Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Ann Oncol ; 28(5): 1098-1104, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28453693

ABSTRACT

Background: In 2012, the United States Preventive Services Task Force (USPSTF) recommended against prostate-specific antigen (PSA) screening, despite evidence that Black men are at a higher risk of prostate cancer-specific mortality (PCSM). We evaluated whether Black men of potentially screening-eligible age (55-69 years) are at a disproportionally high risk of poor outcomes. Patients and methods: The SEER database was used to study 390 259 men diagnosed with prostate cancer in the United States between 2004 and 2011. Multivariable logistic regression modeled the association between Black race and stage of presentation, while Fine-Gray competing risks regression modeled the association between Black race and PCSM, both as a function of screening eligibility (age 55-69 years versus not). Results: Black men were more likely to present with metastatic disease (adjusted odds ratio [AOR] 1.65; 1.58-1.72; P < 0.001) and were at a higher risk of PCSM (adjusted hazard ratio [AHR] 1.36; 1.27-1.46; P < 0.001) compared to non-Black men. There were significant interactions between race and PSA-screening eligibility such that Black patients experienced more disproportionate rates of metastatic disease (AOR 1.76; 1.65-1.87 versus 1.55; 1.47-1.65; Pinteraction < 0.001) and PCSM (AHR 1.53; 1.37-1.70 versus 1.25; 1.14-1.37; Pinteraction = 0.01) in the potentially PSA-screening eligible group than in the group not eligible for screening. Conclusions: Racial disparities in prostate cancer outcome among Black men are significantly worse in PSA-screening eligible populations. These results raise the possibility that Black men could be disproportionately impacted by recommendations to end PSA screening in the United States and suggest that Black race should be included in the updated USPSTF PSA screening guidelines.


Subject(s)
Prostatic Neoplasms/diagnosis , Black or African American , Aged , Early Detection of Cancer , Healthcare Disparities , Humans , Kallikreins/metabolism , Male , Middle Aged , Proportional Hazards Models , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/mortality , Prostatic Neoplasms/therapy , Risk Factors , SEER Program , Treatment Outcome , United States/epidemiology
2.
Neurobiol Dis ; 8(6): 1017-26, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11741397

ABSTRACT

Huntington's disease (HD) is a late onset neurodegenerative disorder caused by a CAG/polyglutamine (polyQ) repeat expansion. PolyQ aggregates can be detected in the nuclei and processes of neurons in HD patients and mouse models prior to the onset of symptoms. The misfolding and aggregation pathway is an important therapeutic target. To better test the efficacy of aggregation inhibitors, we have developed an organotypic slice culture system. We show here that the formation of polyQ aggregates in hippocampal slices established from the R6/2 mouse follows the same prescribed sequence as occurs in vivo. Using this assay, we show that Congo red and chrysamine G can modulate aggregate formation, but show complex dose-response curves. Oral administration of creatine has been shown to delay the onset of all aspects of the phenotype and neuropathology in R6/2 mice. We show here that creatine can similarly inhibit aggregate formation in the slice culture assay.


Subject(s)
Hippocampus/drug effects , Huntington Disease/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Peptides/drug effects , Protein Folding , Trinucleotide Repeat Expansion/drug effects , Animals , Benzoates/pharmacology , Biphenyl Compounds/pharmacology , Cells, Cultured , Coloring Agents/pharmacology , Congo Red/pharmacology , Creatine/pharmacology , Cysteine Endopeptidases/drug effects , Cysteine Endopeptidases/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Energy Metabolism/drug effects , Energy Metabolism/physiology , Female , Hippocampus/metabolism , Hippocampus/pathology , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Immunohistochemistry , Male , Mice , Mice, Transgenic , Multienzyme Complexes/drug effects , Multienzyme Complexes/metabolism , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Nuclear Proteins/drug effects , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organ Culture Techniques , Peptides/genetics , Peptides/metabolism , Proteasome Endopeptidase Complex , Trinucleotide Repeat Expansion/genetics , Ubiquitin/drug effects , Ubiquitin/genetics , Ubiquitin/metabolism
3.
Hum Mol Genet ; 10(21): 2425-35, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11689489

ABSTRACT

Huntington's disease (HD) is a progressive neurological disorder caused by a CAG/polyglutamine repeat expansion. We have previously generated the R6/2 mouse model that expresses exon 1 of the human HD gene containing CAG repeats in excess of 150. These mice develop a progressive neurological phenotype with a rapid onset and progression. We show here that it is impossible to establish fibroblast lines from these mice at 12 weeks of age, whilst this can be achieved without difficulty at 6 and 9 weeks. Cultures derived from mice at 12 weeks contained a high frequency of dysmorphic cells, including cells with an aberrant nuclear morphology and a high frequency of micronuclei and large vacuoles. All of these features were also present in a line derived from a juvenile HD patient. Fibroblast lines derived from R6/2 mice and from HD patients were found to have a high frequency of multiple centrosomes which could account for all of the observed phenotypes including a reduced mitotic index, high frequency of aneuploidy and persistence of the midbody. We were unable to detect large insoluble polyglutamine aggregates in either the mouse or human lines. We have identified a novel progressive HD pathology that occurs in cells of non-central nervous system origin. An investigation of the pathological consequences of the HD mutation in these cells will provide insight into cellular basis of the disease.


Subject(s)
Centrosome/metabolism , Fibroblasts/metabolism , Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Aneuploidy , Animals , Blotting, Western , Brain/metabolism , Cell Line , Cell Nucleus/metabolism , Cellular Senescence/genetics , Cytoplasm/metabolism , Cytoskeleton/metabolism , DNA Replication/genetics , Endocytosis , Endosomes/metabolism , Female , Fibroblasts/cytology , Humans , Huntingtin Protein , Huntington Disease/pathology , Lysosomes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Microscopy, Fluorescence , Mitotic Index , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Trinucleotide Repeat Expansion/genetics , Trinucleotide Repeats/genetics
4.
Neurobiol Dis ; 8(5): 807-21, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11592850

ABSTRACT

Huntington's disease (HD) is a late-onset neurodegenerative disease for which the mutation is CAG/polyglutamine repeat expansion. The R6 mouse lines expressing the HD mutation develop a movement disorder that is preceded by the formation of neuronal polyglutamine aggregates. The phenotype is likely caused by a widespread neuronal dysfunction, whereas neuronal cell death occurs late and is very selective. We show that a decreased mRNA level of the major astroglial glutamate transporter (GLT1) in the striatum and cortex of these mice is accompanied by a concomitant decrease in glutamate uptake. In contrast, the expression of the glutamate transporters, GLAST and EAAC1, remain unchanged. The mRNA level of the astroglial enzyme glutamine synthetase is also decreased. These changes in expression occur prior to any evidence of neurodegeneration and suggest that a defect in astrocytic glutamate uptake may contribute to the phenotype and neuronal cell death in HD.


Subject(s)
Astrocytes/metabolism , Glutamic Acid/pharmacokinetics , Huntington Disease/metabolism , Nerve Tissue Proteins/metabolism , Symporters , Amino Acid Transport System X-AG/biosynthesis , Amino Acid Transport System X-AG/genetics , Animals , Aspartic Acid/metabolism , Biological Transport , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Crosses, Genetic , Disease Models, Animal , Excitatory Amino Acid Transporter 1 , Excitatory Amino Acid Transporter 2/deficiency , Excitatory Amino Acid Transporter 3 , Glial Fibrillary Acidic Protein/analysis , Glutamate Plasma Membrane Transport Proteins , Glutamate-Ammonia Ligase/biosynthesis , Glutamate-Ammonia Ligase/deficiency , Glutamate-Ammonia Ligase/genetics , Humans , Huntingtin Protein , Huntington Disease/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Models, Neurological , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nuclear Proteins/analysis , Peptides/analysis , Phenotype , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
5.
Nutr Rev ; 59(12): 379-90, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11766908

ABSTRACT

Undernutrition is being rapidly reduced in India and China. In both countries the diet is shifting toward higher fat and lower carbohydrate content. Distinct features are high intakes of foods from animal sources and edible oils in China, and high intakes of dairy and added sugar in India. The proportion of overweight is increasing very rapidly in China among all adults; in India the shift is most pronounced among urban residents and high-income rural residents. Hypertension and stroke are relatively higher in China and adult-onset diabetes is relatively higher in India. Established economic techniques were used to measure and project the costs of undernutrition and diet-related noncommunicable diseases in 1995 and 2025. Current WHO mortality projections of diet-related noncommunicable diseases, dietary and body composition survey data, and national data sets of hospital costs for healthcare, are used for the economic analyses. In 1995, China's costs of undernutrition and costs of diet-related noncommunicable diseases were of similar magnitude, but there will be a rapid increase in the costs and prevalence of diet-related noncommunicable diseases by 2025. By contrast with China, India's costs of undernutrition will continue to decline, but undernutrition costs did surpass overnutrition diet-related noncommunicable disease costs in 1995. India's rapid increase in diet-related noncommunicable diseases and their costs projects similar economic costs of undernutrition and overnutrition by 2025.


Subject(s)
Diet/trends , Nutrition Disorders/epidemiology , Obesity/epidemiology , China/epidemiology , Humans , India/epidemiology , Nutrition Disorders/economics , Nutritional Status , Obesity/economics , Rural Health , Urban Health
6.
J Neurosci ; 20(13): 5115-23, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10864968

ABSTRACT

Huntington's disease (HD) is an autosomal dominant progressive and fatal neurodegenerative brain disorder caused by an expanded CAG/polyglutamine repeat in the coding region of the gene. Presymptomatic Huntington's disease patients often exhibit cognitive deficits before the onset of classical symptoms. To investigate the possibility that changes in synaptic plasticity might underlie cognitive impairment in HD, we examined hippocampal synaptic plasticity and spatial cognition in a transgenic mouse (R6/2 line) expressing exon 1 of the human Huntington's disease gene containing an expanded CAG repeat. This mouse exhibits a progressive and fatal neurological phenotype that resembles Huntington's disease. We report that R6/2 mice show marked alterations in synaptic plasticity at both CA1 and dentate granule cell synapses, and impaired spatial cognitive performance in the Morris water maze. The changes in hippocampal plasticity were age dependent, appearing at CA1 synapses several weeks before they were observed in the dentate gyrus. Deficits in synaptic plasticity at CA1 synapses occurred before an overt phenotype. This suggests that altered synaptic plasticity contributes to the pre-symptomatic changes in cognition reported in human carriers of the Huntington' disease gene. The temporal and regional changes in synaptic plasticity within the hippocampus mirror the appearance of neuronal intranuclear inclusions, suggesting a relationship between polyglutamine aggregation and dysfunction.


Subject(s)
Cognition/physiology , Exons , Hippocampus/physiology , Huntington Disease/physiopathology , Maze Learning/physiology , Nerve Tissue Proteins/genetics , Neuronal Plasticity/physiology , Nuclear Proteins/genetics , Pyramidal Cells/physiology , Space Perception/physiology , Synapses/physiology , Action Potentials/physiology , Aging , Animals , Crosses, Genetic , Hippocampus/growth & development , Hippocampus/physiopathology , Humans , Huntingtin Protein , Huntington Disease/genetics , In Vitro Techniques , Long-Term Potentiation , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Neuronal Plasticity/genetics
7.
Proc Natl Acad Sci U S A ; 97(14): 8093-7, 2000 Jul 05.
Article in English | MEDLINE | ID: mdl-10869421

ABSTRACT

Huntington's disease (HD) is a fatal inherited neurodegenerative disorder characterized by personality changes, motor impairment, and subcortical dementia. HD is one of a number of diseases caused by expression of an expanded polyglutamine repeat. We have developed several lines of mice that are transgenic for exon 1 of the HD gene containing an expanded CAG sequence. These mice exhibit a defined neurological phenotype along with neuronal changes that are pathognomonic for the disease. We have previously observed the appearance of neuronal intranuclear inclusions, but did not find evidence for neurodegeneration. In this study, we report that all lines of these mice develop a late onset neurodegeneration within the anterior cingulate cortex, dorsal striatum, and of the Purkinje neurons of the cerebellum. Dying neurons characteristically exhibit neuronal intranuclear inclusions, condensation of both the cytoplasm and nucleus, and ruffling of the plasma membrane while maintaining ultrastructural preservation of cellular organelles. These cells do not develop blebbing of the nucleus or cytoplasm, apoptotic bodies, or fragmentation of DNA. Neuronal death occurs over a period of weeks not hours. We also find degenerating cells of similar appearance within these same regions in brains of patients who had died with HD. We therefore suggest that the mechanism of neuronal cell death in both HD and a transgenic mouse model of HD is neither by apoptosis nor by necrosis.


Subject(s)
Disease Models, Animal , Huntington Disease/genetics , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Trinucleotide Repeat Expansion , Age of Onset , Animals , Cell Death , Cell Nucleus/pathology , Cerebellum/pathology , Corpus Striatum/pathology , Gyrus Cinguli/pathology , Humans , Huntingtin Protein , In Situ Nick-End Labeling , Mice , Neuroglia/pathology , Purkinje Cells/pathology
9.
Ann Neurol ; 47(1): 80-6, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10632104

ABSTRACT

Huntington's disease is a progressive neurodegenerative disease caused by an abnormally expanded (>36) CAG repeat within the ITI5 gene encoding a widely expressed 349-kd protein, huntingtin. The medium spiny neurons of the caudate preferentially degenerate in Huntington's disease, with the presence of neuronal intranuclear inclusions. Excitotoxicity is thought to be important in the pathogenesis of Huntington's disease; the recently described mitochondrial respiratory chain and aconitase defects in Huntington's disease brain are consistent with this hypothesis. A transgenic mouse model (R6/2) of Huntington's disease develops a movement disorder, muscle wasting, and premature death at about 14 to 16 weeks. Selective neuronal death in these mice is not seen until 14 weeks. Biochemical analysis of R6/2 mouse brain at 12 weeks demonstrated a significant reduction in aconitase and mitochondrial complex IV activities in the striatum and a decrease in complex IV activity in the cerebral cortex. Increased immunostaining for inducible nitric oxide synthase and nitrotyrosine was seen in the transgenic mouse model but not control mouse brains. These results extend the parallels between Huntington's disease and the transgenic mouse model to biochemical events and suggest complex IV deficiency and elevated nitric oxide and superoxide radical generation precede neuronal death in the R6/2 mouse and contribute to pathogenesis.


Subject(s)
Disease Models, Animal , Huntington Disease/pathology , Mitochondrial Myopathies/pathology , Animals , Brain/pathology , Immunohistochemistry , Mice , Mice, Transgenic
10.
Health Hum Rights ; 5(1): 88-115, 2000.
Article in English | MEDLINE | ID: mdl-11154525

ABSTRACT

Although India is a signatory to numerous international agreements on the rights of women and has a constitution that prohibits discrimination and exploitation by gender, as well as a plethora of related legislation, it has failed to satisfactorily protect the human rights of women, particularly those of sex workers. This is manifested in high levels of violence in the sex industry, child sex workers, lack of access to health care, and high levels of HIV infection. Policies that revolve around rescue and rehabilitation, or are based on the premise that sex work is immoral, are unlikely to effectively promote the well-being of sex workers. An alternative paradigm, which revolves around an explicit recognition of the human rights of sex workers together with an activist approach to achieve them, involving a collaboration between NGOs and collectives of sex workers, has worked well to protect the human rights and health of sex workers in India.


Subject(s)
Health Policy/legislation & jurisprudence , Occupational Health/legislation & jurisprudence , Prejudice , Sex Work/legislation & jurisprudence , Women's Rights/legislation & jurisprudence , AIDS Serodiagnosis , Adolescent , Adult , Child , Female , Health Services Accessibility , Humans , India , Violence
11.
Philos Trans R Soc Lond B Biol Sci ; 354(1386): 963-9, 1999 Jun 29.
Article in English | MEDLINE | ID: mdl-10434294

ABSTRACT

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by a CAG-polyglutamine repeat expansion. A mouse model of this disease has been generated by the introduction of exon 1 of the human HD gene carrying highly expanded CAG repeats into the mouse germ line (R6 lines). Transgenic mice develop a progressive neurological phenotype with a movement disorder and weight loss similar to that in HD. We have previously identified neuronal inclusions in the brains of these mice that have subsequently been established as the pathological hallmark of polyglutamine disease. Inclusions are present before symptoms, which in turn occur long before any selective neuronal cell death can be identified. We have extended the search for inclusions to skeletal muscle, which, like brain, contains terminally differentiated cells. We have conducted an investigation into the skeletal muscle atrophy that occurs in the R6 lines, (i) to provide possible insights into the muscle bulk loss observed in HD patients, and (ii) to conduct a parallel analysis into the consequence of inclusion formation to that being performed in brain. The identification of inclusions in skeletal muscle might be additionally useful in monitoring the ability of drugs to prevent inclusion formation in vivo.


Subject(s)
Huntington Disease/genetics , Huntington Disease/physiopathology , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Animals , Exons , Humans , Huntingtin Protein , Mice , Mice, Transgenic , Movement Disorders/genetics , Movement Disorders/physiopathology , Peptides/genetics , Repetitive Sequences, Amino Acid , Trinucleotide Repeats
12.
Philos Trans R Soc Lond B Biol Sci ; 354(1386): 971-9, 1999 06 29.
Article in English | MEDLINE | ID: mdl-10434295

ABSTRACT

Huntington's disease (HD) is an inherited progressive neurodegenerative disease caused by the expansion of a polyglutamine repeat sequence within a novel protein. Recent work has shown that abnormal intranuclear inclusions of aggregated mutant protein within neurons is a characteristic feature shared by HD and several other diseases involving glutamine repeat expansion. This suggests that in each of the these disorders the affected nerve cells degenerate as a result of these abnormal inclusions. A transgenic mouse model of HD has been generated by introducing exon 1 of the HD gene containing a highly expanded CAG sequence into the mouse germline. These mice develop widespread neuronal intranuclear inclusions and neurodegeneration specifically within those areas of the brain known to degenerate in HD. We have investigated the sequence of pathological changes that occur after the formation of nuclear inclusions and that precede neuronal cell death in these cells. Although the relation between inclusion formation and neurodegeneration has recently been questioned, a full characterization of the pathways linking protein aggregation and cell death will resolve some of these controversies and will additionally provide new targets for potential therapies.


Subject(s)
Brain/metabolism , Huntington Disease/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , Animals , Gene Expression Regulation , Humans , Huntingtin Protein , Huntington Disease/genetics , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Receptors, AMPA/metabolism , Receptors, GABA-A/metabolism , Receptors, GABA-B/metabolism , Receptors, Kainic Acid/metabolism , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
13.
Hum Mol Genet ; 8(5): 813-22, 1999 May.
Article in English | MEDLINE | ID: mdl-10196370

ABSTRACT

Huntington's disease (HD) is one of a class of inherited progressive neurodegenerative disorders that are caused by a CAG/polyglutamine repeat expansion. We have previously generated mice that are transgenic for exon 1 of the HD gene carrying highly expanded CAG repeats which develop a progressive movement disorder and weight loss with similarities to HD. Neuronal inclusions composed of the exon 1 protein and ubiquitin are present in specific brain regions prior to onset of the phenotype, which in turn occurs long before specific neurodegeneration can be detected. In this report we have extended the search for polyglutamine inclusions to non-neuronal tissues. Outside the central nervous system (CNS), inclusions were identified in a variety of post-mitotic cells. This is consistent with a concentration-dependent nucleation and aggregation model of inclusion formation and indicates that brain-specific factors are not necessary for this process. To possibly gain insights into the wasting that is observed in the human disease, we have conducted a detailed analysis of the timing and progression of inclusion formation in skeletal muscle and an investigation into the cause of the severe muscle atrophy that occurs in the mouse model. The formation of inclusions in non-CNS tissues will be particularly useful with respect to in vivo monitoring of pharmaceutical agents selected for their ability to prevent polyglutamine aggregation in vitro, without the requirement that the agent can cross the blood-brain barrier in the first instance.


Subject(s)
Huntington Disease/genetics , Muscle, Skeletal/ultrastructure , Muscular Atrophy/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Peptides/genetics , Animals , Cell Nucleus/genetics , Cell Nucleus/ultrastructure , Central Nervous System/pathology , Central Nervous System/physiopathology , Disease Models, Animal , Humans , Huntingtin Protein , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Muscular Atrophy/physiopathology , Neurons/pathology
14.
J Neurosci ; 19(8): 3248-57, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10191337

ABSTRACT

Transgenic mice expressing exon 1 of the human Huntington's disease (HD) gene carrying a 141-157 CAG repeat (line R6/2) develop a progressive neurological phenotype with motor symptoms resembling those seen in HD. We have characterized the motor deficits in R6/2 mice using a battery of behavioral tests selected to measure motor aspects of swimming, fore- and hindlimb coordination, balance, and sensorimotor gating [swimming tank, rotarod, raised beam, fore- and hindpaw footprinting, and acoustic startle/prepulse inhibition (PPI)]. Behavioral testing was performed on female hemizygotic R6/2 transgenic mice (n = 9) and female wild-type littermates (n = 22) between 5 and 14 weeks of age. Transgenic mice did not show an overt behavioral phenotype until around 8 weeks of age. However, as early as 5-6 weeks of age they had significant difficulty swimming, traversing the narrowest square (5 mm) raised beam, and maintaining balance on the rotarod at rotation speeds of 33-44 rpm. Furthermore, they showed significant impairment in prepulse inhibition (an impairment also seen in patients with HD). Between 8 and 15 weeks, R6/2 transgenic mice showed a progressive deterioration in performance on all of the motor tests. Thus R6/2 mice show measurable deficits in motor behavior that begin subtly and increase progressively until death. Our data support the use of R6/2 mice as a model of HD and indicate that they may be useful for evaluating therapeutic strategies for HD, particularly those aimed at reducing the severity of motor symptoms or slowing the course of the disease.


Subject(s)
Huntington Disease/genetics , Psychomotor Performance/physiology , Acoustic Stimulation , Analysis of Variance , Animals , Disease Progression , Female , Genotype , Humans , Mice , Mice, Transgenic , Mutation , Reflex, Startle , Swimming/physiology , Walking/physiology
15.
Brain Res ; 805(1-2): 234-40, 1998 Sep 14.
Article in English | MEDLINE | ID: mdl-9733972

ABSTRACT

Huntington's disease transgenic mice were tested in the elevated plus-maze test of anxiety at 6, 8, 10 and 12 weeks of age. At all ages, they showed significant and striking increases in the percentages of open arm entries and time spent on the open arms, compared with their normal littermates, indicating reduced anxiety. These increases were not secondary to a non-specific stimulant effect, since the transgenic mice made fewer closed arm entries, significantly so from 10 weeks of age. The mice were also tested in the holeboard, which provides measures of locomotor activity and directed exploration. From 8 weeks of age, the Huntington's mice were significantly less active than their normal littermates and made fewer exploratory head-dips. The increased open arm activity in the elevated plus-maze cannot therefore be secondary to increased exploration in the transgenic mice. In order to determine whether the reduced anxiety was due to differences in benzodiazepine receptor function, the mice were challenged with the benzodiazepine receptor antagonist, flumazenil. The results indicated that some of the reduced anxiety could be attributed to the presence of an endogenous anxiolytic ligand.


Subject(s)
Anxiety/psychology , Huntington Disease/psychology , Animals , Exploratory Behavior/physiology , Female , Flumazenil/pharmacology , Huntington Disease/genetics , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic/genetics , Species Specificity
16.
Exp Neurol ; 154(1): 31-40, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9875265

ABSTRACT

Striatal grafts have been proposed as a potential strategy for striatal repair in Huntington's disease, but it is unknown whether the diseased brain will compromise graft survival. A transgenic mouse line has recently been described in which hemizygotes with an expanded CAG repeat in exon 1 of the HD gene exhibit a progressive neurological phenotype similar to the motor symptoms of Huntington's disease. We have therefore evaluated the effects of the transgenic brain environment on the survival, differentiation, and function of intrastriatal striatal grafts and undertaken a preliminary analysis of the effects of the grafts on the development of neurological deficits in the host mice. Hemizygote transgenic and wild-type littermate female mice received striatal grafts at 10 weeks of age and were allowed to survive 6 weeks. Normal healthy grafts were seen to survive and differentiate within the striatum of transgenic mice in a manner comparable to that seen in control mice. The transgenic mice exhibited a progressive decline in body weight from 9 weeks of age and a progressive hypoactivity in an open field test of general locomotor behavior. Although striatal grafts exerted a statistically significant influence on several indices of this impairment, all behavioral effects were small and did not exert any clinically relevant effect on the profound neurological deficiency of the transgenic mice.


Subject(s)
Behavior, Animal/physiology , Corpus Striatum/transplantation , Graft Survival , Huntington Disease/therapy , Motor Activity/physiology , Animals , Body Weight/genetics , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Motor Activity/genetics
17.
Nat Genet ; 15(2): 197-200, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9020849

ABSTRACT

Six inherited neurodegenerative diseases are caused by a CAG/polyglutamine expansion, including spinal and bulbar muscular atrophy (SBMA), Huntington's disease (HD), spinocerebellar ataxia type 1 (SCA1), dentatorubral pallidoluysian atrophy (DRPLA) Machado-Joseph disease (MJD or SCA3) and SCA2. Normal and expanded HD allele sizes of 6-39 and 35-121 repeats have been reported, and the allele distributions for the other diseases are comparable. Intergenerational instability has been described in all cases, and repeats tend to be more unstable on paternal transmission. This may present as larger increases on paternal inheritance as in HD, or as a tendency to increase on male and decrease on female transmission as in SCA1 (ref. 15). Somatic repeat instability is also apparent and appears most pronounced in the CNS. The major exception is the cerebellum, which in HD, DRPLA, SCA1 and MJD has a smaller repeat relative to the other brain regions tested. Of non-CNS tissues, instability was observed in blood, liver, kidney and colon. A mouse model of CAG repeat instability would be helpful in unravelling its molecular basis although an absence of CAG repeat instability in transgenic mice has so far been reported. These studies include (CAG) in the androgen receptor cDNA, (CAG) in the HD cDNA, (CAG) in the SCA1 cDNA, (CAG) in the SCA3 cDNA and as an isolated (CAG) tract.


Subject(s)
Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Transgenes/genetics , Trinucleotide Repeats , Animals , Female , Humans , Huntingtin Protein , Male , Mice , Mice, Transgenic , Mosaicism , Mutation , Organ Specificity , Polymerase Chain Reaction , Reproducibility of Results
18.
Hum Mol Genet ; 6(10): 1633-7, 1997.
Article in English | MEDLINE | ID: mdl-9300654

ABSTRACT

CAG/polyglutamine expansion has been shown to form the molecular basis of an increasing number of inherited neurodegenerative diseases. The mutation is likely to act by a dominant gain of function but the mechanism by which it leads to neuronal dysfunction and cell death is unknown. The proteins harbouring these polyglutamine tracts are unrelated and without exception are widely expressed with extensively overlapping expression patterns. The factors governing the cell specific nature of the neurodegeneration have yet to be understood. Upon a certain size threshold, expanded CAG repeats become unstable on transmission and a modest degree of somatic mosaicism is apparent. Similarly, the molecular basis of the instability and its tissue specificity has yet to be unravelled. Recent reports describing the first mouse models of CAG/polyglutamine disorders indicate that it will be possible to model both the pathogenic mechanism and the CAG repeat instability in the mouse. This has great potential and promise for uncovering the molecular basis of these diseases and developing therapeutic interventions.


Subject(s)
Huntington Disease/genetics , Mutation , Animals , Humans , Huntingtin Protein , Mice , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Trinucleotide Repeats
19.
Mol Gen Genet ; 245(6): 716-23, 1994 Dec 15.
Article in English | MEDLINE | ID: mdl-7830719

ABSTRACT

In the budding yeast Saccharomyces cerevisiae, progress of the cell cycle beyond the major control point in G1 phase, termed START, requires activation of the evolutionarily conserved Cdc28 protein kinase by direct association with G1 cyclins. We have used a conditional lethal mutation in CDC28 of S. cerevisiae to clone a functional homologue from the human fungal pathogen Candida albicans. The protein sequence, deduced from the nucleotide sequence, is 79% identical to that of S. cerevisiae Cdc28 and as such is the most closely related protein yet identified. We have also isolated from C. albicans two genes encoding putative G1 cyclins, by their ability to rescue a conditional G1 cyclin defect in S. cerevisiae; one of these genes encodes a protein of 697 amino acids and is identical to the product of the previously described CCN1 gene. The second gene codes for a protein of 465 residues, which has significant homology to S. cerevisiae Cln3. These data suggest that the events and regulatory mechanisms operating at START are highly conserved between these two organisms.


Subject(s)
CDC28 Protein Kinase, S cerevisiae/genetics , Candida albicans/genetics , Cyclins/genetics , Membrane Glycoproteins , Molecular Chaperones , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Base Sequence , Cell Cycle , Cloning, Molecular , Fungal Proteins/genetics , Genes, Fungal , Genetic Complementation Test , Molecular Sequence Data , Restriction Mapping , Sequence Alignment , Sequence Homology, Amino Acid
20.
Pharmacopsychiatry ; 25(6): 273-7, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1494594

ABSTRACT

In 61 acute schizophrenic patients the effects of haloperidol (HPL) and lorazepam combined vs. HPL alone and the interaction between these drugs were evaluated. Patients were assigned to groups randomly. The study design was open. Study duration was 28 days. Psychopathology was evaluated on the basis of BPRS scores. Extrapyramidal side-effects were rated according to Simpson and Angus (1970). Pharmacological parameters included serum levels of lorazepam, HPL, and reduced HPL. Mean daily lorazepam dosage was 0.05 mg/kg, mean HPL dosage 0.5 mg/kg. None of the patients treated with lorazepam and HPL achieved better BPRS total or subscores, nor did their condition improve faster than in patients treated with HPL alone. A significant linear relationship between lorazepam serum levels and oral dosage was found, but none between lorazepam serum levels and BPRS total score, subscore reduction, or extrapyramidal side-effects. The authors conclude that beneficial effects of lorazepam in the treatment of acute psychosis are scant and may not justify the risks incurred with routine comedication of lorazepam.


Subject(s)
Haloperidol/therapeutic use , Lorazepam/therapeutic use , Schizophrenia/drug therapy , Acute Disease , Adult , Drug Therapy, Combination , Female , Haloperidol/analogs & derivatives , Haloperidol/blood , Humans , Lorazepam/blood , Male , Middle Aged , Psychiatric Status Rating Scales , Schizophrenic Psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...