Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Exp Mol Pathol ; 105(1): 23-31, 2018 08.
Article in English | MEDLINE | ID: mdl-29852183

ABSTRACT

Limitations in discovering useful tumor biomarkers and drug targets is not only due to patient-to-patient differences but also due to intratumor heterogeneity. Heterogeneity arises due to the genetic and epigenetic variation of tumor cells in response to microenvironmental interactions and cytotoxic therapy. We explored specific signaling pathway activation in glioblastoma (GBM) by investigating the intratumor activation of the MAPK and PI3K pathways. We present data demonstrating a striking preponderance for mutual exclusivity of MAPK and PI3K activation in GBM tissue, where MAPK activation correlates with proliferation and transcription factor CREB activation and PI3K activation correlates with CD44 expression. Bioinformatic analysis of signaling and CREB-regulated target genes supports the immunohistochemical data, showing that the MAPK-CREB activation correlates with proliferative regions. In-silico analysis suggests that MAPK-CREB signaling activates a pro-inflammatory molecular signature and correlates with a mesenchymal GBM subtype profile, while PI3K-CREB activation correlates with the proneural GBM subtype and a tumor cell invasive gene signature. Overall, the data suggests the existence of intratumor subtype heterogeneity in GBM and that using combinations of both MAPK and PI3K drug inhibitors is necessary for effective targeted therapy.


Subject(s)
Brain Neoplasms/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Genetic Heterogeneity , Glioblastoma/genetics , MAP Kinase Signaling System , Phosphatidylinositol 3-Kinases/metabolism , Transcriptome , Brain Neoplasms/metabolism , Cell Proliferation , Cyclic AMP Response Element-Binding Protein/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Phosphatidylinositol 3-Kinases/genetics
2.
Proc Natl Acad Sci U S A ; 106(2): 381-6, 2009 Jan 13.
Article in English | MEDLINE | ID: mdl-19122148

ABSTRACT

Cognitive decline in Alzheimer's disease (AD) involves pathological accumulation of synaptotoxic amyloid-beta (Abeta) oligomers and hyperphosphorylated tau. Because recent evidence indicates that glycogen synthase kinase 3beta (GSK3beta) activity regulates these neurotoxic pathways, we developed an AD therapeutic strategy to target GSK3beta. The strategy involves the use of copper-bis(thiosemicarbazonoto) complexes to increase intracellular copper bioavailability and inhibit GSK3beta through activation of an Akt signaling pathway. Our lead compound Cu(II)(gtsm) significantly inhibited GSK3beta in the brains of APP/PS1 transgenic AD model mice. Cu(II)(gtsm) also decreased the abundance of Abeta trimers and phosphorylated tau, and restored performance of AD mice in the Y-maze test to levels expected for cognitively normal animals. Improvement in the Y-maze correlated directly with decreased Abeta trimer levels. This study demonstrates that increasing intracellular copper bioavailability can restore cognitive function by inhibiting the accumulation of neurotoxic Abeta trimers and phosphorylated tau.


Subject(s)
Amyloid beta-Peptides/drug effects , Copper/pharmacology , tau Proteins/drug effects , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Cognition/drug effects , Copper/pharmacokinetics , Copper/therapeutic use , Dimerization , Disease Models, Animal , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Glycogen Synthase Kinases/antagonists & inhibitors , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organometallic Compounds/pharmacokinetics , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , tau Proteins/metabolism
3.
J Neurochem ; 108(5): 1198-207, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19141082

ABSTRACT

Accumulation of neurotoxic amyloid-beta (Abeta) is central to the pathology of Alzheimer's disease (AD). Elucidating the mechanisms of Abeta accumulation will therefore expedite the development of Abeta-targeting AD therapeutics. We examined activity of an Abeta-degrading protease (matrix metalloprotease 2) to investigate whether biochemical factors consistent with conditions in the AD brain contribute to Abeta accumulation by altering Abeta sensitivity to proteolytic degradation. An Abeta amino acid mutation found in familial AD, Abeta interactions with zinc (Zn), and increased Abeta hydrophobicity all strongly prevented Abeta degradation. Consistent to all of these factors is the promotion of specific Abeta aggregates where the protease cleavage site, confirmed by mass spectrometry, is inaccessible within an amyloid structure. These data indicate decreased degradation due to amyloid formation initiates Abeta accumulation by preventing normal protease activity. Zn also prevented Abeta degradation by the proteases neprilysin and insulin degrading enzyme. Treating Zn-induced Abeta amyloid with the metal-protein attenuating compound clioquinol reversed amyloid formation and restored the peptide's sensitivity to degradation by matrix metalloprotease 2. This provides new data indicating that therapeutic compounds designed to modulate Abeta-metal interactions can inhibit Abeta accumulation by restoring the catalytic potential of Abeta-degrading proteases.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Amyloid/drug effects , Amyloid beta-Peptides/drug effects , Amyloid beta-Peptides/genetics , Clioquinol/pharmacology , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay/methods , Glutamic Acid/genetics , Glutamine/genetics , Humans , Insulysin/pharmacology , Matrix Metalloproteinase 2/metabolism , Microscopy, Electron, Transmission/methods , Mutation , Neprilysin/pharmacology , Peptide Fragments/drug effects , Peptide Fragments/genetics , Peptide Fragments/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Time Factors , Zinc/pharmacology
4.
Int J Biochem Cell Biol ; 40(5): 1030-42, 2008.
Article in English | MEDLINE | ID: mdl-18160328

ABSTRACT

A growing body of evidence supports a central role for biometals in neurodegenerative disorders. Biometals induce oxidative stress through the generation of reactive oxygen species and contribute to neuronal cell dysfunction in Alzheimer's disease (AD), prion disorders and Parkinson's disease (PD). Therapies based on modulation of biometal metabolism are currently being developed and the metal ligand, 5-chloro-7-iodo-8-hydroxyquinoline (clioquinol or CQ) has been investigated for the treatment of AD. CQ has also shown therapeutic benefits in an animal model of PD. However, little is known about the neuroprotective processes of CQ in vivo. In this study, we examined the effect of CQ in BE(2)-M17 human neuroblastoma cells exposed to increased oxidative stress (hydrogen peroxide (H2O2) treatment). Although CQ alone induced a moderate toxic effect on cells, when added to H2O2-treated M17 cells, CQ induced a significant inhibition of H2O2 toxicity. This correlated with up-regulation of phosphoinositol-3-kinase (PI3K) activity in CQ-treated cells. The protective action of CQ was not observed in murine N2a neuroblastoma cells treated with H2O2 and this cell-line did not reveal CQ-mediated increases in PI3K activation. The protective effect was specific for CQ and was not induced by a number of different metal ligands. Inhibition of PI3K activity with LY294002 prevented CQ protection against H2O2 toxicity, demonstrating a crucial role for CQ activation of PI3K in protection against oxidative stress. Furthermore, CQ inhibited H2O2-mediated up-regulation of p53 activity in the M17 cells and this was dependent on PI3K activation. Our studies demonstrate that in human M17 cells, CQ can protect against oxidative stress by activating the PI3K-dependent survival pathway and blocking p53-mediated cell death. These findings have important implications for the development of protective metal ligand-based therapies for treatment of disorders involving oxidative stress.


Subject(s)
Clioquinol/pharmacology , Hydrogen Peroxide/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Cell Line, Tumor , Humans , Hydrogen Peroxide/toxicity , Mice , Up-Regulation
5.
Int J Biochem Cell Biol ; 40(9): 1901-17, 2008.
Article in English | MEDLINE | ID: mdl-18346929

ABSTRACT

The epidermal growth factor receptor is a receptor tyrosine kinase expressed in a range of tissues and cell-types. Activation of the epidermal growth factor receptor by a number of ligands induces downstream signalling that modulates critical cell functions including growth, survival and differentiation. Abnormal epidermal growth factor receptor expression and activation is also involved in a number of cancers. In addition to its cognate ligands, the epidermal growth factor receptor can be activated by metals such as zinc (Zn) and copper (Cu). Due to the important role of these metals in a number of diseases including neurodegenerative disorders, therapeutic approaches are being developed based on the use of lipid permeable metal-complexing molecules. While these agents are showing promising results in animal models and clinical trials, little is known about the effects of metal-ligand complexes on cell signalling pathways. In this study, we investigated the effects of clioquinol (CQ)-metal complexes on activation of epidermal growth factor receptor. We show here that CQ-Cu complexes induced potent epidermal growth factor receptor phosphorylation resulting in downstream activation of extracellular signal-regulated kinase. Similar levels of epidermal growth factor receptor activation were observed with alternative lipid permeable metal-ligands including neocuproine and pyrrolidine dithiocarbamate. We found that CQ-Cu complexes induced a significant reduction in the level of extracellular Abeta1-40 in cell culture. Inhibition of epidermal growth factor receptor activation by PD153035 blocked extracellular signal-regulated kinase phosphorylation and restored Abeta1-40 levels. Activation of the epidermal growth factor receptor by CQ-Cu was mediated through up-regulation of src kinase activity by a cognate ligand-independent process involving membrane integrins. These findings provide the first evidence that metal-ligand complexes can activate the epidermal growth factor receptor with potentially neuroprotective effects.


Subject(s)
Amyloid beta-Peptides/metabolism , Copper/metabolism , ErbB Receptors/metabolism , Extracellular Space/drug effects , Extracellular Space/metabolism , Organometallic Compounds/pharmacology , Animals , Cell Line , Clioquinol/metabolism , Copper/pharmacology , Cricetinae , Enzyme Activation , Epithelial Cells/drug effects , Epithelial Cells/metabolism , ErbB Receptors/agonists , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Integrins/metabolism , Ligands , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Organometallic Compounds/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , Up-Regulation/drug effects , src-Family Kinases/metabolism
6.
J Pharmacol Exp Ther ; 324(1): 360-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17940196

ABSTRACT

Copper has an important role in cancer growth, angiogenesis, and metastasis. Previous studies have shown that cell-permeable metal ligands, including clioquinol (CQ) and pyrrolidine dithiocarbamate, inhibit cancer cell growth in cell culture and in vivo. The mechanism of action has not been fully determined but may involve metal-mediated inhibition of cancer cell proteasome activity. However, these studies do not fully account for the ability of cell-permeable metal ligands to inhibit cancer cell growth without affecting normal cells. In this study, we examined the effect of CQ on macrophage-mediated inhibition of HeLa cancer cell growth in vitro. When CQ was added to RAW 264.7 macrophage-HeLa cell cocultures, a substantial increase in HeLa cell toxicity was observed compared with CQ treatment of HeLa cells cultured alone. Transfer of conditioned medium from CQ-treated macrophages to HeLa cells also induced HeLa cell toxicity, demonstrating the role of secreted factors in the macrophage-mediated effect. Further investigation revealed that CQ induced copper-dependent activation of macrophages and release of tumor necrosis factor (TNF) alpha. In studies with recombinant TNFalpha, we showed that the level of TNFalpha released by CQ-treated macrophages was sufficient to induce HeLa cell toxicity. Moreover, the toxic effect of conditioned medium from CQ-treated macrophages could be prevented by addition of neutralizing antibodies to TNFalpha. These studies demonstrate that CQ can induce cancer cell toxicity through metal-dependent release of TNFalpha from macrophages. Our results may help to explain the targeted inhibition of tumor growth in vivo by CQ.


Subject(s)
Antineoplastic Agents/pharmacology , Clioquinol/pharmacology , Macrophages/drug effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line , Cell Survival/drug effects , Copper/pharmacology , Cytokines/metabolism , HeLa Cells , Humans , Macrophages/metabolism , Mice , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
7.
Biochem J ; 407(3): 435-50, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17680773

ABSTRACT

Biometals have an important role in AD (Alzheimer's disease) and metal ligands have been investigated as potential therapeutic agents for treatment of AD. In recent studies the 8HQ (8-hydroxyquinoline) derivative CQ (clioquinol) has shown promising results in animal models and small clinical trials; however, the actual mode of action in vivo is still being investigated. We previously reported that CQ-metal complexes up-regulated MMP (matrix metalloprotease) activity in vitro by activating PI3K (phosphoinositide 3-kinase) and JNK (c-jun N-terminal kinase), and that the increased MMP activity resulted in enhanced degradation of secreted Abeta (amyloid beta) peptide. In the present study, we have further investigated the biochemical mechanisms by which metal ligands affect Abeta metabolism. To achieve this, we measured the effects of diverse metal ligands on cellular metal uptake and secreted Abeta levels in cell culture. We report that different classes of metal ligands including 8HQ and phenanthroline derivatives and the sulfur compound PDTC (pyrrolidine dithiocarbamate) elevated cellular metal levels (copper and zinc), and resulted in substantial loss of secreted Abeta. Generally, the ability to inhibit Abeta levels correlated with a higher lipid solubility of the ligands and their capacity to increase metal uptake. However, we also identified several ligands that potently inhibited Abeta levels while only inducing minimal change to cellular metal levels. Metal ligands that inhibited Abeta levels [e.g. CQ, 8HQ, NC (neocuproine), 1,10-phenanthroline and PDTC] induced metal-dependent activation of PI3K and JNK, resulting in JNK-mediated up-regulation of metalloprotease activity and subsequent loss of secreted Abeta. The findings in the present study show that diverse metal ligands with high lipid solubility can elevate cellular metal levels resulting in metalloprotease-dependent inhibition of Abeta. Given that a structurally diverse array of ligands was assessed, the results are consistent with the effects being due to metal transport rather than the chelating ligand interacting directly with a receptor.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Copper/metabolism , Peptides/metabolism , Zinc/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/genetics , Animals , Biological Transport, Active/genetics , CHO Cells , Cricetinae , Cricetulus , Humans , Ligands , Oxyquinoline/analogs & derivatives , Oxyquinoline/metabolism , Peptides/genetics , Phenanthrolines/metabolism
8.
Bio Protoc ; 8(17): e2991, 2018 Sep 05.
Article in English | MEDLINE | ID: mdl-34395790

ABSTRACT

Glioma stem cells (GSC) grown as neurospheres exhibit similar characteristics to neural stem cells (NSC) grown as neurospheres, including the ability to self-renew and differentiate. GSCs are thought to play a role in cancer initiation and progression. Self-renewal potential of GSCs is thought to reflect many characteristics associated with malignancy, including tumor recurrence following cytotoxic therapy due to their proliferative dormancy and capacity to allow for the development of resistant tumor cell sub-clones due to mutations acquired during their differentiation. Here, we demonstrate that using extreme limiting dilution analysis (ELDA), subtle differences in the frequency of sphere-forming potential between PI3K-mutant oncogenic NSCs and non-oncogenic NSCs can be measured, in vitro. We further show how ELDA can be used on cells, before and after forced differentiation to amplify inherent differences in sphere-forming potential between mutant and control NSCs. Ultimately, ELDA exploits a difference in the ability of a single or a few seeded stem cells to self-renew, divide and form neurospheres. Importantly, the assay also allows a comparison between genetically distinct cells or between the same cells under different conditions, where the impact of target-specific drugs or other novel cancer stem cell therapies can be tested.

9.
Neuro Oncol ; 20(10): 1344-1355, 2018 09 03.
Article in English | MEDLINE | ID: mdl-29718345

ABSTRACT

Background: Hyperactivation of phosphoinositide 3-kinase (PI3K) signaling is common in cancers, but the precise role of the pathway in glioma biology remains to be determined. Some understanding of PI3K signaling mechanisms in brain cancer comes from studies on neural stem/progenitor cells (NSPCs), where signals transmitted via the PI3K pathway cooperate with other intracellular pathways and downstream transcription factors to regulate critical cell functions. Methods: To investigate the role of the PI3K pathway in glioma initiation and development, we generated a mouse model targeting the inducible expression of a PIK3CAH1047A oncogenic mutant and deletion of the PI3K negative regulator, phosphatase and tensin homolog (PTEN), to NSPCs. Results: Expression of a Pik3caH1047A was sufficient to generate tumors with oligodendroglial features, but simultaneous loss of PTEN was required for the development of invasive, high-grade glioma. Pik3caH1047A-PTEN mutant NSPCs exhibited enhanced neurosphere formation which correlated with increased Wnt signaling, while loss of cAMP response element binding protein (CREB) in Pik3caH1047A-Pten mutant tumors led to longer symptom-free survival in mice. Conclusion: Taken together, our findings present a novel mouse model for glioma demonstrating that the PI3K pathway is important for initiation of tumorigenesis and that disruption of downstream CREB signaling attenuates tumor expansion.


Subject(s)
Brain Neoplasms/pathology , Carcinogenesis/pathology , Cyclic AMP Response Element-Binding Protein/metabolism , Neural Stem Cells/pathology , PTEN Phosphohydrolase/physiology , Phosphatidylinositol 3-Kinases/physiology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Cycle , Cell Movement , Cell Proliferation , Class I Phosphatidylinositol 3-Kinases , Cyclic AMP Response Element-Binding Protein/genetics , Mice , Mice, Knockout , Neural Stem Cells/metabolism , Phosphorylation , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
PLoS One ; 12(2): e0172791, 2017.
Article in English | MEDLINE | ID: mdl-28241049

ABSTRACT

Glioblastoma (GBM) is a heterogeneous tumor of the brain with a poor prognosis due to recurrence and drug resistance following therapy. Genome-wide profiling has revealed the existence of distinct GBM molecular subtypes that respond differently to aggressive therapies. Despite this, molecular subtype does not predict recurrence or drug resistance and overall survival is similar across subtypes. One of the key features contributing to tumor recurrence and resistance to therapy is proposed to be an underlying subpopulation of resistant glioma stem cells (GSC). CD133 expression has been used as a marker of GSCs, however recent evidence suggests the relationship between CD133 expression, GSCs and molecular subtype is more complex than initially proposed. The expression of CD133, Olig2 and CD44 was investigated using patient derived glioma stem-like cells (PDGCs) in vitro and in vivo. Different PDGCs exhibited a characteristic equilibrium of distinct CD133+ and CD44+ subpopulations and the influence of environmental factors on the intra-tumor equilibrium of CD133+ and CD44+ cells in PDGCs was also investigated, with hypoxia inducing a CD44+ to CD133+ shift and chemo-radiotherapy inducing a CD133+ to CD44+ shift. These data suggest that surveillance and modulation of intra-tumor heterogeneity using molecular markers at initial surgery and surgery for recurrent GBM may be important for more effective management of GBM.


Subject(s)
AC133 Antigen/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Hyaluronan Receptors/metabolism , Neoplastic Stem Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Cell Proliferation , Female , Glioblastoma/pathology , Humans , Hypoxia , Mice , Mice, Inbred BALB C , Neoplasm Recurrence, Local , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2 , Phenotype
11.
Cell Death Dis ; 7(12): e2494, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27906173

ABSTRACT

In some cell types, activation of the second messenger cAMP leads to increased expression of proapoptotic Bim and subsequent cell death. We demonstrate that suppression of the cAMP pathway is a common event across many cancers and that pharmacological activation of cAMP in glioblastoma (GBM) cells leads to enhanced BIM expression and apoptosis in specific GBM cell types. We identified the MAPK signaling axis as the determinant of cAMP agonist sensitivity in GBM cells, with high MAPK activity corresponding to cAMP resistance and low activity corresponding to sensitization to cAMP-induced apoptosis. Sensitive cells were efficiently killed by cAMP agonists alone, while targeting both the cAMP and MAPK pathways in resistant GBM cells resulted in efficient apoptosis. We also show that CD44 is differentially expressed in cAMP agonist-sensitive and -resistant cells. We thus propose that CD44 may be a useful biomarker for distinguishing tumors that may be sensitive to cAMP agonists alone or cAMP agonists in combination with other pathway inhibitors. This suggests that using existing chemotherapeutic compounds in combination with existing FDA-approved cAMP agonists may fast track trials toward improved therapies for difficult-to-treat cancers, such as GBM.


Subject(s)
Apoptosis/drug effects , Brain Neoplasms/pathology , Cyclic AMP/agonists , Drug Resistance, Neoplasm , Glioblastoma/pathology , Hyaluronan Receptors/metabolism , MAP Kinase Signaling System , Bcl-2-Like Protein 11/metabolism , Biomarkers, Tumor/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cyclic AMP/metabolism , Humans , Phosphorylation , Protein Isoforms/metabolism , Up-Regulation
12.
PLoS One ; 6(3): e17669, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21412423

ABSTRACT

We have previously demonstrated that brief treatment of APP transgenic mice with metal ionophores (PBT2, Prana Biotechnology) rapidly and markedly improves learning and memory. To understand the potential mechanisms of action underlying this phenomenon we examined hippocampal dendritic spine density, and the levels of key proteins involved in learning and memory, in young (4 months) and old (14 months) female Tg2576 mice following brief (11 days) oral treatment with PBT2 (30 mg/kg/d). Transgenic mice exhibited deficits in spine density compared to littermate controls that were significantly rescued by PBT2 treatment in both the young (+17%, p<0.001) and old (+32%, p<0.001) animals. There was no effect of PBT2 on spine density in the control animals. In the transgenic animals, PBT2 treatment also resulted in significant increases in brain levels of CamKII (+57%, p = 0.005), spinophilin (+37%, p = 0.04), NMDAR1A (+126%, p = 0.02), NMDAR2A (+70%, p = 0.05), pro-BDNF (+19%, p = 0.02) and BDNF (+19%, p = 0.04). While PBT2-treatment did not significantly alter neurite-length in vivo, it did increase neurite outgrowth (+200%, p = 0.006) in cultured cells, and this was abolished by co-incubation with the transition metal chelator, diamsar. These data suggest that PBT2 may affect multiple aspects of snaptic health/efficacy. In Alzheimer's disease therefore, PBT2 may restore the uptake of physiological metal ions trapped within extracellular ß-amyloid aggregates that then induce biochemical and anatomical changes to improve cognitive function.


Subject(s)
Alzheimer Disease/pathology , Dendritic Spines/drug effects , Dendritic Spines/pathology , Ionophores/pharmacology , Metals/pharmacology , Nerve Tissue Proteins/metabolism , Synapses/metabolism , Alzheimer Disease/metabolism , Animals , Cells, Cultured , Clioquinol/analogs & derivatives , Disease Models, Animal , Female , Hippocampus/drug effects , Hippocampus/pathology , Ionophores/administration & dosage , Memory/drug effects , Metals/administration & dosage , Mice , Mice, Transgenic , Neurites/drug effects , Neurites/metabolism , Neurogenesis/drug effects
13.
Free Radic Biol Med ; 48(9): 1152-61, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20138212

ABSTRACT

Ubiquitinated neuronal aggregates containing TDP-43 are pathological hallmarks in the spectrum of frontotemporal lobar dementia (FTLD) and amyotrophic lateral sclerosis (ALS). In affected neurons, TDP-43 undergoes C-terminal fragmentation, phosphorylation, and ubiquitination and forms aggregates in the cytoplasm or nucleus. Although in vitro studies have been able to recapitulate these features using transfected cell culture models, little is known about the biochemical mechanisms that underlie pathological changes to endogenous TDP-43. As altered metal ion homeostasis and increased oxidative stress are central features of neurodegeneration, including FTLD and ALS, we sought to determine the affects of these factors on endogenous TDP-43 metabolism in mammalian cells. Treatment of SY5Y neuronal-like cells expressing endogenous TDP-43 with zinc (Zn) induced depletion of TDP-43 expression and formation of inclusions that were TDP-43 positive. TDP-43 was also detected in the cytosol of Zn-affected cells but this was not aggregated. No evidence of C-terminal fragmentation, phosphorylation, or ubiquitination was observed. The depletion and aggregation of TDP-43 were associated with the specific action of Zn but were not seen with copper, iron, or H(2)O(2). These studies describe for the first time specific induction of endogenous TDP-43 aggregation in neuronal-like cells and suggest that specific Zn-associated processes could affect TDP-43 metabolism in neurodegenerative diseases.


Subject(s)
Chlorides/toxicity , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Inclusion Bodies/drug effects , Neurons/drug effects , Zinc Compounds/toxicity , Animals , Blotting, Western , Cell Line , Cell Survival/drug effects , Fluorescent Antibody Technique , Humans , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Neurons/metabolism , Neurons/pathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats
14.
J Med Chem ; 52(21): 6606-20, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19807095

ABSTRACT

Bis(thiosemicarbazonato) metal complexes (M(II)(btsc)) have demonstrated potential neuroprotective activity in cell and animal models of Alzheimer's disease (AD). Metal complexes can activate the epidermal growth factor receptor (EGFR), leading to inhibition of amyloid peptide accumulation in neuronal cells. As glial cells also have an important role in modulating neuronal health and survival in AD, we examined the effect of M(II)(btsc) on activity of EGFR in an astroglial cell line. Our findings reveal potent activation of glial EGFR by glyoxalbis(N(4)-methylthiosemicarbazonato)Cu(II)] (Cu(II)(gtsm)). Activation of EGFR by Cu(II)(gtsm) involved phosphorylation of multiple tyrosine residues and was mediated by a cognate ligand-independent process involving M(II)(btsc) inhibition of protein tyrosine phosphatase (PTP) activity. EGFR activation resulted in release of growth factors and cytokines with potential modulatory effects on neuronal function. These studies provide an important insight into the mechanism of action of a neuroprotective M(II)(btsc) and provide a basis for future studies into this novel approach to AD therapy.


Subject(s)
Coordination Complexes/chemical synthesis , Copper , ErbB Receptors/metabolism , Neuroglia/drug effects , Neuroprotective Agents/chemical synthesis , Organometallic Compounds/chemical synthesis , Protein Tyrosine Phosphatases/antagonists & inhibitors , Thiosemicarbazones/chemical synthesis , Cell Line , Cell Line, Tumor , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Cytokines/metabolism , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Phosphorylation , Protein Array Analysis , Proto-Oncogene Proteins c-akt/metabolism , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology
15.
Eur Biophys J ; 37(3): 315-21, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18270696

ABSTRACT

Biometals such as copper and zinc have an important role in Alzheimer's disease (AD). Accumulating evidence indicates that copper homeostasis is altered in AD brain with elevated extracellular and low intracellular copper levels. Studies in animals and cell cultures have suggested that increasing intracellular copper can ameliorate AD-like pathology including amyloid deposition and tau phosphorylation. Modulating copper homeostasis can also improve cognitive function in animal models of AD. Treatments are now being developed that may result in redistribution of copper within the brain. Metal ligands such as clioquinol (CQ), DP-109 or pyrrolidine dithiocarbamate (PDTC) have shown promising results in animal models of AD, however, the actual mode of action in vivo has not been fully determined. We previously reported that CQ-metal complexes were able to increase intracellular copper levels in vitro. This resulted in stimulation of phosphoinositol-3-kinase activity and mitogen activated protein kinases (MAPK). Increased kinase activity resulted in up-regulated matrix metalloprotease (MMP2 and MMP3) activity resulting in enhanced degradation of secreted A beta. These findings are consistent with previous studies reporting metal-mediated activation of MAPKs and MMPs. How this activation occurs is unknown but evidence suggests that copper may be able to activate membrane receptors such as the epidermal growth factor receptor (EGFR) and result in downstream activation of MAPK pathways. This has been supported by studies showing metal-mediated activation of EGFR through ligand-independent processes in a number of cell-types. Our initial studies reveal that copper complexes can in fact activate EGFR. However, further studies are necessary to determine if metal complexes such as CQ-copper induce up-regulation of A beta-degrading MMP activity through this mechanism. Elucidation of this pathway may have important implications for the development of metal ligand based therapeutics for treatment of AD and other neurodegenerative disorders.


Subject(s)
Alzheimer Disease/physiopathology , Brain/drug effects , Brain/physiopathology , Chelating Agents/pharmacology , Copper , Matrix Metalloproteinases, Secreted/metabolism , Amyloid/drug effects , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Animals , Brain/enzymology , Cell Culture Techniques , Chelating Agents/therapeutic use , Clioquinol/therapeutic use , Copper/chemistry , Copper/metabolism , Copper/pharmacology , Disease Models, Animal , Egtazic Acid/analogs & derivatives , Egtazic Acid/therapeutic use , Enzyme Activation/drug effects , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Humans , Intracellular Space/metabolism , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Plaque, Amyloid/drug effects , Proline/analogs & derivatives , Proline/therapeutic use , Thiocarbamates/therapeutic use , Trace Elements/metabolism , Zinc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL