Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
2.
AJNR Am J Neuroradiol ; 36(10): 1803-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26045577

ABSTRACT

Preventive neuroradiology is a new concept supported by growing literature. The main rationale of preventive neuroradiology is the application of multimodal brain imaging toward early and subclinical detection of brain disease and subsequent preventive actions through identification of modifiable risk factors. An insightful example of this is in the area of age-related cognitive decline, mild cognitive impairment, and dementia with potentially modifiable risk factors such as obesity, diet, sleep, hypertension, diabetes, depression, supplementation, smoking, and physical activity. In studying this link between lifestyle and cognitive decline, brain imaging markers may be instrumental as quantitative measures or even indicators of early disease. The purpose of this article is to provide an overview of the major studies reflecting how lifestyle factors affect the brain and cognition aging. In this hot topics review, we will specifically focus on obesity and physical activity.


Subject(s)
Alzheimer Disease/diagnosis , Alzheimer Disease/prevention & control , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/prevention & control , Diagnostic Imaging , Age Factors , Aged , Aging , Alzheimer Disease/etiology , Brain , Cognitive Dysfunction/etiology , Depression/complications , Diabetes Complications/diagnosis , Humans , Hypertension/complications , Life Style , Research , Risk Factors
3.
Gene Ther ; 18(12): 1139-49, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22158031

ABSTRACT

Huntington's disease (HD) is caused by an expansion of CAG triplets at the 5' end of the HD gene, which encodes a pathologically elongated polyglutamine stretch near the N-terminus of huntingtin. HD is an incurable autosomal-dominant neurodegenerative disease characterized by movement disorder, as well as emotional distress and dementia. The newly discovered roles of the non-coding small RNAs in specific degradation or translational suppression of the targeted mRNAs suggest a potential therapeutic approach of post-transcriptional gene silencing that targets the underlying disease etiology rather than the downstream pathological consequences. From pre-clinical trials in different HD animal models to cells from HD patients, small RNA interference has been applied to 'allele-non-specifically or allele-specifically' silence the mutant HD transgene or endogenous mutant HD allele. Silencing the mutant HD transgene significantly inhibits neurodegeneration, improves motor control, and extends survival of HD mice. With future improvement of mutant allele selectivity (preserving the expression of the neuroprotective wild-type allele), target specificity, efficacy and safety, as well as optimization of delivery methods, small non-coding RNA-based therapeutic applications will be a promising approach to treat HD.


Subject(s)
RNA Interference , RNA, Small Untranslated/therapeutic use , Alleles , Animals , Drug Delivery Systems , Humans , Huntingtin Protein , Huntington Disease/genetics , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , RNA, Small Interfering/therapeutic use
4.
Neurosurgery ; 48(6): 1393-9; discussion 1399-401, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11383749

ABSTRACT

OBJECTIVE: Caspase-1 plays an important functional role mediating neuronal cell death and dysfunction after experimental traumatic brain injury (TBI) in mice. Minocycline, a derivative of the antibiotic tetracycline, inhibits caspase-1 expression. This study investigates whether minocycline can ameliorate TBI-mediated injury in mice. METHODS: Brains from mice subjected to traumatic brain injury underwent immunohistochemical analyses for caspase-1, caspase-3, and a neuronal specific marker (NeuN). Minocycline- and saline-treated mice subjected to traumatic brain injury were compared with respect to neurological function, lesion volume, and interleukin-1beta production. RESULTS: Immunohistochemical analysis revealed that activated caspase-1 and caspase-3 are present in neurons 24 hours after TBI. Intraperitoneal administration of minocycline 12 hours before or 30 minutes after TBI in mice resulted in improved neurological function when compared with mice given saline control, as assessed by Rotarod performance 1 to 4 days after TBI. The lesion volume, assessed 4 days after trauma, was significantly decreased in mice treated with minocycline before or after trauma when compared with saline-treated mice. Caspase-1 activity, quantified by measuring mature interleukin-1beta production by enzyme-linked immunosorbent assay, was considerably increased in mice that underwent TBI, and this increase was significantly diminished in minocycline-treated mice. CONCLUSION: We show for the first time that caspase-1 and caspase-3 activities localize specifically within neurons after experimental brain trauma. Further, these results indicate that minocycline is an effective pharmacological agent for reducing tissue injury and neurological deficits that result from experimental TBI, likely through a caspase-1-dependent mechanism. These results provide an experimental rationale for the evaluation of minocycline in human trauma patients.


Subject(s)
Brain Injuries/enzymology , Brain Injuries/pathology , Brain/pathology , Caspase 1/metabolism , Enzyme Inhibitors/pharmacology , Minocycline/pharmacology , Nervous System/physiopathology , Animals , Brain/drug effects , Enzyme Activation/drug effects , Interleukin-1/antagonists & inhibitors , Interleukin-1/biosynthesis , Mice , Mice, Inbred C57BL , Nervous System/drug effects , Neurons/enzymology
5.
Science ; 293(5529): 493-8, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11408619

ABSTRACT

Huntingtin is a 350-kilodalton protein of unknown function that is mutated in Huntington's disease (HD), a neurodegenerative disorder. The mutant protein is presumed to acquire a toxic gain of function that is detrimental to striatal neurons in the brain. However, loss of a beneficial activity of wild-type huntingtin may also cause the death of striatal neurons. Here we demonstrate that wild-type huntingtin up-regulates transcription of brain-derived neurotrophic factor (BDNF), a pro-survival factor produced by cortical neurons that is necessary for survival of striatal neurons in the brain. We show that this beneficial activity of huntingtin is lost when the protein becomes mutated, resulting in decreased production of cortical BDNF. This leads to insufficient neurotrophic support for striatal neurons, which then die. Restoring wild-type huntingtin activity and increasing BDNF production may be therapeutic approaches for treating HD.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Huntington Disease/genetics , Nerve Tissue Proteins/physiology , Neurons/metabolism , Nuclear Proteins/physiology , 3T3 Cells , Animals , Apoptosis , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/metabolism , Cell Survival , Cells, Cultured , Cerebral Cortex/cytology , Corpus Striatum/cytology , Corpus Striatum/pathology , Exons , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/pathology , Mice , Mice, Transgenic , Mutation , Nerve Degeneration , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/genetics , Neurons/pathology , Nuclear Proteins/genetics , Promoter Regions, Genetic , Transcription, Genetic , Transfection
6.
Neuroscientist ; 7(6): 480-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11765125

ABSTRACT

Huntington's disease (HD) is an autosomal dominant condition, resulting from a mutation in huntingtin (htt). Htt is a novel protein, and its normal function is at present not well understood. Nuclear translocation of mutant htt in vitro up-regulates expression of the cell death gene caspase-1. We have demonstrated in a transgenic HD mouse model that caspase-1 and caspase-3 are transcriptionally up-regulated and activated. Underscoring the relevancy of these findings, recent results suggest that caspase-1 is activated in brains of humans with HD. Caspase activation results in the proteolytic cleavage of key cellular targets, including htt, leading to cell dysfunction. Caspase activation leading to cell dysfunction and death correlates with disease progression. In HD-transgenic mice, caspase inhibition resulted in a delayed onset of symptoms, a slowed progression, and prolonged survival. Caspase inhibition is a therapeutic strategy that merits evaluation in humans with HD.


Subject(s)
Caspases/metabolism , Huntington Disease/enzymology , Animals , Apoptosis , Enzyme Activation/physiology , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/physiopathology , Inflammation/physiopathology , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics
9.
Neuroscience ; 99(2): 333-42, 2000.
Article in English | MEDLINE | ID: mdl-10938439

ABSTRACT

Evidence indicates that both necrotic and apoptotic cell death contribute to tissue injury and neurological dysfunction following spinal cord injury. Caspases have been implicated as important mediators of apoptosis following acute central nervous system insults. We investigated whether caspase-1 and caspase-3 are involved in spinal cord injury-mediated cell death, and whether caspase inhibition may reduce tissue damage and improve outcome following spinal cord injury. We demonstrate a 17-fold increase in caspase-1 activity in traumatized spinal cord samples when compared with samples from sham-operated mice. Caspase-1 and caspase-3 activation were also detected by western blot following spinal cord injury, which was significantly inhibited by the broad caspase inhibitor N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone. By immunofluorescence or in situ fluorogenic substrate assay, caspase-1 and caspase-3 expression were detected in neuronal and non-neuronal cells following spinal cord injury. N-Benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone treated mice, and transgenic mice expressing a caspase-1 dominant negative mutant, demonstrated a significant improvement of motor function and a reduction of lesion size compared with vehicle-treated mice. Our results demonstrate for the first time that both caspase-1 and caspase-3 are activated in neurons following spinal cord injury, and that caspase inhibition reduces post-traumatic lesion size and improves motor performance. Caspase inhibitors may be one of the agents to be used for the treatment of spinal cord injury.


Subject(s)
Apoptosis/physiology , Caspase 1/metabolism , Caspases/metabolism , Neurons/metabolism , Spinal Cord Injuries/enzymology , Amino Acid Chloromethyl Ketones/pharmacology , Amino Acid Chloromethyl Ketones/therapeutic use , Animals , Apoptosis/drug effects , Caspase 3 , Caspase Inhibitors , Enzyme Activation , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/physiopathology
10.
J Neurochem ; 75(2): 847-52, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10899963

ABSTRACT

Increasing evidence implicates caspase-1-mediated cell death as a major mechanism of neuronal death in neurodegenerative diseases. In the present study we investigated the role of caspase-1 in neurotoxic experimental animal models of Huntington's disease (HD) by examining whether transgenic mice expressing a caspase-1 dominant-negative mutant are resistant to malonate and 3-nitropropionic acid (3-NP) neurotoxicity. Intrastriatal injection of malonate resulted in significantly smaller striatal lesions in mutant caspase-1 mice than those observed in littermate control mice. Caspase-1 was significantly activated following malonate intrastriatal administration in control mice but significantly attenuated in mutant caspase-1 mice. Systemic 3-NP treatment induced selective striatal lesions that were significantly smaller within mutant caspase-1 mice than in littermate control mice. These results provide further evidence of a functional role for caspase-1 in both malonate- and 3-NP-mediated neurotoxin models of HD.


Subject(s)
Brain/pathology , Caspase 1/genetics , Caspase 1/metabolism , Malonates/toxicity , Neurotoxins/toxicity , Propionates/toxicity , Animals , Brain/drug effects , Brain/enzymology , Crosses, Genetic , Disease Models, Animal , Female , Huntington Disease/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neostriatum/drug effects , Neostriatum/pathology , Nitro Compounds , Point Mutation
11.
Nat Med ; 6(7): 797-801, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10888929

ABSTRACT

Huntington disease is an autosomal dominant neurodegenerative disease with no effective treatment. Minocycline is a tetracycline derivative with proven safety. After ischemia, minocycline inhibits caspase-1 and inducible nitric oxide synthetase upregulation, and reduces infarction. As caspase-1 and nitric oxide seem to play a role in Huntington disease, we evaluated the therapeutic efficacy of minocycline in the R6/2 mouse model of Huntington disease. We report that minocycline delays disease progression, inhibits caspase-1 and caspase-3 mRNA upregulation, and decreases inducible nitric oxide synthetase activity. In addition, effective pharmacotherapy in R6/2 mice requires caspase-1 and caspase-3 inhibition. This is the first demonstration of caspase-1 and caspase-3 transcriptional regulation in a Huntington disease model.


Subject(s)
Caspase 1/biosynthesis , Caspases/biosynthesis , Huntington Disease/drug therapy , Minocycline/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Anti-Bacterial Agents/therapeutic use , Caspase 3 , Disease Models, Animal , Disease Progression , Enzyme Activation/drug effects , Evaluation Studies as Topic , Gene Expression Regulation , Huntington Disease/mortality , Mice , Mice, Transgenic , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase Type II , Transcription, Genetic
12.
Nature ; 405(6784): 360-4, 2000 May 18.
Article in English | MEDLINE | ID: mdl-10830966

ABSTRACT

Cyclin-dependent kinase 5 (cdk5) and its neuron-specific activator p35 are required for neurite outgrowth and cortical lamination. Proteolytic cleavage of p35 produces p25, which accumulates in the brains of patients with Alzheimer's disease. Conversion of p35 to p25 causes prolonged activation and mislocalization of cdk5. Consequently, the p25/cdk5 kinase hyperphosphorylates tau, disrupts the cytoskeleton and promotes the death (apoptosis) of primary neurons. Here we describe the mechanism of conversion of p35 to p25. In cultured primary cortical neurons, excitotoxins, hypoxic stress and calcium influx induce the production of p25. In fresh brain lysates, addition of calcium can stimulate cleavage of p35 to p25. Specific inhibitors of calpain, a calcium-dependent cysteine protease, effectively inhibit the calcium-induced cleavage of p35. In vitro, calpain directly cleaves p35 to release a fragment with relative molecular mass 25,000. The sequence of the calpain cleavage product corresponds precisely to that of p25. Application of the amyloid beta-peptide A beta(1-42) induces the conversion of p35 to p25 in primary cortical neurons. Furthermore, inhibition of cdk5 or calpain activity reduces cell death in A beta-treated cortical neurons. These observations indicate that cleavage of p35 to p25 by calpain may be involved in the pathogenesis of Alzheimer's disease.


Subject(s)
Calpain/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Alzheimer Disease/metabolism , Amino Acid Sequence , Amyloid beta-Peptides/metabolism , Animals , Calcium/metabolism , Calpain/antagonists & inhibitors , Cells, Cultured , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Glutamic Acid/pharmacology , Hydrogen Peroxide/pharmacology , Ionomycin/pharmacology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neurons/drug effects , Peptide Fragments/metabolism , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Recombinant Proteins
13.
Science ; 288(5464): 335-9, 2000 Apr 14.
Article in English | MEDLINE | ID: mdl-10764647

ABSTRACT

Mutations in the copper/zinc superoxide dismutase (SOD1) gene produce an animal model of familial amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disorder. To test a new therapeutic strategy for ALS, we examined the effect of caspase inhibition in transgenic mice expressing mutant human SOD1 with a substitution of glycine to alanine in position 93 (mSOD1(G93A)). Intracerebroventricular administration of zVAD-fmk, a broad caspase inhibitor, delays disease onset and mortality. Moreover, zVAD-fmk inhibits caspase-1 activity as well as caspase-1 and caspase-3 mRNA up-regulation, providing evidence for a non-cell-autonomous pathway regulating caspase expression. Caspases play an instrumental role in neurodegeneration in transgenic mSOD1(G93A) mice, which suggests that caspase inhibition may have a protective role in ALS.


Subject(s)
Amino Acid Chloromethyl Ketones/pharmacology , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/enzymology , Caspase 1/metabolism , Caspases/metabolism , Motor Neurons/drug effects , Neuroprotective Agents/pharmacology , Amino Acid Chloromethyl Ketones/administration & dosage , Amino Acid Chloromethyl Ketones/therapeutic use , Amino Acid Substitution , Amyotrophic Lateral Sclerosis/pathology , Animals , Apoptosis/drug effects , Caspase 1/genetics , Caspase 3 , Caspase Inhibitors , Caspases/genetics , Cysteine Proteinase Inhibitors/administration & dosage , Cysteine Proteinase Inhibitors/pharmacology , Cysteine Proteinase Inhibitors/therapeutic use , Disease Models, Animal , Disease Progression , Enzyme Activation , Gene Expression Regulation, Enzymologic , Humans , Injections, Intraventricular , Interleukin-1/metabolism , Male , Mice , Mice, Transgenic , Motor Neurons/enzymology , Motor Neurons/pathology , Nerve Degeneration , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Psychomotor Performance , Spinal Cord/enzymology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
14.
J Neurosurg ; 92(1): 165-9, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10616097

ABSTRACT

Solitary peripheral nerve lymphomas are exceedingly rare primary manifestations of diffuse peripheral nervous system or central nervous system (CNS) lymphomatosis. A 52-year-old man presented with progressive weakness in gastrocnemius and anterior tibial muscle function, which was associated with radiating pain in the right leg. Magnetic resonance imaging studies revealed a solitary fusiform tumor, extending from the sciatic nerve, at the level of the lesser trochanter of the femur, into the posterior tibial nerve below the popliteal fossa. Intraoperative gross examination found that the tumor diffusely expanded the nerve, but did not extend from or into surrounding muscle or tendons. The final histological diagnosis was a solitary extranodal lymphoma (Burkittlike high-grade B-cell lymphoma). Postoperative staging did not reveal evidence of lymphomatous involvement of other organs, but additional chemo- and radiotherapies were administered. Four months after the surgical biopsy, the patient presented with a right facial nerve palsy. The results of cytological examination of cerebrospinal fluid were positive for the presence of atypical lymphocytes, which was consistent with apparently progressive neurolymphomatosis; however, the results of radiological studies were negative for systemic progression. The patient underwent intrathecal chemotherapy followed by systemic myelosuppressive chemotherapy with bone marrow rescue, but died of respiratory failure while still receiving treatment. Postmortem examination revealed extensive lymphomatosis in the peripheral nerves and spinal nerve roots without evidence of cranial nerve, CNS, or other organ system involvement. The aggressive biological characteristics of these tumors, their management, and pertinent literature are reviewed.


Subject(s)
Lymphoma/diagnosis , Peripheral Nervous System Neoplasms/diagnosis , Sciatic Nerve , Diagnosis, Differential , Fatal Outcome , Humans , Lymphoma/pathology , Lymphoma/therapy , Magnetic Resonance Imaging , Male , Middle Aged , Peripheral Nervous System Neoplasms/pathology , Peripheral Nervous System Neoplasms/therapy , Sciatic Nerve/pathology
16.
Apoptosis ; 5(2): 117-32, 2000 Apr.
Article in English | MEDLINE | ID: mdl-11232240

ABSTRACT

The caspase family proteases are principal components of the apoptotic pathway. In this study we demonstrate that caspase-1-like proteases and interleukin-1 beta are important for death induced by various stimuli in cell lines, primary fibroblasts and primary sensory neurons. Furthermore, we show by immunohistochemistry that during the cell death process endogenous caspase-1-like proteases translocate into the nucleus. This translocation is stimulated by interleukin-1 receptor activation. Translocation of caspase-1-like proteases and cell death can be partially prevented by blocking the interleukin-1 receptor with the interleukin-1 receptor antagonist. This finding offers for the first time a mechanistic explanation for the protective effect of the interleukin-1 receptor antagonist against cell death. Furthermore, our data suggest that caspase-1-like proteases have a function in the nucleus which is necessary for completion of the cell death program. In cultured DRG neurons from embryonic mice the combined inhibition of caspases and the interleukin-1 receptor have an additive effect and fully prevent semaphorin III-induced neuronal death. This shows that endogenous caspases work together with IL-1 beta in Semaphorin III-induced neuronal death. We hypothesize that the cell death process involves a double activation step, probably including an interleukin-1 autocrine loop. This model can explain our finding that combined inhibition of caspases and interleukin-1 receptor is necessary to strongly inhibit the cell death process.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Cell Nucleus/metabolism , Interleukin-1/metabolism , Semaphorin-3A , Active Transport, Cell Nucleus , Animals , Apoptosis/drug effects , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Fractionation , Cells, Cultured , Fibroblasts , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Precipitin Tests , Receptors, Interleukin-1/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Tumor Necrosis Factor-alpha/pharmacology
17.
J Vasc Surg ; 30(4): 752-60, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10514215

ABSTRACT

PURPOSE: Apoptosis is a stereotypical pathway of cell death that is orchestrated by a family of cysteine endoproteases called caspases. This study examined the effect of apoptosis inhibition with a specific caspase inhibitor on murine intestinal viability after ischemia-reperfusion (IR). METHODS: C57Bl6 X SV129 mice underwent segmental small bowel ischemia by vascular isolation of 10 cm of terminal ileum. In separate experiments, the ischemic time was varied from 30 to 130 minutes with a reperfusion interval of 6 hours. The degree of small bowel injury was quantified from 1 to 5 (increasing severity) by standardized, blinded histologic grading. The degree of apoptosis was assessed with a specific assay (terminal deoxyamcleotydil transferase-mediated deoxyuridine triphosphate nick end labeling [TUNEL]) and quantified by calculating the apoptotic index (apoptotic cells/10 high-power fields). To evaluate for activation of interleukin-1beta converting enzyme we measured tissue mature interleukin-1beta levels using a specific enzyme-linked immunosorbent assay. To evaluate the effect of apoptosis inhibition on intestinal viability after IR, mice received 3.0 mg of the caspase inhibitor ZVAD (N-benzyloxycarbonyl Val-Ala-Asp-Ome-fluoromethylketone) subcutaneously before and after IR in five divided doses (n = 11), the same dose of ZFA (N-benzyloxycarbonyl Phe-Ala fluoromethylketone), a structurally similar molecule with no anticaspase activity (n = 9), or sham operation (n = 6). RESULTS: A linear relationship existed between ischemic interval and histologic grade (r = 0.69, P <.006). Increasing the ischemic interval from 0 to 50 minutes was associated with a fivefold increase in apoptotic index (P =.05). Ischemic bowel was measured to have an average of 57.3 +/- 7.8 pg/mL whereas normal bowel had an average of 1.8 +/- 0.5 pg/mL of mature interleukin-1beta present. Mice tolerated multiple injections of ZVAD and ZFA without signs of toxicity. Animals treated with ZVAD (apoptosis inhibitor) had little injury after 50 minutes of ischemia and 6 hours of reperfusion (injury grade 1.8) compared with sham controls (injury grade 1.2, P =.7) and had significantly less injury than mice treated with ZFA (placebo) (injury grade 3.0, P <.006). CONCLUSIONS: Increasing ischemic interval in a segmental small bowel murine IR model is associated with increased histologic injury and augmented apoptosis as evidenced by increased TUNEL staining and interleukin-1beta converting enzyme activation. Inhibition of apoptosis with a specific caspase inhibitor significantly diminishes the degree of small bowel injury.


Subject(s)
Apoptosis/drug effects , Caspase Inhibitors , Enzyme Inhibitors/pharmacology , Intestine, Small/blood supply , Reperfusion Injury/physiopathology , Animals , Cell Survival , In Situ Nick-End Labeling , Interleukin-1/analysis , Mice , Mice, Inbred Strains , Reperfusion Injury/pathology
19.
Nature ; 399(6733): 263-7, 1999 May 20.
Article in English | MEDLINE | ID: mdl-10353249

ABSTRACT

Huntington's disease is an autosomal-dominant progressive neurodegenerative disorder resulting in specific neuronal loss and dysfunction in the striatum and cortex. The disease is universally fatal, with a mean survival following onset of 15-20 years and, at present, there is no effective treatment. The mutation in patients with Huntington's disease is an expanded CAG/polyglutamine repeat in huntingtin, a protein of unknown function with a relative molecular mass of 350,000 (M(r) 350K). The length of the CAG/polyglutamine repeat is inversely correlated with the age of disease onset. The molecular pathways mediating the neuropathology of Huntington's disease are poorly understood. Transgenic mice expressing exon 1 of the human huntingtin gene with an expanded CAG/polyglutamine repeat develop a progressive syndrome with many of the characteristics of human Huntington's disease. Here we demonstrate evidence of caspase-1 activation in the brains of mice and humans with the disease. In this transgenic mouse model of Huntington's disease, expression of a dominant-negative caspase-1 mutant extends survival and delays the appearance of neuronal inclusions, neurotransmitter receptor alterations and onset of symptoms, indicating that caspase-1 is important in the pathogenesis of the disease. In addition, we demonstrate that intracerebroventricular administration of a caspase inhibitor delays disease progression and mortality in the mouse model of Huntington's disease.


Subject(s)
Caspase Inhibitors , Enzyme Inhibitors/therapeutic use , Huntington Disease/enzymology , Animals , Brain/enzymology , Caspase 1/genetics , Disease Progression , Enzyme Activation , Female , Huntingtin Protein , Huntington Disease/drug therapy , Huntington Disease/genetics , Huntington Disease/pathology , Injections, Intraventricular , Interleukin-1/metabolism , Male , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Weight Loss
20.
Neuroreport ; 10(3): 635-8, 1999 Feb 25.
Article in English | MEDLINE | ID: mdl-10208603

ABSTRACT

Increasing evidence implicates apoptosis as a major mechanism of cell death in neurodegenerative diseases. Recent evidence has demonstrated that chronic administration of MPTP can lead to apoptotic cell death. In the present study we examined whether transgenic mice expressing a dominant negative inhibitor of interleukin-1beta convertase enzyme (ICE) are resistant to MPTP induced neurotoxicity. MPTP resulted in a significant depletion of dopamine, DOPAC and HVA in littermate control mice which were completely inhibited in the mutant interleukin-1beta converting enzyme mice. There was also significant protection against MPTP-induced depletion of tyrosine hydroxylase-immunoreactive neurons. There was no alteration in MPTP uptake or metabolism. These results provide further evidence that apoptotic cell death as well as ICE may play an important role in the neurotoxicity of MPTP.


Subject(s)
Caspase 1/genetics , Gene Expression/physiology , Genes, Dominant , MPTP Poisoning , Mice, Transgenic/genetics , Mutation/genetics , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dopamine/metabolism , Drug Resistance/genetics , Homovanillic Acid/metabolism , Mice , Reference Values
SELECTION OF CITATIONS
SEARCH DETAIL
...