Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
RSC Adv ; 14(6): 3712-3722, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38268546

ABSTRACT

In this study, Er-doped CoAl2O4 nanocrystals (NCs) were synthesized via co-precipitation. All the NCs were crystallized in the form of a single phase with a spinel structure and Er3+ ions replaced Al3+ ions in the formation of the CoAl2-xErxO4 alloy structure. The optical characteristics of the Er3+ ion-doped CoAl2O4 NCs were thoroughly investigated by analyzing both the UV-VIS and photoluminescence spectra, using the Judd-Ofelt theory. The effect of Er doping content on the luminescent properties of the CoAl2O4 pigment (using lasers emitting at wavelengths of 413 and 978 nm) has been studied. The values of Judd-Oflet intensity parameters (Ω2, Ω4, and Ω6) were determined from the absorption spectra using the least square fitting method. The J-O parameters were calculated and compared with those of other host materials; the values of the Ω2, Ω4, and Ω6 parameters decreased with an increase in Er concentration. This suggests that the rigidity and local symmetry of the host materials become weaker as the concentration of Er3+ ions increases. The highest value of the Ω2 parameter, when compared with Ω4 and Ω6, suggests that the vibrational frequencies in the given samples are relatively low. The upconversion fluorescence phenomenon was observed and explained in detail under an excitation wavelength of 978 nm when the excitation power was varied.

3.
J Biol Chem ; 297(4): 101191, 2021 10.
Article in English | MEDLINE | ID: mdl-34520759

ABSTRACT

Accumulation of α-synuclein is a main underlying pathological feature of Parkinson's disease and α-synucleinopathies, for which lowering expression of the α-synuclein gene (SNCA) is a potential therapeutic avenue. Using a cell-based luciferase reporter of SNCA expression we performed a quantitative high-throughput screen of 155,885 compounds and identified A-443654, an inhibitor of the multiple functional kinase AKT, as a potent inhibitor of SNCA. HEK-293 cells with CAG repeat expanded ATXN2 (ATXN2-Q58 cells) have increased levels of α-synuclein. We found that A-443654 normalized levels of both SNCA mRNA and α-synuclein monomers and oligomers in ATXN2-Q58 cells. A-443654 also normalized levels of α-synuclein in fibroblasts and iPSC-derived dopaminergic neurons from a patient carrying a triplication of the SNCA gene. Analysis of autophagy and endoplasmic reticulum stress markers showed that A-443654 successfully prevented α-synuclein toxicity and restored cell function in ATXN2-Q58 cells, normalizing the levels of mTOR, LC3-II, p62, STAU1, BiP, and CHOP. A-443654 also decreased the expression of DCLK1, an inhibitor of α-synuclein lysosomal degradation. Our study identifies A-443654 and AKT inhibition as a potential strategy for reducing SNCA expression and treating Parkinson's disease pathology.


Subject(s)
Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation/drug effects , Indazoles/pharmacology , Indoles/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , alpha-Synuclein/biosynthesis , HEK293 Cells , Humans , Parkinson Disease/genetics , Parkinson Disease/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , alpha-Synuclein/genetics
4.
PLoS One ; 16(8): e0256366, 2021.
Article in English | MEDLINE | ID: mdl-34383855

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0136930.].

5.
Am J Physiol Heart Circ Physiol ; 319(3): H642-H650, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32762556

ABSTRACT

The right ventricle (RV) is often overlooked in the evaluation of cardiac performance and treatment of left ventricular (LV) heart diseases. However, recent evidence suggests the RV may play an important role in maintaining systemic cardiac function and delivering stroke volume (SV). We used exercise cardiac magnetic resonance and biomechanical modeling to investigate the role of the RV in LV stroke volume regulation. We studied SV augmentation during exercise by pharmacologically inducing negative chronotropy (sHRi) in healthy volunteers and investigating training-induced SV augmentation in endurance athletes. SV augmentation during exercise after sHRi is achieved differently in the two ventricles. In the RV, the larger SV is driven by increasing contraction down to lower end-systolic volume (ESV; P < 0.001). In the LV, SV augmentation is achieved through an increase in end-diastolic volume (EDV; P < 0.001), avoiding contraction to a lower ESV. The same mechanism underlies the enhanced SV response observed in athletes. Changes in atrial area during SV augmentation suggest that the improved LV EDV response is sustained by the larger RV contractions. Using our biomechanical model, we explain this behavior by showing that the RV systolic function-driven regulation of LV SV optimizes the energetic cost of LV contraction and leads to minimization of the total costs of biventricular contraction. In conclusion, this work provides mechanistic understanding of the pivotal role of the RV in optimizing LV SV during exercise. It demonstrates why optimizing RV function needs to become a key part of therapeutic strategies in patients and training for athletes.NEW & NOTEWORTHY The right ventricle appears to have an important impact on maintaining systemic cardiac function and delivering stroke volume. However, its exact role in supporting left ventricular function has so far been unclear. This study demonstrates a new mechanism of ventricular interaction that provides mechanistic understanding of the key importance of the right ventricle in driving cardiac performance.


Subject(s)
Exercise , Heart/physiology , Stroke Volume , Ventricular Function, Left , Ventricular Function, Right , Adult , Bicycling , Biomechanical Phenomena , Female , Heart/diagnostic imaging , Heart Rate , Humans , Magnetic Resonance Imaging , Male , Systole , Young Adult
6.
PeerJ ; 7: e7779, 2019.
Article in English | MEDLINE | ID: mdl-31579630

ABSTRACT

BACKGROUND: Dengue infection represents a global health issue of growing importance. Dengue non-structural protein 1 (NS1) plays a central role in the early detection of the disease. The most common method for NS1 detection is testing by lateral flow immunoassays (LFIAs) with varying sensitivity. In this study, we present a highly sensitive magneto-enzyme LFIA for prompt diagnosis of dengue. METHODS: We have demonstrated the development of a magneto-enzyme LFIA combining super-paramagnetic nanoparticles as labels and Biotin-Streptavidin signal amplification strategy to detect dengue NS1. Factors affecting the test performance including antibody pair, super-paramagnetic nanoparticle size, nitrocellulose membrane type, amounts of detection and capture antibodies, and amounts of Streptavidin-polyHRP were optimized. Analytical sensitivity and cross-reactivity were determined. Clinical performance of the novel assay was evaluated using a panel of 120 clinical sera. RESULTS: This newly developed assay could detect NS1 of all four serotypes of dengue virus (DENV). The limit of detection (LOD) was found to be as low as 0.25 ng ml-1 for DENV-1 and DENV-3, 0.1 ng ml-1 for DENV-2, and 1.0 ng ml-1 for DENV-4. The LOD for DENV-2 was a 50-fold improvement over the best values previously reported. There was an absence of cross-reactivity with Zika NS1, Hepatitis B virus, Hepatitis C virus, and Japanese encephalitis virus. The sensitivity and specificity of the novel assay were 100% when tested on clinical samples. CONCLUSIONS: We have successfully developed a magneto-enzyme LFIA, allowing rapid and highly sensitive detection of dengue NS1, which is essential for proper management of patients infected with DENV.

8.
PLoS One ; 10(8): e0136930, 2015.
Article in English | MEDLINE | ID: mdl-26317803

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder caused by loss of dopaminergic neurons of the substantia nigra. The hallmark of PD is the appearance of neuronal protein aggregations known as Lewy bodies and Lewy neurites, of which α-synuclein forms a major component. Familial PD is rare and is associated with missense mutations of the SNCA gene or increases in gene copy number resulting in SNCA overexpression. This suggests that lowering SNCA expression could be therapeutic for PD. Supporting this hypothesis, SNCA reduction was neuroprotective in cell line and rodent PD models. We developed novel cell lines expressing SNCA fused to the reporter genes luciferase (luc) or GFP with the objective to enable high-throughput compound screening (HTS) for small molecules that can lower SNCA expression. Because SNCA expression is likely regulated by far-upstream elements (including the NACP-REP1 located at 8852 bp upstream of the transcription site), we employed zinc finger nuclease (ZFN) genome editing to insert reporter genes in-frame downstream of the SNCA gene in order to retain native SNCA expression control. This ensured full retention of known and unknown up- and downstream genetic elements controlling SNCA expression. Treatment of cells with the histone deacetylase inhibitor valproic acid (VPA) resulted in significantly increased SNCA-luc and SNCA-GFP expression supporting the use of our cell lines for identifying small molecules altering complex modes of expression control. Cells expressing SNCA-luc treated with a luciferase inhibitor or SNCA siRNA resulted in Z'-scores ≥ 0.75, suggesting the suitability of these cell lines for use in HTS. This study presents a novel use of genome editing for the creation of cell lines expressing α-synuclein fusion constructs entirely under native expression control. These cell lines are well suited for HTS for compounds that lower SNCA expression directly or by acting at long-range sites to the SNCA promoter and 5'-UTR.


Subject(s)
Deoxyribonucleases/metabolism , Models, Biological , Small Molecule Libraries/pharmacology , Up-Regulation/drug effects , alpha-Synuclein/genetics , Cell Line , Deoxyribonucleases/chemistry , Drug Evaluation, Preclinical , Genes, Reporter , High-Throughput Screening Assays/methods , Humans , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Zinc Fingers , alpha-Synuclein/metabolism
9.
Neurobiol Dis ; 71: 270-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25152487

ABSTRACT

Spinocerebellar ataxia 13 (SCA13) is an autosomal dominant disease resulting from mutations in KCNC3 (Kv3.3), a voltage-gated potassium channel. The KCNC3(R420H) mutation was first identified as causative for SCA13 in a four-generation Filipino kindred with over 20 affected individuals. Electrophysiological analyses in oocytes previously showed that this mutation did not lead to a functional channel and displayed a dominant negative phenotype. In an effort to identify the molecular basis of this allelic form of SCA13, we first determined that human KCNC3(WT) and KCNC3(R420H) display disparate post-translational modifications, and the mutant protein has reduced complex glycan adducts. Immunohistochemical analyses demonstrated that KCNC3(R420H) was not properly trafficking to the plasma membrane and surface biotinylation demonstrated that KCNC3(R420H) exhibited only 24% as much surface expression as KCNC3(WT). KCNC3(R420H) trafficked through the ER but was retained in the Golgi. KCNC3(R420H) expression results in altered Golgi and cellular morphology. Electron microscopy of KCNC3(R420H) localization further supports retention in the Golgi. These results are specific to the KCNC3(R420H) allele and provide new insight into the molecular basis of disease manifestation in SCA13.


Subject(s)
Arginine/genetics , Histidine/genetics , Intracellular Fluid/metabolism , Mutation/genetics , Shaw Potassium Channels/genetics , Spinocerebellar Degenerations/genetics , Animals , Animals, Genetically Modified , Biotinylation , COS Cells , Cadherins/metabolism , Chlorocebus aethiops , Cytoplasm/genetics , Cytoplasm/metabolism , Drosophila , Drosophila Proteins/genetics , Endoplasmic Reticulum/metabolism , Female , Humans , Male , Oocytes , Protein Processing, Post-Translational , Protein Transport , Spinocerebellar Ataxias/congenital , Spinocerebellar Degenerations/metabolism , Transfection
10.
PLoS One ; 4(7): e6235, 2009 Jul 20.
Article in English | MEDLINE | ID: mdl-19617910

ABSTRACT

Mouse models with physiological and behavioral differences attributable to differential plasticity of hippocampal and amygdalar neuronal networks are rare. We previously generated ataxin-2 (Atxn2) knockout mice and demonstrated that these animals lacked obvious anatomical abnormalities of the CNS, but showed marked obesity and reduced fertility. We now report on behavioral changes as a consequence of Atxn2-deficiency. Atxn2-deficiency was associated with impaired long-term potentiation (LTP) in the amygdala, but normal LTP in the hippocampus. Intact hippocampal plasticity was associated behaviorally with normal Morris Water maze testing. Impaired amygdala plasticity was associated with reduced cued and contextual fear conditioning. Conditioned taste aversion, however, was normal. In addition, knockout mice showed decreased innate fear in several tests and motor hyperactivity in open cage testing. Our results suggest that Atxn2-deficiency results in a specific set of behavioral and cellular disturbances that include motor hyperactivity and abnormal fear-related behaviors, but intact hippocampal function. This animal model may be useful for the study of anxiety disorders and should encourage studies of anxiety in patients with spinocerebellar ataxia type 2 (SCA2).


Subject(s)
Fear , Learning , Nerve Tissue Proteins/physiology , Space Perception , Amygdala/physiology , Animals , Ataxins , Behavior, Animal , Conditioning, Operant , Female , Hippocampus/physiology , Homozygote , Long-Term Potentiation , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics
11.
J Neurosci ; 29(29): 9148-62, 2009 Jul 22.
Article in English | MEDLINE | ID: mdl-19625506

ABSTRACT

Spinocerebellar ataxia type 2 (SCA2) is an autosomal dominantly inherited, neurodegenerative disease caused by an expansion of polyglutamine tracts in the cytosolic protein ataxin-2 (Atx2). Cerebellar Purkinje cells (PCs) are predominantly affected in SCA2. The cause of PC degeneration in SCA2 is unknown. Here we demonstrate that mutant Atx2-58Q, but not wild-type (WT) Atx2-22Q, specifically associates with the cytosolic C-terminal region of type 1 inositol 1,4,5-trisphosphate receptor (InsP(3)R1), an intracellular calcium (Ca(2+)) release channel. Association with Atx2-58Q increased the sensitivity of InsP(3)R1 to activation by InsP(3) in planar lipid bilayer reconstitution experiments. To validate physiological significance of these findings, we performed a series of experiments with an SCA2-58Q transgenic mouse model that expresses human full-length Atx2-58Q protein under the control of a PC-specific promoter. In Ca(2+) imaging experiments, we demonstrated that stimulation with 3,5-dihydroxyphenylglycine (DHPG) resulted in higher Ca(2+) responses in 58Q PC cultures than in WT PC cultures. DHPG-induced Ca(2+) responses in 58Q PC cultures were blocked by the addition of ryanodine, an inhibitor of the ryanodine receptor (RyanR). We further demonstrated that application of glutamate induced more pronounced cell death in 58Q PC cultures than in WT PC cultures. Glutamate-induced cell death of 58Q PC cultures was attenuated by dantrolene, a clinically relevant RyanR inhibitor and Ca(2+) stabilizer. In whole animal experiments, we demonstrated that long-term feeding of SCA1-58Q mice with dantrolene alleviated age-dependent motor deficits (quantified in beam-walk and rotarod assays) and reduced PC loss observed in untreated SCA2-58Q mice by 12 months of age (quantified by stereology). Results of our studies indicate that disturbed neuronal Ca(2+) signaling may play an important role in SCA2 pathology and also suggest that the RyanR constitutes a potential therapeutic target for treatment of SCA2 patients.


Subject(s)
Calcium Signaling/physiology , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/metabolism , Spinocerebellar Ataxias/physiopathology , Animals , Ataxins , COS Cells , Calcium/metabolism , Calcium Channel Blockers/administration & dosage , Calcium Signaling/drug effects , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Chlorocebus aethiops , Dantrolene/administration & dosage , Excitatory Amino Acid Agents/administration & dosage , Glutamic Acid/toxicity , Glycine/administration & dosage , Glycine/analogs & derivatives , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mice , Mice, Transgenic , Motor Activity/drug effects , Nerve Degeneration/pathology , Nerve Tissue Proteins/genetics , Purkinje Cells/drug effects , Purkinje Cells/pathology , Purkinje Cells/physiology , Resorcinols/administration & dosage , Ryanodine/administration & dosage , Spinocerebellar Ataxias/genetics
12.
Exp Neurol ; 208(2): 207-15, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17949716

ABSTRACT

Spinocerebellar ataxia 2 (SCA2) belongs to the group of neurodegenerative diseases caused by expansion of a polyglutamine (polyQ) domain. Overexpression of mutant ataxin-2 causes cell death and Golgi dispersion in cell culture as well as morphologic and functional changes in mouse models. To further define the mechanism of ataxin-2 induced cell death, we compared the cytotoxic effects of different domains of normal and mutant ataxin-2. N-terminal truncated ataxin-2(N) with expanded polyQ repeats did not form intranuclear inclusion and was less cytotoxic than the corresponding full-length ataxin-2. Ataxin-2(del42)[Q22], which lacks 42 amino acids (aa) within the Lsm-associated domain (LsmAD) necessary for Golgi localization, showed a diffuse cytoplasmic localization and was more toxic than wild type ataxin-2[Q22]. Mutant ataxin-2(del42)[Q108] displayed the same toxicity as ataxin-2[Q108], but did not disperse the Golgi apparatus to the extent seen with full-length mutant proteins. These observations confirm that ataxin-2 cytotoxicity increases with increasing polyQ expansion and Golgi dispersion and indicate that, in contrast to other polyQ diseases, N-terminal fragments containing the polyQ repeat are less toxic than full-length ataxin-2. Deletion of 42 aa in the Lsm-AD in ataxin-2 results in cytotoxicity without significant abnormalities in the Golgi apparatus. These findings suggest that the C-terminal domains are important for ataxin-2 cytotoxicity and that Golgi abnormalities may not be primary in the pathogenic process.


Subject(s)
COS Cells/physiology , Mutation/physiology , Nerve Tissue Proteins/physiology , Peptides/genetics , Trinucleotide Repeat Expansion , Amino Acid Sequence , Animals , Ataxins , COS Cells/metabolism , Cell Death , Cell Survival , Chlorocebus aethiops , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Nerve Tissue Proteins/genetics , Peptide Fragments/genetics , Peptide Fragments/physiology , Protein Structure, Tertiary/physiology , Sequence Deletion , Transfection , Up-Regulation , trans-Golgi Network/metabolism
13.
Exp Neurol ; 203(2): 531-41, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17097639

ABSTRACT

Expansion of the polyQ repeat in ataxin-2 results in degeneration of Purkinje neurons and other neuronal groups including the substantia nigra in patients with spinocerebellar ataxia type 2 (SCA2). In animal and cell models, overexpression of mutant ataxin-2 induces cell dysfunction and death, but little is known about steady-state levels of normal and mutant ataxin-2 and cellular mechanisms regulating their abundance. Based on preliminary findings that ataxin-2 interacted with parkin, an E3 ubiquitin ligase mutated in an autosomal recessive form of Parkinsonism, we sought to determine whether parkin played a role in regulating the steady-state levels of ataxin-2. Parkin interacted with the N-terminal half of normal and mutant ataxin-2, and ubiquitinated the full-length form of both wild-type and mutant ataxin-2. Parkin also regulated the steady-state levels of endogenous ataxin-2 in PC12 cells with regulatable parkin expression. Parkin reduced abnormalities in Golgi morphology induced by mutant ataxin-2 and decreased ataxin-2 induced cytotoxicity. In brains of SCA2 patients, parkin labeled cytoplasmic ataxin-2 aggregates in Purkinje neurons. These studies suggest a role for parkin in regulating the intracellular levels of both wild-type and mutant ataxin-2, and in rescuing cells from ataxin-2-induced cytotoxicity. The role of parkin variants in modifying the SCA2 phenotype and its use as a therapeutic target should be further investigated.


Subject(s)
Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Ataxins , Blotting, Western , Cell Death/physiology , Cell Line , Cell Survival , Cytoplasm/metabolism , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Mice , Purkinje Cells/metabolism , Transfection , Trypan Blue , Ubiquitin-Protein Ligases/physiology
15.
Biochem Biophys Res Commun ; 339(1): 17-24, 2006 Jan 06.
Article in English | MEDLINE | ID: mdl-16293225

ABSTRACT

Ataxin-2, the gene product of the Spinocerebellar Ataxia Type 2 (SCA2) gene, is a protein of unknown function with abundant expression in embryonic and adult tissues. Its interaction with A2BP1/Fox-1, a protein with an RNA recognition motif, suggests involvement of ataxin-2 in mRNA translation or transport. To study the effects of in vivo ataxin-2 function, we generated an ataxin-2 deficient mouse strain. Ataxin-2 deficient mice were viable. Genotypic analysis of litters from mating of heterozygous mice showed segregation distortion with a significant reduction in the birth of Sca-/- females. Detailed macroscopic and microscopic analysis of surviving nullizygous Sca2 knockout mice showed no major histological abnormalities. On a fat-enriched diet, ataxin-2 deficient animals had increased weight gain. Our results demonstrate that ataxin-2, although widely expressed, is not essential in development or during adult survival in the mouse, but leads to adult-onset obesity.


Subject(s)
Mice, Knockout , Nerve Tissue Proteins/genetics , Animals , Animals, Newborn , Ataxins , Female , Male , Mice , Motor Activity , Nerve Tissue Proteins/physiology , Obesity/genetics , Obesity/physiopathology , Organ Specificity
17.
Hum Mol Genet ; 12(20): 2587-97, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-12925569

ABSTRACT

Inactivating mutations of the gene encoding parkin are responsible for some forms of autosomal recessive juvenile Parkinson disease. Parkin is a ubiquitin ligase that ubiquitinates misfolded proteins targeted for the proteasome-dependent protein degradation pathway. Using the yeast two-hybrid system and co-immunoprecipitation methods, we identified synaptotagmin XI as a protein that interacts with parkin. Parkin binds to the C2A and C2B domains of synaptotagmin XI resulting in the polyubiquitination of synaptotagmin XI. Truncated and missense mutated parkins reduce parkin-sytXI binding affinity and ubiquitination. Parkin-mediated ubiquitination also enhances the turnover of sytXI. In sporadic PD brain sections, sytXI was found in the core of the Lewy bodies. Since synaptotagmin XI is a member of the synaptotagmin family that is well characterized in their importance for vesicle formation and docking, the interaction with this protein suggests a role for parkin in the regulation of the synaptic vesicle pool and in vesicle release. Loss of parkin could thus affect multiple proteins controlling vesicle pools, docking and release and explain the deficits in dopaminergic function seen in patients with parkin mutations.


Subject(s)
Calcium-Binding Proteins , Membrane Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin/metabolism , Animals , Blotting, Western , Brain/pathology , COS Cells , Cell Line , Genes, Recessive , Green Fluorescent Proteins , Humans , Immunohistochemistry , Luminescent Proteins/metabolism , Membrane Glycoproteins/metabolism , Microscopy, Fluorescence , Models, Genetic , Mutation , Mutation, Missense , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Substantia Nigra/pathology , Synaptotagmins , Time Factors , Transfection , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases/metabolism
18.
Hum Mol Genet ; 12(13): 1485-96, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12812977

ABSTRACT

Spinocerebellar ataxia type 2 (SCA2) is caused by the expansion of a polyglutamine (polyQ) repeat in ataxin-2, the SCA2 gene product. In contrast to other polyQ diseases, intranuclear inclusions are not prominent in SCA2. In animal models with expression of mutant ataxin-2 targeted to Purkinje cells, neuronal dysfunction and morphologic changes are observed without the formation of intranuclear aggregates. In this report, we investigated the mechanisms underlying SCA2 pathogenesis using cellular models. We confirmed that the SCA2 gene product, ataxin-2, was predominantly located in the Golgi apparatus. Deletion of ER-exit and trans-Golgi signals in ataxin-2 resulted in an altered subcellular distribution. Expression of full-length ataxin-2 with an expanded repeat disrupted the normal morphology of the Golgi complex and colocalization with Golgi markers was lost. Intranuclear inclusions were only seen when the polyQ repeat was expanded to 104 glutamines, and even then were only observed in a small minority of cells. Expression of ataxin-2 with expanded repeats in PC12 and COS1 cells increased cell death compared with normal ataxin-2 and elevated the levels of activated caspase-3 and TUNEL-positive cells. These results suggest a link between cell death mediated by mutant ataxin-2 and the stability of the Golgi complex. The formation of intranuclear aggregates is not necessary for in vitro cell death caused by expression of full-length mutant ataxin-2.


Subject(s)
Golgi Apparatus/pathology , Peptides/genetics , Proteins/genetics , Animals , Apoptosis , Ataxins , Blotting, Western , Brefeldin A/pharmacology , COS Cells , Caspase 3 , Caspases/metabolism , Cell Death , Cell Division , Golgi Apparatus/metabolism , Green Fluorescent Proteins , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Luminescent Proteins/metabolism , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Mutation , Nerve Tissue Proteins , Neurons/metabolism , PC12 Cells , Peptides/chemistry , Plasmids/metabolism , Protein Synthesis Inhibitors/pharmacology , Rats , Recombinant Fusion Proteins/metabolism , Subcellular Fractions/metabolism , Time Factors , Transfection
19.
Anesth Analg ; 93(4): 832-8, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11574342

ABSTRACT

UNLABELLED: We sought to correlate in vivo microvascular, systemic function, hemodynamic, and oxygenation changes in autologous shed blood (n = 4) and hemoglobin glutamer-200 (Hb-200) (n = 4) resuscitations in hypovolemic dogs. Hemorrhage (approximately 40% blood loss) reduced mean arterial pressure to approximately 50 mm Hg and caused significant (P < 0.01) decreases in hematocrit, total hemoglobin, mean pulmonary arterial pressure, cardiac output, and oxygen delivery and significant (P < 0.01) increases in heart rate, systemic vascular resistance, and lactic acidosis. Significant (P < 0.01) changes in conjunctival microvascular variables also occurred, including a 19% decrease in venular diameter and 79% increase in average blood flow velocity. Shed blood resuscitation returned microvascular, systemic function, hemodynamic, and oxygenation variables to prehemorrhagic baseline values. In contrast, Hb-200 failed to restore hematocrit, total hemoglobin, cardiac output, oxygen delivery index, and systemic venous resistance to baseline, but it restored other systemic functions and all hemodynamic and microvascular changes. In addition, Hb-200 resuscitation in hypovolemic dogs (approximately 40% blood loss) did not cause extreme hemodilution or fatal outcome. This study confirms that real-time (in vivo) microvascular studies, which were conducted only in small rodent models in the past, can be performed simultaneously with systemic function, hemodynamic, and oxygenation studies in a large animal model for relevant data correlation. IMPLICATIONS: This is the first time that changes in the blood circulation have been studied, quantified, and correlated with systemic function, hemodynamic, and oxygenation changes in shock and during shock treatment in a large animal model. This study was performed by a new technology developed in-house to noninvasively and quantitatively study blood vessels in real time.


Subject(s)
Blood Substitutes/therapeutic use , Hypovolemia/drug therapy , Microcirculation/drug effects , Animals , Cattle , Dogs , Female , Hemodynamics/drug effects , Hemoglobins , Hypovolemia/physiopathology , Male , Resuscitation , Splenectomy
20.
Electrophoresis ; 22(10): 2098-102, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11465511

ABSTRACT

A "one-step" procedure, that not only removes the color and blocking proteins used in the colorimetric immunodetection step but also stains the proteins originally on the blot, is presented. Following immunostaining and recording of immunoreactive spots, the blot was allowed to air-dry overnight (or longer) at room temperature and then counterstained with a colloidal gold solution. This "air-drying" process apparently altered the affinity of the blocking proteins (and possibly other proteins added subsequently to the blotting step) towards the nitrocellulose membrane causing them to be removed by the acidic colloidal gold solution while the "blotted" proteins were being stained. The sensitivity of this counterstained blot was comparable to that of the blot without going through the immunodetection process. Since both immunodetection and protein staining were carried out on the same blot, this allowed easy identification of many immunoreactive spots to their corresponding proteins when the two profiles were superimposed. Using this procedure, we have detected 25 immunoreactive spots (or allergens) from the whole body extract of the German cockroach (Blattella germanica) that contribute to asthma and assigned them to their corresponding proteins on a two-dimensional (2-D) protein map. The apparent Mr and pI for each of the allergens were determined. We have also located one of the major cockroach allergens, Bla g 5 (glutathione S-transferase). Two-dimensional zymography revealed the presence of ten gelatinase-type proteolytic enzymes. Only one of the ten proteases comigrated with the immunoreactive proteins indicating that unlike other allergen-producing systems, most of the cockroach allergens do not possess protease activity.


Subject(s)
Allergens/isolation & purification , Electrophoresis, Gel, Two-Dimensional/methods , Immunoblotting/methods , Proteins/immunology , Proteins/isolation & purification , Allergens/chemistry , Animals , Asthma/etiology , Cockroaches/immunology , Humans , Insect Proteins/chemistry , Insect Proteins/immunology , Insect Proteins/isolation & purification , Isoelectric Point , Molecular Weight , Peptide Mapping/methods , Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...