (10% cell loss) and striatum (15% loss) relative to vehicle controls. Terminal uridine deoxynucleotidyl transferase (TUNNEL) positive cell counts and infarct volume were also substantially reduced in P60 mice treated with Compound 1. A motor coordination test performed twice weekly until 5 weeks post-HI confirmed that Compound 1 produced long lasting functional recovery. Our results indicate that Compound 1 produced long lasting neuroprotective effects in adult but not juvenile mice suggesting that inhibition of the CDKs and GSK3beta plays a distinct neuroprotective role in the juvenile and adult brain.


Subject(s)
Cyclin-Dependent Kinases/therapeutic use , Enzyme Inhibitors/therapeutic use , Glycogen Synthase Kinase 3/antagonists & inhibitors , Hypoxia-Ischemia, Brain/drug therapy , Neuroprotective Agents/therapeutic use , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Cell Death/drug effects , Disease Models, Animal , Glycogen Synthase Kinase 3/physiology , Glycogen Synthase Kinase 3 beta , Hypoxia-Ischemia, Brain/pathology , In Situ Nick-End Labeling/methods , Mice , Mice, Inbred C57BL , Time Factors
7.
Neuroscience ; 144(1): 1-7, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17112674

ABSTRACT

Recognition memory was assessed in adult rats that received bilateral injections of saline (sham lesions) or ibotenic acid (lesioned) in the ventral hippocampus as neonates (postnatal day 7, PD7) or young adult (42 days of age, PD42) using the Novel Object Recognition Test (NORT). Normal or sham-lesioned rats were able to distinguish novel from familiar objects over a 0.5 and 2 h delay between the sample and choice phases. Adult rats (PD70) lesioned as neonates performed progressively worse than sham-lesioned animals at delays of 0.5 and 2 h. A single injection of darbepoetin alfa (500 or 5000 U/kg, i.p.), given 1 h before the sample phase restored performance 0.5 or 2 h later in the choice phase to same levels as sham-lesioned rats. Adults lesioned on PD42 displayed deficits in NORT performance with a 2 h delay between the choice and sample phases that were completely reversed by administration of darbepoetin alfa (5000 U/kg, i.p.) 1 h before the sample phase. These results suggest that darbepoetin alfa may have utility in treating memory deficits associated with brain dysfunction related to developmental disorders such as schizophrenia.


Subject(s)
Erythropoietin/analogs & derivatives , Hematinics/therapeutic use , Hippocampus/pathology , Memory/drug effects , Recognition, Psychology/drug effects , Animals , Animals, Newborn , Conditioning, Operant/drug effects , Darbepoetin alfa , Erythropoietin/administration & dosage , Erythropoietin/therapeutic use , Hematinics/administration & dosage , Injections, Intraperitoneal , Rats , Rats, Long-Evans
8.
Hernia ; 11(5): 393-6, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17541495

ABSTRACT

BACKGROUND: Seroma are common early postoperative complications encountered in laparoscopic inguinal hernia repair. Previous anecdotal evidence from our surgical practice suggested a lower incidence of postoperative seroma formation with direct hernia repairs when the lax transversalis fascia (TF) is inverted by tacking to the pubic ramus. We undertook a study to investigate whether TF inversion in this way reduces the incidence of postoperative seroma. METHOD: A total of 216 patients undergoing transabdominal preperitoneal (TAPP) laparoscopic inguinal hernia repairs from August 2003 to December 2005 were included in this prospective non-randomised controlled study. Surgeon 1 would routinely invert the TF whereas surgeon 2 would not. At follow-up the presence of postoperative seroma and pain was recorded. RESULTS: Mann-Whitney U test demonstrated no significant difference in terms of age, sex and time to follow-up between the surgeons' patient groups (P > 0.05), and Chi-square test demonstrated significantly that inversion of the TF is associated with a lower incidence of postoperative seroma (P < 0.05). There was no significant difference in terms of postoperative pain at follow-up. CONCLUSION: Inversion of the TF is associated with a statistically lower incidence of postoperative seroma, without increasing postoperative pain despite the use of one or two additional tacks.


Subject(s)
Hernia, Inguinal/surgery , Laparoscopy/methods , Seroma/prevention & control , Suture Techniques , Abdominal Wall/surgery , Adolescent , Adult , Aged , Aged, 80 and over , Fasciotomy , Female , Follow-Up Studies , Humans , Laparoscopy/adverse effects , Male , Middle Aged , Prospective Studies , Pubic Bone/surgery , Seroma/etiology , Treatment Outcome
9.
Trends Neurosci ; 23(10 Suppl): S92-100, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11052226

ABSTRACT

The MPTP monkey is a well-characterized animal model of parkinsonism and provides an exceptional tool for the study of dyskinesias induced by dopamine-like agents. Several such agents have been tested during the past 15 years, and it has been found that the duration of action of these compounds is the most reliable variable with which to predict their dyskinesiogenic profile. It is proposed that L-dopa-induced dyskinesias represent a form of pathological learning caused by chronic pulsatile (nonphysiological) stimulation of dopamine receptors, which activates a cascade of molecular and biochemical events. These events include defective regulation of Fos proteins that belong to the deltaFosB family, increased expression of neuropeptides, and defective GABA- and glutamate-mediated neurotransmission in the output structures of the basal ganglia.


Subject(s)
Antiparkinson Agents/adverse effects , Basal Ganglia/drug effects , Dopamine Agonists/adverse effects , Dyskinesia, Drug-Induced/metabolism , Levodopa/adverse effects , Parkinsonian Disorders/metabolism , Receptors, Dopamine/drug effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Antiparkinson Agents/administration & dosage , Basal Ganglia/metabolism , Disease Models, Animal , Dopamine Agonists/administration & dosage , Haplorhini , Levodopa/administration & dosage , Neural Inhibition , Neuropeptides/metabolism , Parkinsonian Disorders/chemically induced , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Dopamine/metabolism , Receptors, GABA/metabolism , Receptors, Glutamate/metabolism , Signal Transduction
10.
Neuropharmacology ; 51(5): 974-85, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16901513

ABSTRACT

A major obstacle in the therapeutic development of phosphodiesterase-4 (PDE4) inhibitors is the production of adverse side effects such as nausea and vomiting. Immunohistochemical detection of Fos-like immunoreactivity (FLI) was used to address the neuroanatomical basis for the pharmacological actions of PDE4 inhibitors. The potent and selective PDE4 inhibitors 6-(4-pyridylmethyl)-8-(3-nitrophenyl) quinoline (PMNPQ) and rolipram elevated FLI in brain regions potentially relevant to the anti-depressant and emetic effects of PDE4 inhibition. PMNPQ and rolipram elevated FLI in the locus coeruleus, habenula, paraventricular nucleus of the thalamus, amygdala and nucleus accumbens, all structures with strong limbic connectivity implicated in arousal, memory and affective aspects of behaviour. Consistent with the emetic effects of PDE4 inhibitors such as PMNPQ and rolipram, these compounds elevated FLI in caudal brainstem nuclei such as the area postrema and nucleus of the solitary tract. Administration of the NK(1) antagonist RP 67580 prior to PMNPQ reversed increases in FLI produced by PMNPQ in these regions. RP 67580 did not, however, reduce PMNPQ-induced FLI in limbic structures. These findings suggest that PDE4 inhibitors produce emesis by increasing NK(1) receptor activation in the AP/NTS and implicate brain regions associated with reward and mood such as the amygdala, paraventricular nucleus of the thalamus, habenula and nucleus accumbens in the anti-depressant activity of such compounds.


Subject(s)
Brain/drug effects , Gene Expression/drug effects , Oncogene Proteins v-fos/metabolism , Phosphodiesterase Inhibitors/pharmacology , Analgesics/pharmacology , Animals , Brain/metabolism , Dose-Response Relationship, Drug , Indoles/pharmacology , Isoindoles , Male , Oncogene Proteins v-fos/genetics , Pyridines/pharmacology , Quinolines/pharmacology , Rats , Rats, Sprague-Dawley , Time Factors
11.
Neuroscience ; 141(2): 863-874, 2006 Aug 25.
Article in English | MEDLINE | ID: mdl-16750894

ABSTRACT

Corticocortical disconnection in Alzheimer's disease occurs by the progressive impairment and eventual loss of a small subset of pyramidal neurons in layers III and V of association areas of the neocortex. These neurons exhibit large somatic size, extensive dendritic arborization and high levels of nonphosphorylated neurofilaments of medium and high molecular weight that can be identified using a monoclonal SMI-32 antibody. It is thought that the accumulation of amyloid Abeta and neurofibrillary tangles may provoke metabolic disturbances that result in the loss of these SMI-32 immunoreactive neurons. The recent detection of increased levels of caspase-3 cleaved fodrin in frontal, temporal and parietal association areas in Alzheimer's disease brains suggests that programmed cell death may contribute to the destruction of SMI-32 positive neurons. In the present study, we utilized an antibody that selectively recognizes the 120 kDa breakdown product of alphaIIspectrin (fodrin) generated by caspase-3 to determine whether this protease is activated in vulnerable pyramidal neurons located in layers III and V of Alzheimer's disease brains. Neurons immunoreactive for caspase-3 cleaved alphaIIspectrin were located predominantly in layers III and V of the inferior frontal and superior temporal cortices of patients with Alzheimer's disease but not age-matched controls. Pyramidal neurons immunoreactive for caspase-3 cleaved alphaIIspectrin invariably displayed SMI-32 immunoreactivity suggesting that caspase-3 activation is a pathological event that may be responsible for the loss of a subset of pyramidal neurons that comprise corticocortical projections.


Subject(s)
Alzheimer Disease/metabolism , Caspases/metabolism , Neurofilament Proteins/metabolism , Neurons/metabolism , Spectrin/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Analysis of Variance , Animals , Blotting, Western/methods , Brain/metabolism , Brain/pathology , Caspase 3 , Female , Humans , Immunohistochemistry/methods , Immunoprecipitation/methods , Male , Rats
12.
Cell Death Discov ; 2: 16070, 2016.
Article in English | MEDLINE | ID: mdl-27924223

ABSTRACT

Mitochondrial permeability transition pore (mPTP) opening allows free movement of ions and small molecules leading to mitochondrial membrane depolarization and ATP depletion that triggers cell death. A multi-protein complex of the mitochondrial ATP synthase has an essential role in mPTP. However, the molecular identity of the central 'pore' part of mPTP complex is not known. A highly purified fraction of mammalian mitochondria containing C-subunit of ATPase (C-subunit), calcium, inorganic polyphosphate (polyP) and polyhydroxybutyrate (PHB) forms ion channels with properties that resemble the native mPTP. We demonstrate here that amount of this channel-forming complex dramatically increases in intact mitochondria during mPTP activation. This increase is inhibited by both Cyclosporine A, an inhibitor of mPTP and Ruthenium Red, an inhibitor of the Mitochondrial Calcium Uniporter. Similar increases in the amount of complex formation occurs in areas of mouse brain damaged by ischemia-reperfusion injury. These findings suggest that calcium-induced mPTP is associated with de novo assembly of a channel comprising C-subunit, polyP and PHB.

13.
J Neurosci ; 19(18): 7860-9, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10479688

ABSTRACT

p53 is a pivotal molecule regulating the death of neurons both after acute injury and during development. The molecular mechanisms by which p53 induces apoptosis in neuronal cells, however, are not well understood. We have shown previously that adenovirus-mediated p53 gene delivery to neurons was sufficient to induce apoptosis. In the present study we have examined the molecular mechanism by which p53 evokes neuronal cell death. Adenovirus-mediated delivery of p53 to cerebellar granule neurons resulted in caspase-3 (CPP32) activation followed by terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling (TUNEL) staining and loss of viability as determined by an MTT survival assay. To determine whether Bax is essential for caspase-3 activation, p53 was expressed in Bax-deficient cells. Bax null neurons did not exhibit caspase-3 activation in response to p53 and were protected from apoptosis. To determine whether Bax-dependent caspase-3 activation was required in p53-mediated neuronal cell death, caspase-3-deficient neurons were examined. Our results indicate that caspase-3-deficient neurons exhibit a remarkable delay in apoptosis and a dramatic decrease in TUNEL-positive cells. These studies demonstrate that p53-induced cell death in postmitotic neurons involves a Bax-dependent caspase-3 activation, suggesting that these molecules are important determinants in neuronal cell death after injury.


Subject(s)
Apoptosis , Brain/cytology , Caspases/metabolism , Neurons/cytology , Neurons/physiology , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/physiology , Tumor Suppressor Protein p53/metabolism , Adenoviridae , Animals , Animals, Newborn , Brain/physiology , Caspase 3 , Caspases/genetics , Cells, Cultured , Cerebellum/cytology , Cerebellum/physiology , Genes, p53 , Kinetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein
14.
J Neurosci ; 19(12): 5026-33, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10366635

ABSTRACT

Transient forebrain ischemia produced by four-vessel occlusion (4-VO) triggers the delayed death of CA1 neurons in the hippocampus, resulting in behavioral deficits of spatial learning performance. We demonstrate that CA1 neuronal loss induced by 4-VO (12 min) is preceded by a selective and marked elevation of catalytically active caspase-3 in these neurons, indicative of apoptosis. Virally mediated overexpression of the anti-apoptotic gene X chromosome-linked inhibitor of apoptosis protein (XIAP) prevented both the production of catalytically active caspase-3 and degeneration of CA1 neurons after transient forebrain ischemia. CA1 neurons protected in this manner appeared to function normally, as assessed by immunohistochemical detection of the neuronal activity marker nerve growth factor inducible-A and by spatial learning performance in the Morris water maze. These findings indicate that caspase-3 activation is a key event in ischemic neuronal death and that blockade of this event by XIAP overexpression permits CA1 neurons to survive and operate properly after an ischemic insult.


Subject(s)
Apoptosis/genetics , Hippocampus/cytology , Ischemic Attack, Transient/physiopathology , Nerve Tissue Proteins/genetics , X Chromosome , Animals , Behavior, Animal/physiology , Brain Chemistry/genetics , Caspase 3 , Caspases/metabolism , DNA Fragmentation , Gene Expression Regulation, Enzymologic/physiology , Hippocampus/blood supply , Hippocampus/chemistry , Male , Maze Learning/physiology , Neuronal Apoptosis-Inhibitory Protein , Neurons/chemistry , Neurons/cytology , Neurons/enzymology , Proteins/genetics , Rats , Rats, Wistar , X-Linked Inhibitor of Apoptosis Protein
15.
J Neurosci ; 21(18): 7089-98, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11549719

ABSTRACT

The retinoblastoma tumor suppressor protein, pRb, is a key regulator of cell cycle and has been implicated in the terminal differentiation of neuronal cells. Mice nullizygous for pRb die by embryonic day 14.5 from hematopoietic and neurological defects attributed to failed differentiation (Clarke et al., 1992; Jacks et al., 1992; Lee et al., 1992). Previous studies by MacLeod et al. (1996) have demonstrated that the loss of p53 protects Rb-deficient CNS neurons but not peripheral nervous system (PNS) neurons from cell death. Thus, the mechanisms by which PNS neurons undergo apoptosis in response to Rb deficiency remain unknown. In view of the pivotal role of caspase 3 in the regulation of neuronal apoptosis during development, we examined its function in the execution of the wide-spread neuronal cell death induced by Rb deficiency. Our results support a number of conclusions. First, we show that caspase 3 becomes activated in all neuronal populations undergoing apoptosis. Second, caspase 3 deficiency does not extend the life span of Rb null embryos, because double null mutants exhibit high rates of liver apoptosis resulting in erythropoietic failure. Third, Rb/caspase 3 double-mutant neurons of the CNS exhibit widespread apoptosis similar to that seen in Rb mutants alone; thus caspase 3 deficiency does not protect this population from apoptosis. Finally, in contrast to the CNS, neurons of the PNS including those comprising the trigeminal ganglia and the dorsal root ganglia are protected from apoptosis in Rb/caspase 3 double-mutant embryos. Examination of the mechanistic differences between these two cell types suggest that CNS neurons may invoke other caspases to facilitate apoptosis in the absence of caspase 3. These findings suggest that PNS neurons are dependent on caspase 3 for the execution of apoptosis and that caspase 3 may serve as a key therapeutic target for neuroprotection after injury of this cell type.


Subject(s)
Caspases/deficiency , Peripheral Nervous System/physiopathology , Retinoblastoma Protein/deficiency , Amyloid beta-Protein Precursor/metabolism , Animals , Apoptosis , Caspase 3 , Caspases/biosynthesis , Caspases/genetics , Central Nervous System/cytology , Central Nervous System/embryology , Central Nervous System/metabolism , Crosses, Genetic , Enzyme Induction/physiology , Fluorescent Dyes , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Ganglia, Spinal/metabolism , Gene Expression Regulation, Developmental/physiology , Genotype , In Situ Nick-End Labeling , Mice , Mice, Knockout , Neurons/classification , Neurons/metabolism , Neurons/pathology , Organ Specificity , Peripheral Nervous System/embryology , Peripheral Nervous System/pathology , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/genetics
16.
Neuroscience ; 134(2): 575-82, 2005.
Article in English | MEDLINE | ID: mdl-15939545

ABSTRACT

In the present study, we compared the in vivo neuroprotective efficacy of intraperitoneally administered tetracycline and minocycline to enhance the survival of retinal ganglion cells (RGCs) following unilateral axotomy of the adult rat optic nerve. We also examined the effects of the tetracycline drugs on the activation of retinal microglia. RGCs in retinal whole-mounts were visualized by retrograde labeling with fluorogold. The presence of activated microglia was confirmed immunohistochemically using OX-42 monoclonal antibodies. Optic nerve axotomy produced RGC death and increased activation of microglia. No significant RGC loss was seen prior to 5 days and approximately 50% and 80-90% cell loss occurred at 7 and 14 days, respectively. Examination of the effects of tetracycline and minocycline on RGC survival at 7 days post-axotomy, revealed increased numbers of RGCs in minocycline-treated animals (75% of non-axotomized control) compared with vehicle-only (52% of control) and tetracycline-treated (58% of control) animals. The densities of RGCs (RGCs/mm2+/-S.D.) for control, vehicle-, tetracycline- and minocycline-treated axotomized animals were 1996+/-81, 1029+/-186, 1158+/-190 and 1497+/-312, respectively. The neuroprotective effect of minocycline seen at 7 days was transient, since RGCs present in minocycline-treated animals at 14 days post-axotomy (281+/-43, 14% of control) were not significantly different to vehicle-treated animals (225+/-47, 11% of control). OX-42 staining of activated retinal microglia was reduced in tetracycline- and minocycline-treated axotomized animals compared with axotomized animals receiving vehicle-only. These results demonstrate that systemic administration of the second-generation tetracycline derivative, minocycline, delays the death of axotomized RGCs by a mechanism that may be associated with inhibition of microglia activation. The neuroprotective efficacy of minocycline following optic nerve axotomy was superior to that of tetracycline.


Subject(s)
Axotomy , Cell Survival/drug effects , Minocycline/pharmacology , Retinal Ganglion Cells/cytology , Tetracycline/pharmacology , Animals , Optic Nerve/physiology , Rats , Rats, Long-Evans , Retina/cytology , Retina/drug effects , Retinal Ganglion Cells/drug effects
17.
J Mol Med (Berl) ; 77(1): 130-2, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9930947

ABSTRACT

Islet autotransplantation offers the potential for preventing the surgically induced diabetes that is an inevitable consequence of total pancreatectomy. This paper describes the first islet autotransplant programme in the United Kingdom and the first series in the world to use the spleen as a site for the islet graft. Over an 11 month period, 7 patients underwent total pancreatectomy for chronic pancreatitis combined with a simultaneous islet autotransplant. All 7 patients had normal glucose-tolerance levels and normal C-peptide levels pre-operatively. In 6 patients, islets were embolized into the liver via the portal vein (median transplanted volume=8.5 ml). In addition, 3 patients received islets into the splenic sinusoids via a short gastric vein (median transplanted volume=4 ml). One patient received islets into the spleen alone. One patient died of a stroke 4 weeks post transplantation. Two patients have achieved insulin independence, with a further two patients achieving "transient" insulin independence (<1 month). The remaining 2 patients, although requiring reduced insulin doses, have not achieved insulin-independence. However, all patients have C-peptide levels within the normal range. In trying to explain these findings, split proinsulin levels were measured and found to be elevated. High levels of split proinsulin cross react with the C-peptide assay and this would explain the falsely elevated C-peptide levels. Indeed insulin levels in these patients were all below the normal range. These findings would suggest that the use of C-peptide levels as the "gold standard" for monitoring islet autograft function, may require reappraisal.


Subject(s)
Islets of Langerhans Transplantation , Pancreatectomy , Pancreatitis/surgery , C-Peptide/analysis , Chronic Disease , Diabetes Mellitus, Type 2/prevention & control , Graft Survival , Humans , Insulin/analysis , Proinsulin/analysis , Transplantation, Autologous
18.
Neuroscience ; 308: 75-94, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26363153

ABSTRACT

In view of evidence that increased consumption of epicatechin (E) and quercetin (Q) may reduce the risk of stroke, we have measured the effects of combining E and Q on mitochondrial function and neuronal survival following oxygen-glucose deprivation (OGD). Relative to mouse cortical neuron cultures pretreated (24h) with either E or Q (0.1-10µM), E+Q synergistically attenuated OGD-induced neuronal cell death. E, Q and E+Q (0.3µM) increased spare respiratory capacity but only E+Q (0.3µM) preserved this crucial parameter of neuronal mitochondrial function after OGD. These improvements were accompanied by corresponding increases in cyclic AMP response element binding protein (CREB) phosphorylation and the expression of CREB-target genes that promote neuronal survival (Bcl-2) and mitochondrial biogenesis (PGC-1α). Consistent with these findings, E+Q (0.1 and 1.0µM) elevated mitochondrial gene expression (MT-ND2 and MT-ATP6) to a greater extent than E or Q after OGD. Q (0.3-3.0µM), but not E (3.0µM), elevated cytosolic calcium (Ca(2+)) spikes and the mitochondrial membrane potential. Conversely, E and E+Q (0.1 and 0.3µM), but not Q (0.1 and 0.3µM), activated protein kinase B (Akt). Nitric oxide synthase (NOS) inhibition with L-N(G)-nitroarginine methyl ester (1.0µM) blocked neuroprotection by E (0.3µM) or Q (1.0µM). Oral administration of E+Q (75mg/kg; once daily for 5days) reduced hypoxic-ischemic brain injury. These findings suggest E and Q activate Akt- and Ca(2+)-mediated signaling pathways that converge on NOS and CREB resulting in synergistic improvements in neuronal mitochondrial performance which confer profound protection against ischemic injury.


Subject(s)
Brain Ischemia/drug therapy , Catechin/pharmacology , Mitochondria/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Quercetin/pharmacology , Animals , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Calcium/metabolism , Cell Death/drug effects , Cell Death/physiology , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Cerebral Cortex/physiology , Drug Synergism , Glucose/deficiency , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mice , Mitochondria/physiology , Neurons/pathology , Neurons/physiology , Oxygen Consumption/drug effects , Oxygen Consumption/physiology
19.
Brain Pathol ; 10(2): 283-92, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10764048

ABSTRACT

Accumulating evidence strongly suggests that apoptosis contributes to neuronal cell death in a variety of neurodegenerative contexts. Activation of the cysteine protease caspase-3 appears to be a key event in the execution of apoptosis in the central nervous system (CNS). As a result, mice null for caspase-3 display considerable neuronal expansion usually resulting in death by the second week of life. At present, 14 caspase family members have been identified and subdivided into three subgroups on the basis of preference for specific tetrapeptide motifs using a positional scanning combinatorial substrate library. Caspase-3 is a group II member (2, 3, 7) categorized by an absolute substrate requirement for aspartic acid in the P4 position of the scissile bond. The preferred cleavage motif (DExD) for group II caspases is found in many structural, metabolic and repair proteins essential for cellular homeostasis. Consistent with the proposal that apoptosis plays a central in role human neurodegenerative disease, caspase-3 activation has recently been observed in stroke, spinal cord trauma, head injury and Alzheimer's disease. Indeed, peptide-based caspase inhibitors prevent neuronal loss in animal models of head injury and stroke suggesting that these compounds may be the forerunners of non-peptide small molecules that halt apoptosis processes implicated in these neurodegenerative disorders. A clear link between an hereditary neurodegenerative disorder and failed caspase inhibition has recently been proposed for spinal muscular atrophy (SMA). In severe SMA, the neuronal specific inhibitor of apoptosis (IAP) family member known as NAIP is often dysfunctional due to missense and truncation mutations. IAPs such as NAIP potently block the enzymatic activity of group II caspases (3 and 7) suggesting that NAIP mutations may permit unopposed developmental apoptosis to occur in sensory and motor systems resulting in lethal muscular atrophy. Conversely, adenovirally-mediated overexpression of NAIP or the X-linked IAP called XIAP reduces the loss of CA1 hippocampal neurons following transient forebrain ischemia. Taken together, these findings suggest that anti-apoptotic strategies may some day have utility in the treatment of neurodegenerative disease. The present review will summarize some of the recent evidence suggesting that apoptosis inhibitors may become a practical therapeutic approach for both acute and chronic neurodegenerative conditions.


Subject(s)
Apoptosis/drug effects , Neuroprotective Agents/pharmacology , Animals , Apoptosis/physiology , Brain Ischemia/enzymology , Brain Ischemia/physiopathology , Caspase 3 , Caspases/chemistry , Caspases/metabolism , Enzyme Activation/physiology , Humans , Multigene Family/physiology , Neoplasm Proteins/genetics , Neoplasm Proteins/pharmacology , Neurodegenerative Diseases/drug therapy
20.
Hypertension ; 30(6): 1572-7, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9403585

ABSTRACT

The effects of regular (RNa) or high (HNa) sodium diet for 3, 7, and 14 days on Fra-like immunoreactivity (Fra-LI) in the brains of Wistar-Kyoto rats (WKY) and spontaneously hypertensive rats (SHR) were examined using an antibody that recognizes all known members of the Fos family (Fos, Fos-B, Fra-1, and Fra-2). Two weeks of HNa significantly exacerbated hypertension in SHR but had no effects in WKY. On RNa, compared with WKY, SHR showed higher Fra-LI in the median preoptic nucleus, supraoptic nucleus, both parts of the paraventricular nucleus, nucleus of the solitary tract, and central gray. Fra-LI in the subfornical organ did not differ between the two strains. On RNa, Fra-LI in the anterior hypothalamic area could be detected only in WKY. In osmoregulatory areas, HNa diet increased Fra-LI in both SHR and WKY to comparable extents, but in the median preoptic nucleus, Fra-LI was increased to a greater extent in SHR. HNa produced smaller increases in the subfornical organ of SHR compared with WKY. In both the parvocellular and magnocellular paraventricular nuclei, increases in Fra-LI by HNa were more pronounced in SHR than in WKY. In the anterior hypothalamic area, Fra-LI could no longer be detected in WKY on HNa, whereas it appeared in SHR. HNa increased Fra-LI in the NTS and central gray to similar levels in WKY and SHR. These results indicate that WKY and SHR differ in the pattern of neuronal activation accompanying maturation on RNa. HNa activates neurons in a number of brain areas, and the pattern of these changes also differs between WKY and SHR.


Subject(s)
Brain/physiopathology , Hypertension/physiopathology , Neurons/physiology , Proto-Oncogene Proteins c-fos/biosynthesis , Sodium, Dietary/pharmacology , Animals , Blood Pressure/drug effects , Body Weight , Brain/drug effects , Brain/physiology , Male , Neurons/drug effects , Organ Specificity , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Reference Values , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL