Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 139
Filter
1.
Int J Tuberc Lung Dis ; 26(11): 1016-1022, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36281048

ABSTRACT

BACKGROUND: Recommendations have been made to integrate screening for common non-communicable diseases (NCDs) within TB programs. However, we must ensure screening is tied to evidence-based interventions before scale-up. We aimed to map the existing evidence regarding interventions that address NCDs that most commonly affect people with TB.METHODS: We systematically searched PubMed, Medline, and Embase for studies that evaluated interventions to mitigate respiratory disease, cardiovascular disease, alcohol and substance use disorder, and mental health disorders among people with TB. We excluded studies that only screened for comorbidity but resulted in no further intervention. We also excluded studies focusing on smoking cessation interventions for which evidence-based guidelines are well established.RESULTS: The search identified 20 studies that met our inclusion criteria. The most commonly evaluated intervention was referral for diabetes care (6 studies). Other interventions included pulmonary rehabilitation (5 studies), care programs for alcohol use disorder (4 studies), and psychosocial support or individual counselling (5 studies).CONCLUSION: There is limited robust evidence to support identified interventions in changing individual outcomes, and a significant knowledge gap remains on the long-term durability of the interventions´ clinical benefit, reach, and effectiveness. Implementation research demonstrating feasibility and effectiveness is needed before scaling up.


Subject(s)
Noncommunicable Diseases , Substance-Related Disorders , Tuberculosis , Humans , Alcohol Drinking , Comorbidity , Counseling , Noncommunicable Diseases/prevention & control , Substance-Related Disorders/prevention & control , Tuberculosis/epidemiology
2.
BMJ Open ; 1(1): e000155, 2011 May 15.
Article in English | MEDLINE | ID: mdl-22021778

ABSTRACT

Background Medical devices are used widely for virtually every disease and condition. Although devices are subject to regulation, the number of recalls, the clinical data requirements for regulation and the impact on patient safety are poorly understood. Methods The authors defined a device using European directives and used publicly available information on the Medicines and Health Regulatory Authority website to determine the number of devices recalled from January 2006 to December 2010. Two reviewers independently assessed Field Safety Notices and Medical Device Alerts. The authors wrote to manufacturers to obtain further information and clinical data, and summarised data by year, Conformité Européenne classification, indication, and Food and Drug Administration recall system of severity. Results In total, 2124 field safety notices were issued over the 5-year period, an increase of 1220% (62 in 2006 to 757 in 2010). 447 Medical Device Alerts were issued in the same period, and 44% were assessed as a reasonable probability of causing serious adverse health consequences or death. The authors wrote to 192 manufacturers of withdrawn devices and received 101 (53%) replies; only four (2.1%) provided the clinical data the authors requested. A lack of available transparent data prevented full analyses of the safety impact. Of the highest-risk recalled devices, more than half were related to the cardiovascular system (25%) or musculoskeletal system (33%), and 88% (95% CI 80% to 97%) were assessed as a reasonable probability of causing serious adverse health consequences or death. For low-risk devices, the figure was 34% (95% CI 26% to 42%). Conclusion The number of medical devices subject to recalls or warnings in the UK has risen dramatically. A substantial number of these devices may have caused serious adverse effects in patients and contributed to healthcare costs. Significant problems exist in the UK with a lack of access to transparent data and a registry of the highest-risk devices.

3.
J Cell Biochem ; 98(2): 243-50, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16453279

ABSTRACT

Stannin (Snn) was discovered using subtractive hybridization methodology designed to find gene products related to selective organotin toxicity and apoptosis. The cDNAs for Snn were first isolated from brain tissues sensitive to trimethyltin, and were subsequently used to localize, characterize, and identify genomic DNA, and other gene products of Snn. Snn is a highly conserved, 88 amino acid protein found primarily in vertebrates. There is a minor divergence in the C-terminal sequence between amphibians and primates, but a nearly complete conservation of the first 60 residues in all vertebrates sequenced to date. Snn is a membrane-bound protein and is localized, in part, to the mitochondria and other vesicular organelles, suggesting that both localization and conservation are significant for the overall function of the protein. The structure of Snn in a micellar environment and its architecture in lipid bilayers have been determined using a combination of solution and solid-state NMR, respectively. Snn structure comprised a single transmembrane domain (residues 10-33), a 28-residue linker region from residues 34-60 that contains a conserved CXC metal binding motif and a putative 14-3-3xi binding region, and a cytoplasmic helix (residues 61-79), which is partially embedded into the membrane. Of primary interest is understanding how this highly-conserved peptide with an interesting structure and cellular localization transmits both normal and potentially toxic signals within the cell. Evidence to date suggests that organotins such as trimethyltin interact with the CXC region of Snn, which is vicinal to the putative 14-3-3 binding site. In vitro transfection analyses and microarray experiments have inferred a possible role of Snn in several key signaling systems, including activation of the p38-ERK cascade, p53-dependent pathways, and 14-3-3xi protein-mediated processes. TNFalpha can induce Snn mRNA expression in endothelial cells in a PKC-epsilon dependent manner. Studies with Snn siRNA suggest that this protein may be involved in growth regulation, since inhibition of Snn expression alone leads to reduced endothelial cells growth and induction of COP-1, a negative regulator of p53 function. A key piece of the puzzle, however, is how and why such a highly-conserved protein, localized to mitochondria, interacts with other regulatory proteins to alter growth and apoptosis. By knowing the structure, location, and possible signaling pathways involved, we propose that Snn constitutes an important sensor of mitochondrial damage, and plays a key role in the mediation of cross-talk between mitochondrial and nuclear compartments in specific cell types.


Subject(s)
Mitochondria/metabolism , Neuropeptides/chemistry , Neuropeptides/metabolism , Animals , Apoptosis/physiology , Cell Proliferation/drug effects , Environmental Pollutants/toxicity , Humans , Mitochondria/drug effects , Models, Molecular , Organotin Compounds/toxicity , Sequence Homology, Amino Acid , Signal Transduction/physiology , Species Specificity
4.
Neuroscience ; 117(3): 541-55, 2003.
Article in English | MEDLINE | ID: mdl-12617961

ABSTRACT

Calcineurin, a calmodulin-dependent protein phosphatase, regulates transcription and possibly apoptosis. Previous studies demonstrated that in baby hamster kidney-21 cells after co-transfection calcineurin interacts with Bcl-2, thereby altering transcription and apoptosis. Using co-immunoprecipitation and subcellular fractionation techniques, we observed that calcineurin occurred as a complex with Bcl-2 in various regions of rat and mouse brain. The calcineurin-Bcl-2 complex was identified in mitochondrial, nuclear, microsomal and cytosol fractions. In vitro induction of hypoxia and aglycia or N-methyl-D-aspartate treatment markedly altered both extent of complex formation and its subcellular localization. These observations suggest that Bcl-2 either sequesters calcineurin, that calcineurin dephosphorylates Bcl-2, or that Bcl-2 shuttles calcineurin to specific substrates. Calcineurin also co-immunoprecipitated with the inositol-tris-phosphate receptor. This interaction increased after in vitro hypoxia/aglycia. In Bcl-2 (-/-) mice, interactions between calcineurin- and inositol-tris-phosphate receptor occurred less frequently than in wild-type mice under both control and hypoxic conditions. Experiments involving cell-free systems, as well as brain slices treated with thapsigargin or with N-methyl-D-aspartate suggested that calcium and calmodulin activation of calcineurin leads to interactions between calcineurin and Bcl-2. These data indicate that during times of cellular stress and damage, Bcl-2 targets activated calcineurin to specific compartments and substrates.


Subject(s)
Calcineurin/metabolism , Calcium/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Blotting, Western/methods , Calcium Channels/drug effects , Calcium Channels/metabolism , Calmodulin/pharmacology , Cerebellum/cytology , Cerebellum/drug effects , Cerebellum/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Crosses, Genetic , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/physiopathology , In Vitro Techniques , Inositol 1,4,5-Trisphosphate Receptors , Male , Mice , Mice, Knockout/metabolism , Mice, Transgenic/metabolism , N-Methylaspartate/pharmacology , Neurons/drug effects , Precipitin Tests/methods , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Subcellular Fractions/classification , Subcellular Fractions/metabolism , Thapsigargin/pharmacology , Time Factors
5.
Neuroscience ; 117(3): 557-65, 2003.
Article in English | MEDLINE | ID: mdl-12617962

ABSTRACT

Although pathogenesis of neuronal ischemia is incompletely understood, evidence indicates apoptotic neuronal death after ischemia. Bcl-2, an anti-apoptotic and neuroprotective protein, interacts with calcineurin in non-neuronal tissues. Activation of calcineurin, which is abundant in the brain, may play a role in apoptosis. Using co-immunoprecipitation experiments in biopsy-derived, fresh human cortical and hippocampal slices, we examined possible interactions between calcineurin and Bcl-2. Calcineuin-Bcl-2 interactions increased after exposure in vitro to excitotoxic agents and conditions of hypoxia/aglycia. This interaction may shuttle calcineurin to substrates such as the inositol-1,4,5-tris-phosphate receptor because under these experimental conditions interactions between calcineurin and inositol-1,4,5-tris-phosphate receptor also increased. A specific calcineurin inhibitor, FK-520, attenuated insult-induced increases in calcineurin-Bcl-2 interactions and augmented caspase-3 like activity. These data suggest that Bcl-2 modulates neuroprotective effects of calcineurin and that calcineurin inhibitors increase ischemic neuronal damage.


Subject(s)
Calcineurin/metabolism , Hypoxia, Brain/metabolism , Kainic Acid/analogs & derivatives , Neurotoxins/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Tacrolimus/analogs & derivatives , Adult , Blotting, Western , Calcium Channels/drug effects , Calcium Channels/metabolism , Caspase 3 , Caspases/drug effects , Caspases/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Precursors/drug effects , Enzyme Precursors/metabolism , Female , Humans , Hypoxia, Brain/drug therapy , Hypoxia, Brain/physiopathology , Immunosuppressive Agents/therapeutic use , In Vitro Techniques , Inositol 1,4,5-Trisphosphate Receptors , Kainic Acid/pharmacology , Male , Middle Aged , N-Methylaspartate/pharmacology , Okadaic Acid/pharmacology , Precipitin Tests/methods , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Spectrin/drug effects , Spectrin/metabolism , Tacrolimus/therapeutic use
6.
J Biol Chem ; 275(45): 35442-7, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-10931839

ABSTRACT

Syk is an important protein-tyrosine kinase in immunoreceptor signaling. FcepsilonRI aggregation in mast cells induces tyrosine phosphorylation and increased enzymatic activity of Syk. The two adjacent tyrosines in the Syk activation loop are thought to be important for the propagation of FcepsilonRI signaling. To evaluate the phosphorylation of these tyrosines in vivo and further understand the relationship of Syk tyrosine phosphorylation with its function, an antibody was developed specific for phosphorylated tyrosines in the activation loop of Syk. FcepsilonRI aggregation on mast cells induced the phosphorylation of both tyrosine residues of the activation loop. The kinase activity of Syk played the major role in phosphorylating its activation loop tyrosines both in vivo and in vitro. In FcepsilonRI-stimulated mast cells, the total Syk tyrosine phosphorylation paralleled the phosphorylation of its activation loop tyrosines and downstream propagation of signals for histamine release. In contrast, the cell surface binding of anti-ganglioside monoclonal antibody AA4 induced only strong general tyrosine phosphorylation of Syk and minimal histamine release and weak phosphorylation of activation loop tyrosines. These results demonstrate that phosphorylation of the activation loop tyrosines is important for mediating receptor signaling and is a better marker of Syk function than is total Syk tyrosine phosphorylation.


Subject(s)
Enzyme Precursors/metabolism , Enzyme Precursors/physiology , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/physiology , Tyrosine/metabolism , Animals , Dose-Response Relationship, Drug , Enzyme Activation , Histamine/metabolism , Immunoblotting , Intracellular Signaling Peptides and Proteins , Mast Cells/metabolism , Phosphorylation , Precipitin Tests , Rats , Signal Transduction , Syk Kinase , Time Factors , Transfection , Tumor Cells, Cultured , Tyrosine/chemistry
7.
Toxicol Pathol ; 28(1): 43-53, 2000.
Article in English | MEDLINE | ID: mdl-10668990

ABSTRACT

Neurotoxicants with similar structural features or common mechanisms of chemical action frequently produce widely divergent neuropathologic outcomes. Methylmercury (MeHg) produces marked cerebellar dysmorphogenesis during critical periods of development. The pathologic picture is characterized by complete architectural disruption of neuronal elements within the cerebellum. MeHg binds strongly to protein and soluble sulphydryl groups. Binding to microtubular -SH groups results in catastrophic depolymerization of immature tyrosinated microtubules. However, more mature acetylated microtubules are resistant to MeHg-induced depolymerization. In contrast to MeHg, the structurally similar organotin trimethyltin (TMT) elicits specific apoptotic destruction of pyramidal neurons in the CA3 region of the hippocampus and in other limbic structures. Expression of the phylogenetically conserved protein stannin is required for development of TMT-induced lesions. Inhibition of expression using antisense oligonucleotides against stannin protects neurons from the effects of TMT, suggesting that this protein is required for expression of neurotoxicity. However, expression of stannin alone is insufficient for induction of apoptotic pathways in neuronal populations. The aromatic nitrocompound 1,3-dinitrobenzene (DNB) has 2 independent nitro groups that can redox cycle in the presence of molecular oxygen. Despite its ability to deplete neural glutathione stores, DNB produces edematous gliovascular lesions in the brain stem of rats. Glial cells are susceptible despite high concentrations of reduced glutathione compared with neuronal somata in the central nervous system (CNS). The severity of lesions produced by DNB is modulated by the activity of neurons in the affected pathways. The inherent discrepancy between susceptibility of neuronal and glial cell populations is likely mediated by differential control of the mitochondrial permeability transition in astrocytes and neurons. Lessons learned in the mechanistic investigation of neurotoxicants suggest caution in the evaluation and interpretation of structure-activity relationships, eg, TMT, MeHg, and DNB all induce oxidative stress, whereas TMT and triethyltin produce neuronal damage and myelin edema, respectively. The precise CNS molecular targets of cell-specific lipophilic neurotoxicants remain to be determined.


Subject(s)
Nervous System Diseases/chemically induced , Nervous System Physiological Phenomena/drug effects , Neurotoxins/toxicity , Animals , Humans , Nervous System Diseases/pathology , Rats
11.
Nursingconnections ; 12(1): 59-61, 1999.
Article in English | MEDLINE | ID: mdl-10401402
12.
Dig Dis Sci ; 44(6): 1136-41, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10389685

ABSTRACT

Peutz-Jeghers syndrome is characterized by gastrointestinal hamartomatous polyposis, mucocutaneous pigmentation, and a predisposition to cancer. The etiology of this syndrome is unknown. We investigated the expression of epidermal growth factor receptor (EGFr), transforming growth factor-alpha (TGF-alpha), transforming growth factor-beta1 (TGF-beta1) and transforming growth factor-beta receptor (TGF-beta RII) between normal and Peutz-Jeghers small bowel tissues. In addition, immunoprecipitation by phosphotyrosine antibodies followed by EGFr western blotting was measured and compared between a Peutz-Jeghers hamartoma and normal duodenal tissue. EGFr expression was increased 2.5-fold in normal and hamartomatous tissue of Peutz-Jeghers patients compared to normal small bowel tissue. In Peutz-Jeghers tissues, the major EGFr immunoreactive band was increased size from 170 to approximately 200 kDa. Using an antibody specific for activated EGFr, this larger size band was predominant in Peutz-Jeghers tissue. Immunoprecipitation of a hamartoma by a phosphotyrosine specific antibody followed by western blotting for EGFr demonstrated this 200-kDa band. Expression of TGF-alpha, TGF-beta1, TGF-beta1 RII was not significantly different between normal and Peutz-Jeghers tissues. In conclusion, EGFr was overexpressed in normal and hamartomatous small bowel tissue of Peutz-Jeghers patients, which suggests that EGFr in Peutz-Jeghers tissue is persistently activated or highly stimulated by endogenous ligands and also suggests a possible role for EGFr in the pathogenesis of Peutz-Jeghers syndrome.


Subject(s)
ErbB Receptors/metabolism , Gene Expression Regulation , Peutz-Jeghers Syndrome/metabolism , Biopsy , Blotting, Western/methods , Digestive System/metabolism , Digestive System/pathology , ErbB Receptors/analysis , Hamartoma/genetics , Hamartoma/metabolism , Hamartoma/pathology , Humans , Immunohistochemistry , Peutz-Jeghers Syndrome/genetics , Peutz-Jeghers Syndrome/pathology , Phosphorylation , Precipitin Tests , Proteins/analysis , Proteins/metabolism , Reference Values
13.
Mol Pharmacol ; 55(4): 632-41, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10101020

ABSTRACT

Phosphorylation of tau protein promotes stability of the axonal cytoskeleton; aberrant tau phosphorylation is implicated in the biogenesis of paired helical filaments (PHF) seen in Alzheimer's disease. Protein kinases and phosphatases that modulate tau phosphorylation have been identified using in vitro techniques; however, the role of these enzymes in vivo has not been determined. We used intraventricular infusions of antisense oligodeoxynucleotides (ODNs) directed against the major brain isoforms of the Ca2+/calmodulin-dependent phosphatase calcineurin to determine how reduced activity of this enzyme would affect tau dephosphorylation. Five-day infusions of antisense ODNs (5 and 10 nmol/day) in rats decreased immunoreactive levels and activity of calcineurin throughout the brain; sense ODNs, scrambled ODNs, and infusion vehicle alone had no effect. When neocortical slices were prepared from antisense ODN-treated rats and incubated for 1 to 2 h in vitro, tau protein remained phosphorylated as determined by using the phosphorylation-sensitive monoclonal antibodies AT-180 (Thr231) and AT-270 (Thr181). In contrast, AT-180 and AT-270 sites were completely dephosphorylated during incubation of neocortical slices from vehicle-infused controls and sense ODN-treated rats. Neocortical slices from antisense-treated rats were incubated with the phosphatase inhibitors okadaic acid (100 nM; 10 microM) and FK-520 (5 microM); these preparations showed enhanced tau phosphorylation, consistent with a significant loss of calcineurin activity. Thus, we conclude that phosphorylation of at least two sites on tau protein, namely, Thr181 and Thr231, is regulated by calcineurin.


Subject(s)
Brain/drug effects , Calcineurin/metabolism , Oligonucleotides, Antisense/pharmacology , tau Proteins/metabolism , Animals , Brain/metabolism , Calcineurin/drug effects , Calcineurin/genetics , In Vitro Techniques , Male , Neocortex/drug effects , Neocortex/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Threonine/metabolism
14.
Nursingconnections ; 12(4): 54-6, 1999.
Article in English | MEDLINE | ID: mdl-12016642
17.
Methods Mol Med ; 22: 99-103, 1999.
Article in English | MEDLINE | ID: mdl-21380826

ABSTRACT

Transfection of specific elements of DNA into cultured mammalian cells allows for the analysis of a range of functional and toxicologic mechanisms. At the heart of this technique is the ability to promote the uptake of DNA into actively growing cells, and to detect and analyze the expression of the gene(s) encoded by the DNA (1). The two basic types of transfection analyses are transient transfections, in which the DNA is expressed during the few days postapplication, and stable transfections, in which cells expressing the gene of interest are actively selected via cointroduction of a marker for positive selections (2). Choosing which technique is appropriate for a given experiment is determined by the temporal aspects of the question, the types of assays performed, and whether the gene of interest is expressed constitutively or via an inducible promoter. Illustrations for both methods will be provided.

18.
Mamm Genome ; 9(7): 556-64, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9657854

ABSTRACT

Stannin is a protein that has been localized to trimethyltin-sensitive cell populations, and evidence suggests it plays a role in the toxic effects of organotins. In this study, we have isolated a mouse stannin genomic clone and have characterized the gene's intron-exon organization, promoter region, and chromosomal location. We have also isolated a partial human stannin cDNA clone and analyzed the open reading frame. The mouse genomic clone spans approximately 19 kb and consists of one intron and two exons. The splice site consensus sequence was maintained at all intron-exon junctions. Promoter analysis suggests that two putative promoter sites exist, each containing multiple regulatory elements and transcription factor-binding sites. Fluorescence in situ hybridization analysis localized stannin to mouse Chromosome (Chr) 16 at band A2. This region is homologous to the proximal region of human Chr 16 (16p13) to which stannin has been previously mapped. Sequence analysis revealed that the 264-bp open reading frame was identical between rat and mouse. The human sequence was 98% identical, with two amino acid substitutions near the c-terminal end of the peptide. These data suggest that stannin is highly conserved between species, and its unusual pattern of cellular expression may, in part, be explained via cell-specific promoters.


Subject(s)
Chromosome Mapping , Neuropeptides/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosomes, Human, Pair 16 , Cloning, Molecular , Conserved Sequence , DNA , Exons , Gene Library , Humans , Introns , Male , Mice , Molecular Sequence Data , Open Reading Frames , Promoter Regions, Genetic , RNA Splicing , Transcription, Genetic
20.
Environ Health Perspect ; 106 Suppl 1: 131-58, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9539010

ABSTRACT

Risk assessment is a process often divided into the following steps: a) hazard identification, b) dose-response assessment, c) exposure assessment, and d) risk characterization. Regulatory toxicity studies usually are aimed at providing data for the first two steps. Human case reports, environmental research, and in vitro studies may also be used to identify or to further characterize a toxic hazard. In this report the strengths and limitations of in vitro techniques are discussed in light of their usefulness to identify neurotoxic hazards, as well as for the subsequent dose-response assessment. Because of the complexity of the nervous system, multiple functions of individual cells, and our limited knowledge of biochemical processes involved in neurotoxicity, it is not known how well any in vitro system would recapitulate the in vivo system. Thus, it would be difficult to design an in vitro test battery to replace in vivo test systems. In vitro systems are well suited to the study of biological processes in a more isolated context and have been most successfully used to elucidate mechanisms of toxicity, identify target cells of neurotoxicity, and delineate the development and intricate cellular changes induced by neurotoxicants. Both biochemical and morphological end points can be used, but many of the end points used can be altered by pharmacological actions as well as toxicity. Therefore, for many of these end points it is difficult or impossible to set a criterion that allows one to differentiate between a pharmacological and a neurotoxic effect. For the process of risk assessment such a discrimination is central. Therefore, end points used to determine potential neurotoxicity of a compound have to be carefully selected and evaluated with respect to their potential to discriminate between an adverse neurotoxic effect and a pharmacologic effect. It is obvious that for in vitro neurotoxicity studies the primary end points that can be used are those affected through specific mechanisms of neurotoxicity. For example, in vitro systems may be useful for certain structurally defined compounds and mechanisms of toxicity, such as organophosphorus compounds and delayed neuropathy, for which target cells and the biochemical processes involved in the neurotoxicity are well known. For other compounds and the different types of neurotoxicity, a mechanism of toxicity needs to be identified first. Once identified, by either in vivo or in vitro methods, a system can be developed to detect and to evaluate predictive ability for the type of in vivo neurotoxicity produced. Therefore, in vitro tests have their greatest potential in providing information on basic mechanistic processes in order to refine specific experimental questions to be addressed in the whole animal.


Subject(s)
Nervous System/drug effects , Toxicology/methods , Animals , Calcium/metabolism , Cells, Cultured , Homeostasis , Humans , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...