Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
J Intern Med ; 285(6): 594-607, 2019 06.
Article in English | MEDLINE | ID: mdl-30963638

ABSTRACT

Traumatic brain injury (TBI) is caused by a head impact with a force exceeding regular exposure from normal body movement which the brain normally can accommodate. People affected include, but are not restricted to, sport athletes in American football, ice hockey, boxing as well as military personnel. Both single and repetitive exposures may affect the brain acutely and can lead to chronic neurodegenerative changes including chronic traumatic encephalopathy associated with the development of dementia. The changes in the brain following TBI include neuroinflammation, white matter lesions, and axonal damage as well as hyperphosphorylation and aggregation of tau protein. Even though the human brain gross anatomy is different from rodents implicating different energy transfer upon impact, especially rotational forces, animal models of TBI are important tools to investigate the changes that occur upon TBI at molecular and cellular levels. Importantly, such models may help to increase the knowledge of how the pathologies develop, including the spreading of tau pathologies, and how to diagnose the severity of the TBI in the clinic. In addition, animal models are helpful in the development of novel biomarkers and can also be used to test potential disease-modifying compounds in a preclinical setting.


Subject(s)
Brain Injuries, Traumatic/pathology , Disease Models, Animal , Animals , Humans
3.
J Intern Med ; 280(2): 177-202, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27165517

ABSTRACT

There are more than 30 human proteins whose aggregation appears to cause degenerative maladies referred to as amyloid diseases or amyloidoses. These disorders are named after the characteristic cross-ß-sheet amyloid fibrils that accumulate systemically or are localized to specific organs. In most cases, current treatment is limited to symptomatic approaches and thus disease-modifying therapies are needed. Alzheimer's disease is a neurodegenerative disorder with extracellular amyloid ß-peptide (Aß) fibrils and intracellular tau neurofibrillary tangles as pathological hallmarks. Numerous clinical trials have been conducted with passive and active immunotherapy, and small molecules to inhibit Aß formation and aggregation or to enhance Aß clearance; so far such clinical trials have been unsuccessful. Novel strategies are therefore required and here we will discuss the possibility of utilizing the chaperone BRICHOS to prevent Aß aggregation and toxicity. Type 2 diabetes mellitus is symptomatically treated with insulin. However, the underlying pathology is linked to the aggregation and progressive accumulation of islet amyloid polypeptide as fibrils and oligomers, which are cytotoxic. Several compounds have been shown to inhibit islet amyloid aggregation and cytotoxicity in vitro. Future animal studies and clinical trials have to be conducted to determine their efficacy in vivo. The transthyretin (TTR) amyloidoses are a group of systemic degenerative diseases compromising multiple organ systems, caused by TTR aggregation. Liver transplantation decreases the generation of misfolded TTR and improves the quality of life for a subgroup of this patient population. Compounds that stabilize the natively folded, nonamyloidogenic, tetrameric conformation of TTR have been developed and the drug tafamidis is available as a promising treatment.


Subject(s)
Amyloidosis/therapy , Alzheimer Disease/therapy , Amyloid/physiology , Amyloidosis/physiopathology , Animals , Diabetes Mellitus, Type 2/therapy , Humans , Liver Transplantation , Prealbumin/physiology
4.
J Cell Mol Med ; 11(4): 810-25, 2007.
Article in English | MEDLINE | ID: mdl-17760842

ABSTRACT

Inflammation is associated with both acute and chronic neurological disorders, including stroke and Alzheimer's disease (AD). Cytokines such as interleukin (IL)-1 have several activities in the brain both under physiological and pathophysiological conditions. The objective of this study was to evaluate consequences of the central blockade of IL-1 transmission in a previously developed transgenic mouse strain with brain-directed overexpression of human soluble IL-1 receptor antagonist (Tg hsIL-1ra). Effects on brain morphology and brain levels of the AD-related proteins beta-amyloid precursor protein (APP) and presenilin 1(PS1), as well as the levels of IL-1beta, IL-6 and tumour necrosis factor-alpha (TNF-alpha) were analysed in homozygotic and heterozygotic mice and wild type (WT) controls, of both genders and of young (30-40 days) and adult (13-14 months) age. A marked reduction in brain volume was observed in transgenic mice as determined by volumetry. Western blot analysis showed higher levels of APP, but lower levels of PS1, in adult animals than in young ones. In the cerebellum, heterozygotic (Tg hsIL-1ra(+/-)) mice had lower levels of APP and PS1 than WT mice. With one exception, there were no genotypic differences in the levels of IL-1beta, IL-6 and TNF-alpha. The cytokine levels were generally higher in adult than in young mice. In conclusion, the chronic blockade of IL-1 signalling in the brain was associated with an atrophic phenotype of the brain, and with modified levels of APP and PS1. Brain-directed overexpression of hsIL-1ra was not followed by major compensatory changes in the levels of pro-inflammatory cytokines.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/metabolism , Brain/pathology , Cytokines/metabolism , Interleukin 1 Receptor Antagonist Protein/metabolism , Presenilin-1/metabolism , Animals , Brain/anatomy & histology , Female , Humans , Male , Mice , Organ Size
5.
J Neurosci Res ; 84(3): 596-605, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16802321

ABSTRACT

PTEN is a dual phosphatase that negatively regulates the phosphatidylinositol 3-kinase (PI3K)/Akt signalling pathway important for cell survival. We determined effects of the inflammation and oxidative stresses of tumor necrosis factor-alpha (TNFalpha) and trans-4-hydroxy-2-nonenal (HNE), respectively, on PTEN, Akt, and GSK3beta signalling in rat primary cortical neurons. The inhibitors bisperoxovanadium [bpV(Pic)] and LY294002 were also used to determine PTEN and PI3K involvement in TNFalpha and HNE modulation of neuronal cell death. PTEN inhibition with bpV(Pic) alone did not affect Ser(473)Akt or Ser(9)GSK3beta phosphorylation. Instead, effects of this inhibitor were manifest when it was used together with TNFalpha and to a lesser extent with HNE. TNFalpha together with PTEN inhibition increased phosphorylation of Ser(473)Akt and Ser(9)GSK3beta. TNFalpha and HNE both gave decreased numbers of viable and increased numbers of early apoptotic neurons. PTEN inhibition partially reversed the toxic effect of TNFalpha as shown by an increased number of viable and a decreased number of early apoptotic neurons. All effects were reversed by PI3K inhibition. HNE together with inhibition of PTEN gave increased Ser(473)Akt but not Ser(9)GSK3beta phosphorylation and no effects on the number of viable or early apoptotic cells. In conclusion, PTEN inhibition gives a mild reversal of TNFalpha- but not HNE-induced cell death via the PI3K pathway.


Subject(s)
Apoptosis/physiology , Glycogen Synthase Kinase 3/metabolism , Neurons/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Aldehydes/pharmacology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Cysteine Proteinase Inhibitors/pharmacology , Encephalitis/chemically induced , Encephalitis/metabolism , Encephalitis/physiopathology , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/chemistry , Glycogen Synthase Kinase 3 beta , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neurons/drug effects , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/chemistry , Rats , Rats, Sprague-Dawley , Serine/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology
6.
J Neurosci Res ; 82(3): 432-41, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16184608

ABSTRACT

The mechanisms associated with cell death have been an important focus for neurobiology research. In the present study, the methodology of flow cytometry was used to optimize quantification of the toxic effects of tumor necrosis factor-alpha (TNF-alpha), trans-4-hydroxy-2-nonenal (4-HNE), and aged amyloid-beta (Abeta1-42) on rat primary cortical neurons. The fluorescent dyes annexin V-FITC and propidium iodide (PI) were used to identify populations of viable, early apoptotic, necrotic and late apoptotic cells by flow cytometry. Prior to exposure, the primary cultures showed 83% cell viability. Flow cytometry following labeling of cells with a specific neuronal marker, TUJ-1, revealed 82% pure neuronal populations, whereas approximately 7% were astrocytic as shown by glial fibrillary acidic protein positivity. Exposure of primary cultures to TNF-alpha, 4-HNE, and aged Abeta1-42 gave an increased number of early apoptotic cells. We show that flow cytometry is a suitable method for quantifying effects of different stressors on neurons in primary cultures. This technique could be useful for screening and testing of pharmacological compounds relevant to neurodegenerative disorders.


Subject(s)
Cerebral Cortex/metabolism , Flow Cytometry/methods , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Oxidative Stress/physiology , Aldehydes/pharmacology , Amyloid beta-Peptides/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Biomarkers/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Disease Models, Animal , Necrosis/chemically induced , Necrosis/metabolism , Necrosis/physiopathology , Neurodegenerative Diseases/physiopathology , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurotoxins/pharmacology , Oxidative Stress/drug effects , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley , Tubulin/metabolism , Tumor Necrosis Factor-alpha/pharmacology
7.
J Neurosci Res ; 66(1): 122-34, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11599009

ABSTRACT

Presenilins (PSs) are mutated in a majority of familial Alzheimer disease (FAD) cases. Mutated PSs may cause FAD by a number of pro-apoptotic mechanisms, or by regulating gamma-secretase activity, a protease involved in beta-amyloid precursor protein processing to the neurotoxic beta-amyloid peptide. Besides their normal endoproteolytic processing, PSs are substrates for caspases, being cleaved to alternative N-terminal and C-terminal fragments. So far little is known about the role of PSs cleavage in the apoptotic machinery. Here, we used SH-SY5Y neuroblastoma cells stably transfected with wild-type or exon 9 deleted presenilin 1 (PS1) in a time-course study after the exposure to the calcium ionophore A23187. During and after exposure to A 23187, intracellular calcium levels were higher in exon 9 deleted PS1 cells as compared with non-transfected and wild-type PS1 transfected cells. Cell death and the enrichment of apoptotic cells after A23187 exposure were increased by overexpression of exon 9 deleted PS1 as compared with the control cell lines. Wild-type PS1 cells were compared with exon 9 deleted PS1 cells and the temporal relationship between PS1 and other caspase substrates cleavages was analyzed. Exon 9 deleted PS1 cells exhibited a higher caspase-3 activation and a greater cleavage of PS1 and poly(ADP-ribose) polymerase (PARP) compared with wild-type PS1 cells. Exon 9 deleted PS1 cleavage occurred earlier than other caspase substrate cleavages (i.e., PARP and gelsolin), simultaneous with minimum detectable caspase-3 activation. Therefore, alternative cleavage of PS1 may play an important role for the regulation of the proteolytic cascade activated during apoptosis.


Subject(s)
Apoptosis/physiology , Calcimycin/pharmacology , Caspases/metabolism , Ionophores/pharmacology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neuroblastoma , Apoptosis/drug effects , Buffers , Calcium/metabolism , Cell Adhesion/physiology , Exons/genetics , Gene Deletion , Gene Expression/physiology , Homeostasis/drug effects , Homeostasis/physiology , Humans , Neurons/cytology , Neurons/enzymology , Presenilin-1 , Transfection , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/enzymology
8.
J Neurosci Res ; 63(5): 429-37, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11223918

ABSTRACT

Alzheimer's disease (AD) is characterized by the degeneration and loss of neurons, intracellular neurofibrillary tangles and the accumulation of extracellular senile plaques consisting mainly of beta-amyloid (A beta). A beta is generated from the amyloid precursor protein (APP) by sequential beta- and gamma-secretase cleavage. Alternatively, APP may be cleaved within the A beta region by alpha-secretase, preventing A beta formation. Here we investigated APP processing and secretion in primary neurons, using either colchicine or the calcium ionophore A23187 to induce apoptosis. Cell viability was determined by MTT measurements and apoptosis was further confirmed by annexin V and propidium iodide staining. We found that exposure to A23187 significantly decreased the secretion of soluble beta-secretase cleaved APP (beta-sAPP) in a caspase-dependent manner, although the secretion of total soluble APP beta sAPP) did not change. In addition, caspase inhibition restored cell viability to control levels. Exposure to colchicine did not change the amount of either secreted beta-sAPP or total sAPP and caspase inhibition was only partially able to restore cell viability. We conclude that calcium homeostasis is an important apoptotic effector specifically affecting the beta-secretase cleavage of APP.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Apoptosis/drug effects , Calcimycin/pharmacology , Calcium/physiology , Cerebral Cortex/cytology , Ionophores/pharmacology , Neurons/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Amyloid Precursor Protein Secretases , Animals , Annexin A5/analysis , Apoptosis/physiology , Aspartic Acid Endopeptidases/metabolism , Biological Transport/drug effects , Blotting, Western , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cerebral Cortex/embryology , Colchicine/pharmacology , Culture Media, Conditioned/analysis , Cysteine Proteinase Inhibitors/pharmacology , Cytoskeleton/drug effects , Endopeptidases , Neurons/enzymology , Neurons/metabolism , Rats , Rats, Sprague-Dawley
9.
Neuroscience ; 95(2): 593-601, 2000.
Article in English | MEDLINE | ID: mdl-10658639

ABSTRACT

Mutations in the presenilin-1 (PS1) and presenilin-2 (PS2) genes account for the majority of early-onset familial Alzheimer's disease cases. Recent studies suggest that presenilin gene mutations predispose cells to apoptosis by mechanisms involving altered calcium homeostasis and oxidative damage. In the present study, we determined whether PS1 mutations also sensitize cells to hyperosmotic stress-induced apoptosis. For this, we established SH-SY5Y neuroblastoma cell lines stably transfected with wild-type PS1 or either the PS1 exon 9 deletion (deltaE9) or PS1 L250S mutants. Cultured cells were exposed to an overnight (17 h) serum deprivation, followed by a 30 min treatment with either 20 mM glucose, 10 nM insulin-like growth factor-1 or 20 mM glucose + 10 nM insulin-like growth factor-1. Cells were then cultured for a further 3, 6 or 24 h and stained for apoptotic condensed nuclei using propidium iodide. Confirmation that cells were undergoing an active apoptotic process was achieved by labelling of DNA strand breaks using the terminal dUTP nick end labelling (TUNEL) technique. We also determined cell viability using 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction. Propidium iodide staining revealed that all cell lines and controls showed an increased number of apoptotic cells appearing with condensed nuclei at 24 h compared with 6 h and 3 h. High glucose-induced hyperosmotic stress resulted in significantly more apoptotic cells in the PS1 deltaE9 and PS1 L250S mutation cell lines at 24 h, compared with the wild-type PS1 lines (P < 0.001, ANOVA for both comparisons). Mean values (+/-S.D.) for the percentage number of apoptotic cells at 24 h following high glucose treatment were 16.1 +/- 3.5%, 26.7 +/- 5.5% and 31.0 +/- 5.7% for the wild-type PS1, PS1 deltaE9 and PS1 L250S lines, respectively. The pro-apoptotic effects of high glucose treatment were reversed by 10 nM insulin-like growth factor-1, although to a lesser extent in the mutation cell lines (5.8 +/- 2.4%, 15.2 +/- 7.3% and 13.2 +/- 2.0% for the wild-type PS1, PS1 deltaE9 (P < 0.01 for comparison with wild-type PS1) and PS1 L250S (P < 0.01 for comparison with wild-type PS1) transfected lines, respectively. TUNEL labelling of cells at 24 h following treatment gave essentially the same results pattern as obtained using propidium iodide. The percentage number of apoptotic cells with DNA strand breaks (means +/- S.D.) following high glucose treatment was 15.4 +/- 2.6% for the wild-type PS1, 26.8 +/- 3.2% for the PS1 deltaE9 (P < 0.001 for comparison with wild-type PS1) and 29.7 +/- 6.1% for the PS1 L250S transfected lines (P < 0.001 for comparison with wild-type PS1). The PS1 deltaE9 and PS1 L250S transfected lines also showed a higher number of apoptotic cells with DNA strand breaks at 24 h following high glucose plus insulin-like growth factor-1 treatment (11.4 +/- 2.0% and 14.3 +/- 2.8%, respectively), compared with values for the wild-type PS1 lines (8.5 +/- 2.4%). These differences were significant (P < 0.01) for the comparison of wild-type PS1 and PS1 L250S, but not PS1 deltaE9 lines. The mutation-related increases in number of apoptotic cells at 24 h following high glucose treatment were not accompanied by significant differences in cell viability at this time-point. Our results indicate that PS1 mutations predispose to hyperosmotic stress-induced apoptosis and that the anti-apoptotic effects of insulin-like growth factor-1 are compromised by these mutations. Perturbations of insulin-like growth factor-1 signalling may be involved in PS1 mutation-related apoptotic neuronal cell death in Alzheimer's disease.


Subject(s)
Alzheimer Disease/genetics , Apoptosis/genetics , Gene Deletion , Membrane Proteins/genetics , Alzheimer Disease/pathology , Brain Neoplasms , Cell Movement/physiology , Coloring Agents , Glucose/metabolism , Humans , Hypertonic Solutions , In Situ Nick-End Labeling , Insulin-Like Growth Factor I/metabolism , Mutation , Neurites/physiology , Neuroblastoma , Osmotic Pressure , Presenilin-1 , Signal Transduction/physiology , Tetrazolium Salts , Thiazoles , Transfection , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/metabolism
11.
Brain Res Bull ; 43(4): 393-403, 1997.
Article in English | MEDLINE | ID: mdl-9241442

ABSTRACT

Various forms of cell death induced by the glutamate receptor agonist, alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA), were analyzed by determining the capacity of cultured cerebellar granule cells to metabolize 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) into formazan, by measuring the leakage of lactate dehydrogenase (LDH), by using confocal microscopy to visualize propidium iodide staining of apoptotic nuclei, and by using field inversion gel electrophoresis (FIGE) for the detection of AMPA-produced cleavage of DNA into high molecular-weight fragments (50 kbp). All these measures indicated that stimulation of AMPA receptors may be involved in the neurotoxic effects of glutamate, and that AMPA-induced neurotoxicity in cerebellar granule cells display morphologically distinct features of both necrotic and apoptotic modes of cell death. In agreement with previous observations, a blockade of AMPA receptor desensitization was necessary to unmask AMPA-induced functional responses in cultured cerebellar granule neurons in vitro. Microfluorimetric measurements of free cytoplasmic calcium concentrations ([Ca2+]i) in single cerebellar neurons revealed that AMPA neurotoxicity was accompanied by a pronounced elevation of [Ca2+]i. Our current results add further evidence to the notion that glutamate-induced neurotoxicity in cerebellar granule cells is mediated not only through NMDA receptors but also through a direct activation of AMPA receptor-regulated cation channels.


Subject(s)
Cell Death/physiology , Cerebellum/drug effects , Neurotoxins/toxicity , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/toxicity , Animals , Benzothiadiazines/pharmacology , Cells, Cultured/drug effects , Diuretics , Dose-Response Relationship, Drug , Models, Neurological , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Sodium Chloride Symporter Inhibitors/pharmacology
13.
Neuroreport ; 7(15-17): 2659-64, 1996 Nov 04.
Article in English | MEDLINE | ID: mdl-8981442

ABSTRACT

During ischaemic brain injury, glutamate accumulation with overstimulation of postsynaptic glutamate receptors and intracellular Ca2+ overload lead to neuronal death. We have shown previously that delayed neuronal death in cultures of cerebellar granule cells (CGCs) exposed to glutamate occurs by apoptosis. Here, we report that lamin cleavage and dissolution of the microtubule network precede chromatin fragmentation in glutamate-induced CGC apoptosis. Like other events that characterize excitotoxic cell death, degradation of lamins, beta-tubulin and disruption of microtubule architecture is inhibited by the NMDA-receptor antagonist MK-801. Our findings suggest that cleavage of key cytoskeletal elements is an important step in glutamate-induced neuronal apoptosis.


Subject(s)
Apoptosis/physiology , Cerebellum/drug effects , Chromatin/metabolism , Glutamic Acid/pharmacology , Nuclear Proteins/metabolism , Tubulin/metabolism , Animals , Cells, Cultured/drug effects , Immunohistochemistry , Lamins , Rats
14.
FEBS Lett ; 394(3): 321-4, 1996 Oct 07.
Article in English | MEDLINE | ID: mdl-8830666

ABSTRACT

We have previously reported that glutamate can trigger a succession of necrosis and apoptosis in cerebellar granule cells (CGC). Since specific blockers of the N-methyl-D-aspartate (NMDA) receptor channel prevented both types of cell death, the role of Ca2+-dependent processes in the initiation of glutamate toxicity was further investigated. We examined the possible involvement of mitochondria and the role of the Ca2+/calmodulin-regulated protein phosphatase, calcineurin, in the development of either type of cell death. Cyclosporin A and the more selective calcineurin inhibitor, FK-506, prevented the development of both early necrosis and delayed apoptosis. In addition, cyclosporin A prevented the collapse of mitochondrial membrane potential observed during the exposure to glutamate and the concomitant necrotic phase. When CsA was added immediately after glutamate removal, it also prevented delayed apoptosis of neurons that had survived the necrotic phase. Altogether, these results suggest the involvement of calcineurin and a role for mitochondrial deenergization as early signals in neuronal apoptosis induced by glutamate.


Subject(s)
Apoptosis , Calmodulin-Binding Proteins/metabolism , Glutamic Acid/pharmacology , Mitochondria/metabolism , Neurons/drug effects , Neurons/pathology , Phosphoprotein Phosphatases/metabolism , Animals , Calcineurin , Calmodulin-Binding Proteins/antagonists & inhibitors , Cells, Cultured , Cerebellum/cytology , Cyclosporine/pharmacology , Membrane Potentials/drug effects , Necrosis , Phosphoprotein Phosphatases/antagonists & inhibitors , Rats , Tacrolimus/pharmacology
15.
16.
Exp Cell Res ; 221(2): 404-12, 1995 Dec.
Article in English | MEDLINE | ID: mdl-7493640

ABSTRACT

To investigate the involvement of proteases in apoptosis, rat thymocytes were treated with the glucocorticoid hormone methylprednisolone or the topoisomerase II inhibitor etoposide in the presence of selective substrate inhibitors of either interleukin-1 beta-converting enzyme (ICE), (Z-Val-Ala-Asp-chloromethylketone, VADcmk) or Ca(2+)-regulated serine protease (Suc-Ala-Ala-Pro-Phe-chloromethylketone, AAPFcmk). VADcmk protected from lamin proteolysis, chromatin fragmentation, cell shrinkage, and formation of apoptotic nuclei in both methylprednisolone- and etoposide-treated thymocytes when present during the initiation of the apoptotic process. AAPFcmk prevented lamin breakdown, chromatin fragmentation, and apoptotic morphological changes in thymocytes treated with methylprednisolone, but not with etoposide. Both MPS- and etoposide-treated thymocytes exhibited enhanced ICE-like protease activity which was maximal 1 h after treatment. This increase in proteolytic activity was blocked by VADcmk, but not AAPFcmk. Our findings suggest that ICE-like protease activity is critically involved in the early phase of both methylprednisolone- and etoposide-induced apoptosis in thymocytes, whereas the Ca(2+)-regulated serine protease is an obligatory component of the proteolytic cascade in methylprednisolone-induced apoptosis.


Subject(s)
Apoptosis , Cysteine Endopeptidases/physiology , Serine Endopeptidases/physiology , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Calcium/physiology , Caspase 1 , Cell Size , Cells, Cultured , Chromatin/metabolism , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , DNA/metabolism , Enzyme Inhibitors/pharmacology , Etoposide/pharmacology , Lamins , Male , Methylprednisolone/pharmacology , Nuclear Proteins/metabolism , Oligopeptides/pharmacology , Rats , Rats, Sprague-Dawley , Serine Proteinase Inhibitors/pharmacology , Topoisomerase II Inhibitors
17.
Neuron ; 15(4): 961-73, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7576644

ABSTRACT

During ischemic brain injury, glutamate accumulation leads to overstimulation of postsynaptic glutamate receptors with intracellular Ca2+ overload and neuronal cell death. Here we show that glutamate can induce either early necrosis or delayed apoptosis in cultures of cerebellar granule cells. During and shortly after exposure to glutamate, a subpopulation of neurons died by necrosis. In these cells, mitochondrial membrane potential collapsed, nuclei swelled, and intracellular debris were scattered in the incubation medium. Neurons surviving the early necrotic phase recovered mitochondrial potential and energy levels. Later, they underwent apoptosis, as shown by the formation of apoptotic nuclei and by chromatin degradation into high and low molecular weight fragments. These results suggest that mitochondrial function is a critical factor that determines the mode of neuronal death in excitotoxicity.


Subject(s)
Apoptosis/drug effects , Cerebellum/pathology , Glutamic Acid/pharmacology , Mitochondria/physiology , Neurons/drug effects , Animals , Chromatin/drug effects , Chromatin/ultrastructure , DNA/metabolism , Membrane Potentials/drug effects , Molecular Weight , Necrosis/chemically induced , Neurons/physiology , Neurons/ultrastructure , Rats , Rats, Sprague-Dawley
18.
Proc Natl Acad Sci U S A ; 92(16): 7162-6, 1995 Aug 01.
Article in English | MEDLINE | ID: mdl-7638161

ABSTRACT

N-Methyl-D-aspartate (NMDA) receptor-mediated neurotoxicity may depend, in part, on the generation of nitric oxide (NO.) and superoxide anion (O2.-), which react to form peroxynitrite (OONO-). This form of neurotoxicity is thought to contribute to a final common pathway of injury in a wide variety of acute and chronic neurologic disorders, including focal ischemia, trauma, epilepsy, Huntington disease, Alzheimer disease, amyotrophic lateral scelerosis, AIDS dementia, and other neurodegenerative diseases. Here, we report that exposure of cortical neurons to relatively short durations or low concentrations of NMDA, S-nitrosocysteine, or 3-morpholinosydnonimine, which generate low levels of peroxynitrite, induces a delayed form of neurotoxicity predominated by apoptotic features. Pretreatment with superoxide dismutase and catalase to scavenge O2.- partially prevents the apoptotic process triggered by S-nitrosocysteine or 3-morpholinosydnonimine. In contrast, intense exposure to high concentrations of NMDA or peroxynitrite induces necrotic cell damage characterized by acute swelling and lysis, which cannot be ameliorated by superoxide dismutase and catalase. Thus, depending on the intensity of the initial insult, NMDA or nitric oxide/superoxide can result in either apoptotic or necrotic neuronal cell damage.


Subject(s)
Apoptosis/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , N-Methylaspartate/toxicity , Nitric Oxide/toxicity , S-Nitrosothiols , Superoxides/toxicity , Animals , Cells, Cultured , Cysteine/analogs & derivatives , Cysteine/toxicity , DNA/drug effects , DNA/metabolism , Free Radicals , Kinetics , Molsidomine/analogs & derivatives , Molsidomine/toxicity , Necrosis , Neurons/drug effects , Neurons/pathology , Nitrates/toxicity , Oxidative Stress , Rats , Superoxide Dismutase/pharmacology
19.
J Biol Chem ; 269(48): 30553-60, 1994 Dec 02.
Article in English | MEDLINE | ID: mdl-7982974

ABSTRACT

Increasing concentrations (1-100 microM) of the redox cycling quinone, 2,3-dimethoxy-1,4-naphthoquinone (DMNQ), stimulated growth, triggered apoptosis, or caused necrosis of pancreatic RINm5F cells, depending on the dose and duration of the exposure. Following the exposure of RINm5F cells to 10 microM DMNQ, ornithine decarboxylase activity and polyamine biosynthesis increased. This was accompanied by enhanced cell proliferation. Conversely, exposure to 30 microM DMNQ for 3 h resulted in the inhibition of ornithine decarboxylase, intracellular polyamine depletion, and apoptotic cell killing. Pretreatment of the cultures with the phorbol ester, 12-O-tetradecanoylphorbol-13-acetate, restored polyamine levels and prevented apoptosis. Exposure to the same DMNQ concentration for only 1 h, with subsequent re-incubation in growth medium, neither caused polyamine depletion nor resulted in apoptosis. Finally, exposure to an even higher DMNQ concentration (100 microM) for either 1 or 3 h caused rapid intracellular Ca2+ overload, ATP, NAD+, and glutathione depletion, and extensive DNA single strand breakage, which resulted in necrotic cell death. Our results show that a disturbance of polyamine biosynthesis occurred prior to cell growth or apoptosis elicited by oxidative stress. In addition, we show that effects as opposite as cell proliferation and deletion, by either apoptosis or necrosis, can be induced, in the same system, by varying the exposure to a prooxidant.


Subject(s)
Apoptosis/drug effects , Cell Division/drug effects , Insulin/metabolism , Naphthoquinones/pharmacology , Oxidants/pharmacology , Polyamines/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Adenosylmethionine Decarboxylase/metabolism , Animals , Cell Line , Cell Survival/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Dose-Response Relationship, Drug , Insulin Secretion , Islets of Langerhans , Kinetics , Necrosis , Tetradecanoylphorbol Acetate/pharmacology , Time Factors
20.
FEBS Lett ; 355(1): 23-6, 1994 Nov 21.
Article in English | MEDLINE | ID: mdl-7525358

ABSTRACT

Nitric oxide (NO) is a diffusible messenger involved in several patho-physiological processes including immune-mediated cytotoxicity and neural cell killing. NO or the products of its redox chemistry can cause DNA damage and activate subsequent lethal reactions including energy depletion and cell necrosis. However, regardless of whether it is endogenously produced in response to cytokines, or generated by chemical breakdown of donor molecules, NO can also induce apoptosis in different systems. Here, we report that NO generation in response to a cytokine induced NO-synthase or by NO donors stimulates the expression of the tumor suppressor gene, p53, in RAW 264.7 macrophages or pancreatic RINm5F cells prior to apoptosis. NO-synthase inhibitors such as NG-monomethyl-L-arginine prevent the inducible NO generation as well as p53 expression and apoptosis. Since p53 expression is linked to apoptosis in some cells exposed to DNA damaging agents, we suggest that NO-induced apoptosis in these cell systems is the consequence of DNA damage and subsequent expression of this tumor suppressor gene.


Subject(s)
Apoptosis , Genes, p53 , Nitric Oxide/physiology , Tumor Suppressor Protein p53/biosynthesis , Amino Acid Oxidoreductases/metabolism , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Cell Line , Glutathione/analogs & derivatives , Glutathione/pharmacology , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/cytology , Mice , Nitric Oxide Synthase , Nitroso Compounds/pharmacology , Rats , S-Nitrosoglutathione , Up-Regulation , omega-N-Methylarginine
SELECTION OF CITATIONS
SEARCH DETAIL
...