14 research outputs found

    Postauditing in the EIA process

    Get PDF
    ABSTACT Environmental impact assessment (EIA) is an integral part of the protection of the environment. Although it is a tool which considers the impact on many different elements of the environment, the process also has many deficiencies. Because of its limitations, an audit has been forming since the beginning of EIA. An EIA audit evaluates the performance of an EIA by comparing actual impacts detected after the realization of the project to those that were predicted, those that were listed in the environmental impact statement. This paper will offer an insight into the EIA and the post-auditing process and contains three basic parts of these areas. The first part explores the EIA, its historical evolution and application in the Czech Republic. The second part will discuss the post-audit itself. In this part, the different post-auditing processes, which were carried out in different parts of the world, were chosen. Within the selected post-auditing, the predicted data from the environmental impact statements are statistically compared with resultant data from reality. It is therefore necessary to analyze the percentage given for how many predictions were wrong in a positive way, accurate or wrong in negative way. From different results of post-auditing, we can tell that only a small percentage of the..

    Effect of BNN27 treatment on paw grip endurance (PaGE) and rotarod in male and female mice.

    No full text
    <p>Male Tg vehicle-treated mice demonstrated a significant decrease in time spent on the PaGE test (a, left) at p95 (p = 0.0023, Mann-Whitney <i>U</i> test), p115 (p = 0.0003, Mann-Whitney <i>U</i> test), and p135 (p = 0.0385, Mann-Whitney <i>U</i> test) and the rotarod test (b, left) at p55 (p = 0.0089, Mann-Whitney <i>U</i> test), p95 (p = 0.0192, Mann-Whitney <i>U</i> test), and p135 (p = 0.0152, Mann-Whitney <i>U</i> test) relative to Wt vehicle-treated mice. Female Tg vehicle-treated mice demonstrated a decrease in time spent on the PaGE test (c, left) at p95 (p = 0.0128, Mann-Whitney <i>U</i> test), p115 (p<0.0001, Mann-Whitney <i>U</i> test), and p135 (p<0.0001, Mann-Whitney <i>U</i> test) and the rotarod test (d, left) at p55 (p = 0.0379, Mann-Whitney <i>U</i> test), p95 (p = 0.0026, Mann-Whitney <i>U</i> test), and p135 (p = 0.0002, Mann-Whitney <i>U</i> test) relative to Wt vehicle-treated mice. In Tg female mice, BNN27 10 mg/kg treatment significantly increased the time spent on the PaGE (c, right) (p<0.05, Dunn’s multiple comparison test) and rotarod (d, right) (p<0.01, Dunn’s multiple comparison test) tests compared to vehicle treatment at p95. Wt-vehicle (male: n = 10, female: n = 9; grey), Tg-vehicle (male: n = 11, female: n = 11; grey), Tg-BNN27 10 mg/kg (male: n = 9, female: n = 9; black), Tg-BNN27 50 mg/kg (male: n = 9, female: n = 9; red). Data are presented as median ± interquartile ranges. * p<0.05, ** p<0.01, ***p<0.001, ****p<0.0001.</p

    Effect of BNN27 treatment on stride length and width in male and female mice.

    No full text
    <p>Male Tg vehicle-treated mice exhibited a decrease in stride length (a, left) at p115 (p = 0.0136, Mann-Whitney <i>U</i> test) and stride width (b, left) at p95 (p = 0.0202, Mann-Whitney <i>U</i> test) relative to Wt vehicle-treated mice. Female Tg vehicle-treated mice showed a decrease in stride length (c, left) at p95 (p = 0.0155, Mann-Whitney <i>U</i> test), p115 (p = 0.0024, Mann-Whitney <i>U</i> test), and p135 (p = 0.0010, Mann-Whitney <i>U</i> test) but no change in stride width (d, left) (p>0.05, Mann-Whitney <i>U</i> test) compared to Wt vehicle-treated mice. There was no significant effect of BNN27 treatment (10 mg/kg or 50 mg/kg) on stride length or width in either male or female Tg mice compared to vehicle treatment (p>0.05, Kruskal-Wallis test) (a-d, right). Wt-vehicle (male: n = 10, female: n = 9; grey), Tg-vehicle (male: n = 8, female: n = 11; grey), Tg-BNN27 10 mg/kg (male: n = 9, female: n = 9; black), Tg-BNN27 50 mg/kg (male: n = 9, female: n = 9; red). Data are presented as median ± interquartile ranges. * p<0.05, ** p<0.01, ***p<0.001.</p

    Effect of BNN27 treatment on motor neuron survival in male and female mice.

    No full text
    <p>There was no change in motor neuron counts in a) male and b) female mice following BNN27 treatment. Top, representative H&E stained longitudinal sections (10μM) of lumbar spinal cord from Wt vehicle-treated mice and Tg mice treated with vehicle, BNN27 10 mg/kg, or BNN27 50 mg/kg. Arrows indicate motor neurons; scale bar = 150μM. Bottom, quantification of motor neuron counts from Tg mice treated with vehicle (male: n = 7, female: n = 11; grey), BNN27 10 mg/kg (male: n = 8, female: n = 10; black), and BNN27 50 mg/kg (male: n = 9, female: n = 12; red) normalized to Wt vehicle-treated mice (male: n = 12, female: n = 9; white). Tg vehicle-treated mice showed a decrease in the relative motor neuron number compared to Wt vehicle-treated mice in both males (a; p = 0.0095, Mann-Whitney <i>U</i> test) and females (b; p = 0.0042, Mann-Whitney <i>U</i> test). BNN27 treatment (10 mg/kg or 50 mg/kg) had no effect on motor neuron counts in male (a; p>0.05, Kruskal-Wallis test) or female (b; p>0.05, Kruskal-Wallis test) mice. Data are presented as median and 10-90th percentiles. ** p<0.01.</p

    MNTs attenuated survival of mouse motor neurons co-cultured with human astrocytes from SOD1 ALS patients.

    No full text
    <p>a) Survival of mouse motor neurons (MNs), expressing GFP under the Hb9 promoter, was significantly decreased in co-cultures with human astrocytes (iAstrocytes) from an ALS patient a SOD1 mutation (Patient 2/SOD1 210; light grey) compared to those from 3 compiled control patients (3 compiled; white) (p<0.0001, unpaired t-test). BNN20, BNN27, and BNN23 were each tested at 1 μM (dark grey), 10 μM (black), and 30 μM (medium grey) in iAstrocyte-mouse MN co-cultures. At 10 μM, BNN20 (p<0.01, Tukey’s test) and BNN27 (p<0.001, Tukey’s test) significantly increased the survival of MNs co-cultured with SOD1 iAstrocytes relative to untreated samples. b) The effects of BNN27 (red) were tested, along with Riluzole (dark grey), on iAstrocytes from 3 control patients (compiled) and 2 individual ALS patients each carrying a distinct SOD1 mutation (Patient 1/SOD1 91 and Patient 2/SOD1 210). Left, BNN27 (10 μM) treatment yielded a significant increase in MN survival in co-cultures with iAstrocytes from Patient 1 (p<0.0001, Tukey’s test) and Patient 2 (p<0.0001, Tukey’s test) relative to untreated co-cultures (light grey). Similarly, Riluzole (10 μM) treatment also significantly increased MN survival in co-cultures with Patient 1 (p<0.0001, Tukey’s test) and Patient 2 (p<0.0001, Tukey’s test) iAstroctyes. Right, representative images of mouse MNs co-cultured with iAstrocytes from control untreated (control), SOD1 210 untreated (untreated), SOD1 210 Riluzole (Riluzole), and SOD1 210 BNN27 (BNN27). Scale bar = 20μM. c) Oxidative stress levels were significantly increased in iAstrocytes from 2 ALS patients containing a SOD1 mutation (Patient 1/SOD1 91 and Patient 2/SOD1 210; light grey) compared to those from 2 control patients (white) (p<0.0001, Tukey’s test). BNN27 treatment (red) produced a significant decrease in oxidative stress levels from SOD1 iAstrocytes relative to untreated cells (p<0.001, Tukey’s test). Scale bar = 10μM. Experiments were performed in triplicate (a, c) and quadruplicate (b). Data are presented as mean ± SEMs. ** p<0.01, ***p<0.001, ****p<0.0001.</p

    Effect of BNN27 treatment on neuromuscular junction integrity in male and female mice.

    No full text
    <p>There was no change in neuromuscular junction (NMJ) integrity in a) male and b) female mice following BNN27 treatment. Top, representative tibialis anterior sections (20μM) from Wt vehicle-treated mice and Tg mice treated with vehicle, BNN27 10 mg/kg or 50 mg/kg. Sections were stained for α-Bungarotoxin (left: TMR-α-Bungarotoxin 1:750; red) and the vesicular acetylcholine transporter (middle: VAChT 1:10,000; green). Overlay images (right) show overlap of α-Bungarotoxin and VAChT. Scale bar = 20μM. Bottom, quantification of the percent overlay between α-Bungarotoxin and VAChT from Tg mice treated with vehicle (male: n = 8, female: n = 11; grey), BNN27 10 mg/kg (male: n = 10, female: n = 10; black) or BNN27 50 mg/kg (male: n = 11, female: n = 13; red) normalized to Wt vehicle-treated mice (male: n = 10, female: n = 9; white). Tg vehicle-treated mice showed a decrease in the relative overlap compared to Wt vehicle-treated males (p = 0.0005, Mann-Whitney <i>U</i> test) and females (p = 0.0018, Mann-Whitney <i>U</i> test). Data are presented as median and 10-90th percentiles. ** p<0.01, *** p<0.001.</p

    Effect of BNN27 treatment on paresis onset and survival in male and female mice.

    No full text
    <p>There was no significant effect of BNN27 (10 mg/kg or 50 mg/kg) treatment on paresis onset or survival compared to vehicle treatment in either male (p>0.05, Kruskal-Wallis test) or female (p>0.05, Kruskal-Wallis test) Tg mice. However, BNN27 10mg/kg treatment in Tg female mice produced a trend (p = 0.0523, Mann-Whitney <i>U</i> test) towards an increase in the age of paresis onset relative to Tg vehicle-treated female (b, left). Tg-vehicle (male: n = 8, female: n = 11; grey), Tg-BNN27 10 mg/kg (male: n = 10, female: n = 10; black), Tg-BNN27 50 mg/kg (male: n = 9, female: n = 9; red). Data are presented as percent values.</p

    Transient exposure to rotenone causes degeneration and progressive parkinsonian motor deficits, neuroinflammation, and synucleinopathy

    No full text
    Abstract Individuals with Parkinson’s disease (PD) typically receive a diagnosis once they have developed motor symptoms, at which point there is already significant loss of substantia nigra dopamine neurons, α-synuclein accumulation in surviving neurons, and neuroinflammation. Consequently, the point of clinical presentation may be too late to initiate disease-modifying therapy. In contrast to this clinical reality, animal models often involve acute neurodegeneration and potential therapies are tested concurrently or shortly after the pathogenic insult has begun rather than later when diagnostic clinical symptoms emerge. Therefore, we sought to develop a model that reflects the clinical situation more accurately. Middle-aged rats (7–9 months-old) received a single daily intraperitoneal injection of rotenone for 5 consecutive days and were observed over the next 8–9 months. Rotenone-treated rats showed transient motor slowing and postural instability during exposure but recovered within 9 days of rotenone cessation. Rats remained without behavioral deficits for 3–4 months, then developed progressive motor abnormalities over the ensuing months. As motor abnormalities began to emerge 3 months after rotenone exposure, there was significant loss of nigral dopaminergic neurons and significant microglial activation. There was delayed accumulation of α-synuclein in neurons of the substantia nigra and frontal cortex, which was maximal at 9 months post-rotenone. In summary, a brief temporally-remote exposure to rotenone causes delayed and progressive behavioral and neuropathological changes similar to Parkinson’s disease. This model mimics the human clinical situation, in which pathogenesis is well-established by the time diagnostic motor deficits appear. As such, this model may provide a more relevant experimental system in which to test disease-modifying therapeutics

    Dose-dependent reduction of somatic expansions but not Htt aggregates by di-valent siRNA-mediated silencing of MSH3 in HdhQ111 mice

    No full text
    Abstract Huntington's disease (HD) is a progressive neurodegenerative disorder caused by CAG trinucleotide repeat expansions in exon 1 of the HTT gene. In addition to germline CAG expansions, somatic repeat expansions in neurons also contribute to HD pathogenesis. The DNA mismatch repair gene, MSH3, identified as a genetic modifier of HD onset and progression, promotes somatic CAG expansions, and thus presents a potential therapeutic target. However, what extent of MSH3 protein reduction is needed to attenuate somatic CAG expansions and elicit therapeutic benefits in HD disease models is less clear. In our study, we employed potent di-siRNAs to silence mouse Msh3 mRNA expression in a dose-dependent manner in HdhQ111/+ mice and correlated somatic Htt CAG instability with MSH3 protein levels from simultaneously isolated DNA and protein after siRNA treatment. Our results reveal a linear correlation with a proportionality constant of ~ 1 between the prevention of somatic Htt CAG expansions and MSH3 protein expression in vivo, supporting MSH3 as a rate-limiting step in somatic expansions. Intriguingly, despite a 75% reduction in MSH3 protein levels, striatal nuclear HTT aggregates remained unchanged. We also note that evidence for nuclear Msh3 mRNA that is inaccessible to RNA interference was found, and that MSH6 protein in the striatum was upregulated following MSH3 knockdown in HdhQ111/+ mice. These results provide important clues to address critical questions for the development of therapeutic molecules targeting MSH3 as a potential therapeutic target for HD

    SVA insertion in X-linked Dystonia Parkinsonism alters histone H3 acetylation associated with TAF1 gene.

    No full text
    X-linked Dystonia-Parkinsonism (XDP) is a neurodegenerative disease linked to an insertion of a SINE-VNTR-Alu (SVA)-type retrotransposon within an intron of TAF1. This SVA insertion induces aberrant TAF1 splicing and partial intron retention, thereby decreasing levels of the full-length transcript. Here we sought to determine if these altered transcriptional dynamics caused by the SVA are also accompanied by local changes in histone acetylation, given that these modifications influence gene expression. Because TAF1 protein may itself exhibit histone acetyltransferase activity, we also examined whether decreased TAF1 expression in XDP cell lines and post-mortem brain affects global levels of acetylated histone H3 (AcH3). The results demonstrate that total AcH3 are not altered in XDP post-mortem prefrontal cortex or cell lines. We also did not detect local differences in AcH3 associated with TAF1 exons or intronic sites flanking the SVA insertion. There was, however, a decrease in AcH3 association with the exon immediately proximal to the intronic SVA, and this decrease was normalized by CRISPR/Cas-excision of the SVA. Collectively, these data suggest that the SVA insertion alters histone status in this region, which may contribute to the dysregulation of TAF1 expression
    corecore