Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
J Biol Chem ; 293(47): 18270-18284, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30287689

ABSTRACT

In response to an accumulation of unfolded proteins in the endoplasmic reticulum (ER) lumen, three ER transmembrane signaling proteins, inositol-requiring enzyme 1 (IRE1), PRKR-like ER kinase (PERK), and activating transcription factor 6α (ATF6α), are activated. These proteins initiate a signaling and transcriptional network termed the unfolded protein response (UPR), which re-establishes cellular proteostasis. When this restoration fails, however, cells undergo apoptosis. To investigate cross-talk between these different UPR enzymes, here we developed a high-content live cell screening platform to image fluorescent UPR-reporter cell lines derived from human SH-SY5Y neuroblastoma cells in which different ER stress signaling proteins were silenced through lentivirus-delivered shRNA constructs. We observed that loss of ATF6 expression results in uncontrolled IRE1-reporter activity and increases X box-binding protein 1 (XBP1) splicing. Transient increases in both IRE1 mRNA and IRE1 protein levels were observed in response to ER stress, suggesting that IRE1 up-regulation is a general feature of ER stress signaling and was further increased in cells lacking ATF6 expression. Moreover, overexpression of the transcriptionally active N-terminal domain of ATF6 reversed the increases in IRE1 levels. Furthermore, inhibition of IRE1 kinase activity or of downstream JNK activity prevented an increase in IRE1 levels during ER stress, suggesting that IRE1 transcription is regulated through a positive feed-forward loop. Collectively, our results indicate that from the moment of activation, IRE1 signaling during ER stress has an ATF6-dependent "off-switch."


Subject(s)
Activating Transcription Factor 6/metabolism , Endoplasmic Reticulum Stress , Activating Transcription Factor 6/chemistry , Activating Transcription Factor 6/genetics , Endoplasmic Reticulum Chaperone BiP , Endoribonucleases/genetics , Endoribonucleases/metabolism , Gene Expression Regulation , Humans , Protein Domains , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
3.
J Transl Med ; 14(1): 196, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27369741

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is a leading cause of cancer mortality in the Western world and commonly treated with genotoxic chemotherapy. Stress in the endoplasmic reticulum (ER) was implicated to contribute to chemotherapeutic resistance. Hence, ER stress related protein may be of prognostic or therapeutic significance. METHODS: The expression levels of ER stress proteins calnexin, calreticulin, GRP78 and GRP94 were determined in n = 23 Stage II and III colon cancer fresh frozen tumour and matched normal tissue samples. Data were validated in a cohort of n = 11 rectal cancer patients treated with radiochemotherapy in the neoadjuvant setting. The calnexin gene was silenced using siRNA in HCT116 cells. RESULTS: There were no increased levels of ER stress proteins in tumour compared to matched normal tissue samples in Stage II or III CRC. However, increased calnexin protein levels were predictive of poor clinical outcome in the patient cohort. Data were validated in the rectal cancer cohort treated in the neoadjuvant setting. Calnexin gene-silencing significantly reduced cell survival and increased cancer cell susceptibility to 5FU chemotherapy. CONCLUSION: Increased tumour protein levels of calnexin may be of prognostic significance in CRC, and calnexin may represent a potential target for future therapies.


Subject(s)
Biomarkers, Tumor/metabolism , Calnexin/metabolism , Colorectal Neoplasms/metabolism , Endoplasmic Reticulum/metabolism , Molecular Targeted Therapy , Cell Death/drug effects , Cell Survival/drug effects , Clone Cells , Colorectal Neoplasms/pathology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Fluorouracil/pharmacology , Gene Knockdown Techniques , Gene Silencing/drug effects , HCT116 Cells , Humans , Immunohistochemistry , Neoadjuvant Therapy , Neoplasm Staging , Prognosis , Rectal Neoplasms/therapy , Treatment Outcome
4.
Eur J Pharmacol ; 780: 53-64, 2016 Jun 05.
Article in English | MEDLINE | ID: mdl-26997367

ABSTRACT

Deregulation of the ubiquitin-proteasome pathway has been frequently observed in a number of malignancies. Using quantitative Western blotting of normal and matched tumour tissue, we here identified a significant increase in the 19S proteasome subunit Rpt4 in response to chemoradiation in locally advanced rectal cancer patients with unfavourable outcome. We therefore explored the potential of Rpt4 reduction as a therapeutic strategy in colorectal cancer (CRC). Utilizing siRNA to down regulate Rpt4 expression, we show that silencing of Rpt4 reduced proteasomal activity and induced endoplasmic reticulum stress. Gene silencing of Rpt4 also inhibited cell proliferation, reduced clonogenic survival and induced apoptosis in HCT-116 colon cancer cells. We next developed a cell penetrating peptide-based nanoparticle delivery system to achieve in vivo gene silencing of Rpt4. Administration of Rpt4 siRNA nanoparticles reduced tumour growth and improved survival in a HCT-116 colon cancer xenograft tumour model in vivo. Collectively, our data suggest that inhibition of Rpt4 represents a novel strategy for the treatment of CRC.


Subject(s)
Colorectal Neoplasms/enzymology , Colorectal Neoplasms/therapy , Molecular Targeted Therapy , Proteasome Endopeptidase Complex/metabolism , Apoptosis/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Drug Carriers/chemistry , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Gene Silencing , HCT116 Cells , Humans , Nanoparticles/chemistry , Proteasome Endopeptidase Complex/deficiency , Proteasome Endopeptidase Complex/genetics , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Treatment Failure
5.
Eur J Neurosci ; 43(5): 640-52, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26750440

ABSTRACT

Disturbance of homeostasis within the endoplasmic reticulum (ER) lumen leads to the accumulation of unfolded and misfolded proteins. This results in the activation of an evolutionary conserved stress response termed ER stress that, if unresolved, induces apoptosis. Previously the Bcl-2 homology domain 3-Only Protein Puma was identified as a mediator of ER stress-induced apoptosis in neurons. In the search of alternative contributors to ER stress-induced apoptosis, a downregulation of the anti-apoptotic Bcl-2 family protein Mcl-1 was noted during ER stress in both mouse cortical neurons and human SH-SY5Y neuroblastoma cells. Downregulation of Mcl-1 was associated with an upregulation of microRNA-29a (miR-29a) expression, and subsequent experiments showed that miR-29a targeted the 3'-untranslated region of the anti-apoptotic Bcl-2 family protein, Mcl-1. Inhibition of miR-29a expression using sequence-specific antagomirs or the overexpression of Mcl-1 decreased cell death following tunicamycin treatment, while gene silencing of Mcl-1 increased cell death. miR-29a did not alter the signalling branches of the ER stress response, rather its expression was controlled by the ER stress-induced transcription factor activating-transcription-factor-4 (ATF4). The current data demonstrate that the ATF4-mediated upregulation of miR-29a enhances the sensitivity of neurons to ER stress-induced apoptosis.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , MicroRNAs/genetics , Neurons/metabolism , Up-Regulation , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cells, Cultured , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
6.
J Mol Neurosci ; 53(2): 231-41, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24696165

ABSTRACT

Endoplasmic reticulum (ER) stress has been implicated in a number of neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS). MicroRNAs are small ribonucleic acids which can modulate protein expression by binding to the 3'UTR of target mRNAs. We recently identified increased miR-29a expression in response to ER stress in neurons, with members of the miR-29 family implicated in cancer and neurodegeneration. We found high expression of miR-29a in the mouse brain and spinal cord by quantitative PCR analysis and increased expression of miR-29a in the spinal cord of SOD1(G93A) transgenic mice, a mouse model of familial ALS. In situ hybridisation experiments revealed increased miR-29a expression in the lumbar spinal cord of SOD1(G93A) transgenic mice from postnatal day 70 onward when compared to wild-type mice. miR-29a knockdown was achieved in the CNS in vivo after a single intracerebroventricular injection of a miR-29a-specific antagomir. While analysis of disease progression and motor function could not identify a significant alteration in ALS disease manifestations, a trend towards increased lifespan was observed in male SOD1(G93A) mice. These findings demonstrate that miR-29a may act as a marker for disease progression in SOD1(G93A) mice, and provide first proof-of-concept for a therapeutic modulation of miR-29a function in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , MicroRNAs/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/drug effects , Brain/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Oligoribonucleotides/pharmacology , Spinal Cord/drug effects , Spinal Cord/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
7.
J Cell Sci ; 127(Pt 3): 609-19, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24284067

ABSTRACT

Targeting the proteasome is a valuable approach for cancer therapy, potentially limited by pro-survival pathways that are induced in parallel to cell death. Whether these pro-survival pathways are activated in all cells, show different activation kinetics in sensitive versus resistant cells or interact functionally with cell death pathways is unknown. We monitored activation of the heat-shock response (HSR), a key survival pathway induced by proteasome inhibition, relative to apoptosis activation in HCT116 colon cancer cells expressing enhanced green fluorescent protein (EGFP) under the control of the HSP70 promoter. Single-cell and high-content time-lapse imaging of epoxomicin treatment revealed that neither basal activity nor the time of onset of the HSR differed between resistant and sensitive populations. However, resistant cells had significantly higher and prolonged reporter activity than those that succumbed to cell death. p53 deficiency protected against cell death but failed to modulate the HSR. By contrast, inhibition of the HSR significantly increased the cytotoxicity of epoxomicin. Our data provide novel insights into the kinetics and heterogeneity of the HSR during proteasome inhibition, suggesting that the HSR modulates cell death signalling unidirectionally.


Subject(s)
Apoptosis/genetics , Colonic Neoplasms/pathology , HSP70 Heat-Shock Proteins/genetics , Single-Cell Analysis , Cell Survival , Colonic Neoplasms/metabolism , HCT116 Cells , HSP70 Heat-Shock Proteins/ultrastructure , Heat-Shock Response/genetics , Humans , Time-Lapse Imaging
8.
Brain ; 136(Pt 2): 577-92, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23361066

ABSTRACT

Hippocampal sclerosis is a frequent pathological finding in patients with temporal lobe epilepsy and can be caused by prolonged single or repeated brief seizures. Both DNA damage and endoplasmic reticulum stress have been implicated as underlying molecular mechanisms in seizure-induced brain injury. The CCAAT/enhancer-binding protein homologous protein (CHOP) is a transcriptional regulator induced downstream of DNA damage and endoplasmic reticulum stress, which can promote or inhibit apoptosis according to context. Recent work has proposed inhibition of CHOP as a suitable neuroprotective strategy. Here, we show that transcript and protein levels of CHOP increase in surviving subfields of the hippocampus after prolonged seizures (status epilepticus) in mouse models. CHOP was also elevated in the hippocampus from epileptic mice and patients with pharmacoresistant epilepsy. The hippocampus of CHOP-deficient mice was much more vulnerable to damage in mouse models of status epilepticus. Moreover, compared with wild-type animals, CHOP-deficient mice subject to status epilepticus developed more spontaneous seizures, displayed protracted hippocampal neurodegeneration and a deficit in a hippocampus-dependent object-place recognition task. The absence of CHOP was associated with a supra-maximal induction of p53 after status epilepticus, and inhibition of p53 abolished the cell death-promoting consequences of CHOP deficiency. The protective effect of CHOP could be partly explained by activating transcription of murine double minute 2 that targets p53 for degradation. These data demonstrate that CHOP is required for neuronal survival after seizures and caution against inhibition of CHOP as a neuroprotective strategy where excitotoxicity is an underlying pathomechanism.


Subject(s)
Neurons/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Seizures/metabolism , Transcription Factor CHOP/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Cell Survival/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/pathology , Proto-Oncogene Proteins c-mdm2/physiology , Seizures/genetics , Seizures/pathology , Tumor Suppressor Protein p53/physiology
9.
Cancer Res ; 73(2): 519-28, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23329644

ABSTRACT

Apoptotic desensitization is a hallmark of cancer cells, but present knowledge of molecular systems controlling apoptosis has yet to provide significant prognostic insights. Here, we report findings from a systems study of the intrinsic pathway of apoptosis by BCL2 family proteins and clinical translation of its findings into a model with applications in colorectal cancer (CRC). By determining absolute protein quantifications in CRC cells and patient tumor samples, we found that BAK and BAX were expressed more highly than their antiapoptotic inhibitors. This counterintuitive finding suggested that sole inhibition of effector BAX and BAK could not be sufficient for systems stability in nonstressed cells. Assuming a model of direct effector activation by BH3-only proteins, we calculated that the amount of stress-induced BH3-only proteins required to activate mitochondrial apoptosis could predict individual death responses of CRC cells to 5-fluorouracil/oxaliplatin. Applying this model predictor to protein profiles in tumor and matched normal tissue samples from 26 patients with CRCs, we found that differences in protein quantities were sufficient to model the increased tumor sensitivity to chemotherapy compared with normal tissue. In addition, these differences were sufficient to differentiate clinical responders from nonresponders with high confidence. Applications of our model, termed DR_MOMP, were used to assess the impact of apoptosis-sensitizing drugs in lowering the necessary dose of state-of-the-art chemotherapy in individual patients. Together, our findings offer a ready clinical tool with the potential to tailor chemotherapy to individual patients.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Systems Analysis , Aged , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Computational Biology , Female , Humans , Male , Models, Biological , Protein Array Analysis , Treatment Outcome , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
10.
J Neurochem ; 124(5): 721-34, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23199202

ABSTRACT

Neuronal preconditioning is a phenomenon where a previous exposure to a sub-lethal stress stimulus increases the resistance of neurons towards a second, normally lethal stress stimulus. Activation of the energy stress sensor, AMP-activated protein kinase (AMPK) has been shown to contribute to the protective effects of ischaemic and mitochondrial uncoupling-induced preconditioning in neurons, however, the molecular basis of AMPK-mediated preconditioning has been less well characterized. We investigated the effect of AMPK preconditioning using 5-aminoimidazole-4-carboxamide riboside (AICAR) in a model of NMDA-mediated excitotoxic injury in primary mouse cortical neurons. Activation of AMPK with low concentrations of AICAR (0.1 mM for 2 h) induced a transient increase in AMPK phosphorylation, protecting neurons against NMDA-induced excitotoxicity. Analysing potential targets of AMPK activation, demonstrated a marked increase in mRNA expression and protein levels of the anti-apoptotic BCL-2 family protein myeloid cell leukaemia sequence 1 (MCL-1) in AICAR-preconditioned neurons. Interestingly, over-expression of MCL-1 protected neurons against NMDA-induced excitotoxicity while MCL-1 gene silencing abolished the effect of AICAR preconditioning. Monitored intracellular Ca²âº levels during NMDA excitation revealed that MCL-1 over-expressing neurons exhibited improved bioenergetics and markedly reduced Ca²âº elevations, suggesting a potential mechanism through which MCL-1 confers neuroprotection. This study identifies MCL-1 as a key effector of AMPK-induced preconditioning in neurons.


Subject(s)
Adaptation, Physiological/physiology , Cerebral Cortex/metabolism , Neurons/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , AMP-Activated Protein Kinase Kinases , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Blotting, Western , Cerebral Cortex/drug effects , Flow Cytometry , Hypoglycemic Agents/pharmacology , Mice , Microscopy, Confocal , Myeloid Cell Leukemia Sequence 1 Protein , N-Methylaspartate/toxicity , Neurons/drug effects , Ribonucleotides/pharmacology , Stress, Physiological/physiology
11.
J Neurosci ; 32(15): 5024-38, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22496549

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an incurable neurodegenerative disorder affecting motoneurons. Mutations in angiogenin, encoding a member of the pancreatic RNase A superfamily, segregate with ALS. We previously demonstrated that angiogenin administration shows promise as a neuroprotective therapeutic in studies using transgenic ALS mice and primary motoneuron cultures. Its mechanism of action and target cells in the spinal cord, however, are largely unknown. Using mixed motoneuron cultures, motoneuron-like NSC34 cells, and primary astroglia cultures as model systems, we here demonstrate that angiogenin is a neuronally secreted factor that is endocytosed by astroglia and mediates neuroprotection in paracrine. We show that wild-type angiogenin acts unidirectionally to induce RNA cleavage in astroglia, while the ALS-associated K40I mutant is also secreted and endocytosed, but fails to induce RNA cleavage. Angiogenin uptake into astroglia requires heparan sulfate proteoglycans, and engages clathrin-mediated endocytosis. We show that this uptake mechanism exists for mouse and human angiogenin, and delivers a functional RNase output. Moreover, we identify syndecan 4 as the angiogenin receptor mediating the selective uptake of angiogenin into astroglia. Our data provide new insights into the paracrine activities of angiogenin in the nervous system, and further highlight the critical role of non-neuronal cells in the pathogenesis of ALS.


Subject(s)
Astrocytes/metabolism , Astrocytes/physiology , Motor Neurons/metabolism , RNA Cleavage/physiology , Ribonuclease, Pancreatic/metabolism , Ribonuclease, Pancreatic/physiology , Animals , Blotting, Western , Cell Survival/physiology , Cells, Cultured , Clathrin/physiology , Culture Media, Conditioned , Endocytosis/physiology , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents , Paracrine Communication/physiology , Protein Binding , Real-Time Polymerase Chain Reaction , Syndecan-4/metabolism , Transfection
12.
Biochim Biophys Acta ; 1817(5): 744-53, 2012 May.
Article in English | MEDLINE | ID: mdl-22336583

ABSTRACT

The preconditioning response conferred by a mild uncoupling of the mitochondrial membrane potential (Δψ(m)) has been attributed to altered reactive oxygen species (ROS) production and mitochondrial Ca(2+) uptake within the cells. Here we have explored if altered cellular energetics in response to a mild mitochondrial uncoupling stimulus may also contribute to the protection. The addition of 100 nM FCCP for 30 min to cerebellar granule neurons (CGNs) induced a transient depolarization of the Δψ(m), that was sufficient to significantly reduce CGN vulnerability to the excitotoxic stimulus, glutamate. On investigation, the mild mitochondrial 'uncoupling' stimulus resulted in a significant increase in the plasma membrane levels of the glucose transporter isoform 3, with a hyperpolarisation of Δψ(m) and increased cellular ATP levels also evident following the washout of FCCP. Furthermore, the phosphorylation state of AMP-activated protein kinase (AMPK) (Thr 172) was increased within 5 min of the uncoupling stimulus and elevated up to 1h after washout. Significantly, the physiological changes and protection evident after the mild uncoupling stimulus were lost in CGNs when AMPK activity was inhibited. This study identifies an additional mechanism through which protection is mediated upon mild mitochondrial uncoupling: it implicates increased AMPK signalling and an adaptive shift in energy metabolism as mediators of the preconditioning response associated with FCCP-induced mild mitochondrial uncoupling.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cytoprotection/drug effects , Mitochondria/metabolism , Neurons/cytology , Neurons/enzymology , Neurotoxins/toxicity , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cerebellum/cytology , Energy Metabolism , Enzyme Activation/drug effects , Glutamic Acid/toxicity , Intracellular Space/drug effects , Intracellular Space/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Neurons/drug effects , Stress, Physiological/drug effects
13.
Gut ; 61(5): 725-33, 2012 May.
Article in English | MEDLINE | ID: mdl-22082587

ABSTRACT

OBJECTIVE: Key to the clinical management of colorectal cancer is identifying tools which aid in assessing patient prognosis and determining more effective and personalised treatment strategies. We evaluated whether an experimental systems biology strategy which analyses the susceptibility of cancer cells to undergo caspase activation can be exploited to predict patient responses to 5-fluorouracil-based chemotherapy and to case-specifically identify potential alternative targeted treatments to reactivate apoptosis. DESIGN: We quantified five essential apoptosis-regulating proteins (Pro-Caspases 3 and 9, APAF-1, SMAC and XIAP) in samples of Stage II (n = 13) and III (n=17) tumour and normal colonic (n = 8) tissue using absolute quantitative immunoblotting and employed systems simulations of apoptosis signalling to predict the susceptibility of tumour cells to execute apoptosis. Additional systems analyses assessed the efficacy of novel apoptosis-inducing therapeutics such as XIAP antagonists, proteasome inhibitors and Pro-Caspase-3-activating compounds in restoring apoptosis execution in apoptosis-incompetent tumours. RESULTS: Comparisons of caspase activity profiles demonstrated that the likelihood of colorectal tumours to undergo apoptosis decreases with advancing disease stage. Systems-level analysis correctly predicted positive or negative outcome in 85% (p=0.004) of colorectal cancer patients receiving 5-fluorouracil based chemotherapy and significantly outperformed common uni- and multi-variate statistical approaches. Modelling of individual patient responses to novel apoptosis-inducing therapeutics revealed markedly different inter-individual responses. CONCLUSIONS: Our study represents the first proof-of-concept example demonstrating the significant clinical potential of systems biology-based approaches for predicting patient outcome and responsiveness to novel targeted treatment paradigms.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Apoptosis/physiology , Colorectal Neoplasms/drug therapy , Decision Support Techniques , Fluorouracil/therapeutic use , Models, Biological , Systems Biology , Aged , Apoptosis/drug effects , Biomarkers/metabolism , Caspases/metabolism , Chemotherapy, Adjuvant , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/physiopathology , Female , Humans , Logistic Models , Male , Middle Aged , Molecular Targeted Therapy , Neoplasm Staging , Principal Component Analysis , Treatment Outcome
14.
Neurobiol Aging ; 33(9): 2200-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-21803450

ABSTRACT

Here we employed human SHEP neuroblastoma cells either stably or inducibly expressing the amyloid precursor protein (APP) intracellular domain (AICD) to investigate its ability to modulate stress-induced cell death. Analysis of effector caspase activation revealed that AICD overexpression was specifically associated with an increased sensitivity to apoptosis induced by the 2 endoplasmic reticulum (ER) stressors thapsigargin and tunicamycin, but not by staurosporine (STS). Basal and ER stress-induced expression of Bip/Grp78 and C/EBP-homologous protein/GADD153 were not altered by AICD implying that AICD potentiated cell death downstream or independent of the conserved unfolded protein response (UPR). Interestingly, quantitative polymerase chain reaction analysis and reporter gene assays revealed that AICD significantly downregulated messenger RNA levels of the Alzheimer's disease susceptibility gene ApoJ/clusterin, indicating transcriptional repression. Knockdown of ApoJ/clusterin mimicked the effect of AICD on ER stress-induced apoptosis, but had no discernible effect on staurosporine-induced cell death. Our data suggest that altered levels of AICD may abolish the prosurvival function of ApoJ/clusterin and increase the susceptibility of neurons to ER stress-mediated cell death, a pathway that may contribute to the pathogenesis of Alzheimer's disease.


Subject(s)
Apoptosis/drug effects , Cytidine Deaminase/pharmacology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Cell Line, Tumor , Clusterin/genetics , Clusterin/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Endoplasmic Reticulum Chaperone BiP , Enzyme Inhibitors/pharmacology , Flow Cytometry , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Neuroblastoma , RNA, Messenger/metabolism , Signal Transduction/drug effects , Thapsigargin/pharmacology , Time Factors , Transfection , Tunicamycin/pharmacology , Unfolded Protein Response/drug effects
15.
Int J Cancer ; 131(4): E494-500, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-21960357

ABSTRACT

Colorectal cancer is a leading cause of cancer-related deaths worldwide. Early diagnosis and treatment of colorectal cancer is the key to improving survival rates and as such a need exists to identify patients who may benefit from adjuvant chemotherapy. The dysregulation of the ubiquitin-proteasome system (UPS) has been implicated in oncogenesis and cancer cell survival, and proteasome inhibitors are in clinical use for a number of malignancies including multiple myeloma. In our study, we examined the protein expression of several key components of the UPS in colorectal cancer using immunohistochemistry to determine expression levels of ubiquitinylated proteins and the proteasomal subunits, 20S core and Rpt4 in a cohort of 228 patients with colon cancer. Multivariate Cox analysis revealed that neither the intensity of either ubiquitinylated proteins or the 20S core was predictive in either Stage II or III colon cancer for disease free survival or overall survival. In contrast, in Stage II patients increased Rpt4 staining was significantly associated with disease free survival (Cox proportional hazard ratio 0.605; p = 0.0217). Our data suggest that Rpt4 is an independent prognostic variable for Stage II colorectal cancer and may aid in the decision of which patients undergo adjuvant chemotherapy.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Aged , Colorectal Neoplasms/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Multivariate Analysis , Prognosis
16.
J Biol Chem ; 286(29): 25719-28, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21628466

ABSTRACT

Inactivating mutations in the transcription factor hepatocyte nuclear factor (HNF) 1A cause HNF1A-maturity-onset diabetes of the young (HNF1A-MODY), the most common monogenic form of diabetes. To examine HNF1A-MODY-induced defects in gene expression, we performed a microarray analysis of the transcriptome of rat INS-1 cells inducibly expressing the common hot spot HNF1A frameshift mutation, Pro291fsinsC-HNF1A. Real-time quantitative PCR (qPCR), Western blotting, immunohistochemistry, reporter assays, and chromatin immunoprecipitation (ChIP) were used to validate alterations in gene expression and to explore biological activities of target genes. Twenty-four hours after induction of the mutant HNF1A protein, we identified a prominent down-regulation of the bone morphogenetic protein 3 gene (Bmp-3) mRNA expression. Reporter assays, qPCR, and Western blot analysis validated these results. In contrast, inducible expression of wild-type HNF1A led to a time-dependent increase in Bmp-3 mRNA and protein levels. Moreover, reduced protein levels of BMP-3 and insulin were detected in islets of transgenic HNF1A-MODY mice. Interestingly, treatment of naïve INS-1 cells or murine organotypic islet cultures with recombinant human BMP-3 potently increased their insulin levels and restored the decrease in SMAD2 phosphorylation and insulin gene expression induced by the HNF1A frameshift mutation. Our study suggests a critical link between HNF1A-MODY-induced alterations in Bmp-3 expression and insulin gene levels in INS-1 cells and indicates that the reduced expression of growth factors involved in tissue differentiation may play an important role in the pathophysiology of HNF1A-MODY.


Subject(s)
Bone Morphogenetic Protein 3/pharmacology , Down-Regulation/drug effects , Frameshift Mutation/drug effects , Hepatocyte Nuclear Factor 1-alpha/genetics , Insulin/genetics , Animals , Cell Line, Tumor , Down-Regulation/genetics , Gene Expression Profiling , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Mice , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Rats
17.
Eur J Neurosci ; 33(3): 401-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21198986

ABSTRACT

Bcl-2 homology domain 3 (BH3)-only proteins are pro-apoptotic Bcl-2 family members that play important roles in upstream cell death signalling during apoptosis. Proteasomal stress has been shown to contribute to the pathology of cerebral ischaemia and many neurodegenerative disorders. Here we explored the contribution of BH3-only proteins in mediating proteasome-inhibition-induced apoptosis in the murine brain in vivo. Stereotactic intrahippocampal microinjection of the selective proteasome inhibitor epoxomicin (2.5 nmol) induced a delayed apoptosis within only the CA1 hippocampal neurons and not neurons within the CA3 or dentate gyrus regions, a selective vulnerability similar to that seen during ischaemia. This injury developed over a time-course of 3 days and was characterized by positive terminal deoxynucleotidyl transferase dUTP nick end labelling staining and nuclear condensation. Previous work from our laboratory has identified the BH3-only protein p53-upregulated mediator of apoptosis (Puma) as mediating proteasome-inhibition-induced apoptosis in cultured neural cells. Genetic deletion of puma reduced the number of terminal deoxynucleotidyl transferase dUTP nick end labelling-positive cells within the CA1 following epoxomicin microinjection but it did not provide a complete protection. Subsequent studies identified the BH3-only protein Bim as also being upregulated during proteasome inhibition in organotypic hippocampal slice cultures and after epoxomicin treatment in vivo. Interestingly, the genetic deletion of bim also afforded significant neuroprotection, although this protection was less pronounced. In summary, we demonstrate that the BH3-only proteins Puma and Bim mediate the delayed apoptosis of CA1 hippocampal neurons induced by proteasome inhibition in vivo, and that either BH3-only protein can only partly compensate for the deficiency of the other.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , Hippocampus/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Proteasome Inhibitors , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Blotting, Western , Hippocampus/drug effects , Hippocampus/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/pathology , Oligopeptides/toxicity , Proto-Oncogene Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Proteins/genetics
18.
Antioxid Redox Signal ; 14(10): 1863-76, 2011 May 15.
Article in English | MEDLINE | ID: mdl-20712420

ABSTRACT

5'-Adenosine monophosphate (AMP)-activated protein kinase (AMPK) is a key sensor of cellular energy status. AMPK signaling regulates energy balance at the cellular, organ, and whole-body level. More recently, it has become apparent that AMPK plays also an important role in long-term decisions that determine cell fate, in particular cell cycle progression and apoptosis activation. Here, we describe the diverse mechanisms of AMPK activation and the role of AMPK in the regulation of cellular energy balance. We summarize recent studies implicating AMPK activation in the regulation of neuronal survival and as a key player during ischemic stroke. We also suggest that AMPK activation may have dual functions in the regulation of neuronal survival: AMPK provides a protective effect during transient energy depletion as exemplified in a model of neuronal Ca(2+) overloading, and this effect is partially mediated by the activation of neuronal glucose transporter 3. Prolonged AMPK activation, on the contrary, can lead to neuronal apoptosis via the transcriptional activation of the proapoptotic Bcl-2 family member, bim. Molecular switches that determine the protective versus cell death-inducing effects of AMPK activation are discussed.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cell Survival/physiology , Neurons/enzymology , Neurons/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Survival/genetics , Humans
19.
Mol Cell Biol ; 30(23): 5484-501, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20921277

ABSTRACT

Proteasomal stress and the accumulation of polyubiquitinated proteins are key features of numerous neurodegenerative disorders. Previously we demonstrated that stabilization of p53 and activation of its target gene, puma (p53-upregulated mediator of apoptosis), mediated proteasome inhibitor-induced apoptosis in cancer cells. Here we demonstrated that Puma also contributed to proteasome inhibitor-induced apoptosis in mouse neocortical neurons. Although protection afforded by puma gene deletion was incomplete, we found little evidence indicating contributions from other proapoptotic BH3-only proteins. Attenuation of bax expression did not further reduce Puma-independent apoptosis, suggesting that pathways other than the mitochondrial apoptosis pathway were activated. Real-time imaging experiments in wild-type and puma-deficient neurons using a fluorescence resonance energy transfer (FRET)-based caspase sensor confirmed the involvement of a second cell death pathway characterized by caspase activation prior to mitochondrial permeabilization and, more prominently, a third, caspase-independent and Puma-independent pathway characterized by rapid cell shrinkage and nuclear condensation. This pathway involved lysosomal permeabilization in the absence of autophagy activation and was sensitive to cathepsin but not autophagy inhibition. Our data demonstrate that proteasomal stress activates distinct cell death pathways in neurons, leading to both caspase-dependent and caspase-independent apoptosis, and demonstrate independent roles for Puma and lysosomal permeabilization in this model.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Proteasome Inhibitors , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Autophagy , BH3 Interacting Domain Death Agonist Protein/deficiency , BH3 Interacting Domain Death Agonist Protein/genetics , Base Sequence , Bcl-2-Like Protein 11 , Caspase 3/metabolism , Cathepsins/metabolism , Cytochromes c/metabolism , DNA Primers/genetics , Fluorescence Resonance Energy Transfer , Gene Expression , Lysosomes/drug effects , Lysosomes/metabolism , Membrane Potential, Mitochondrial , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Neurological , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/drug effects , Protease Inhibitors/pharmacology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/genetics , Stress, Physiological , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
20.
J Biol Chem ; 285(46): 36199-206, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20841353

ABSTRACT

Heterozygous loss-of-function mutations in the hepatocyte nuclear factor 1A (HNF1A) gene result in the pathogenesis of maturity-onset diabetes-of-the-young type 3, (HNF1A-MODY). This disorder is characterized by a primary defect in metabolism-secretion coupling and decreased beta cell mass, attributed to excessive beta cell apoptosis. Here, we investigated the link between energy stress and apoptosis activation following HNF1A inactivation. This study employed single cell fluorescent microscopy, flow cytometry, gene expression analysis, and gene silencing to study the effects of overexpression of dominant-negative (DN)-HNF1A expression on cellular bioenergetics and apoptosis in INS-1 cells. Induction of DN-HNF1A expression led to reduced ATP levels and diminished the bioenergetic response to glucose. This was coupled with activation of the bioenergetic stress sensor AMP-activated protein kinase (AMPK), which preceded the onset of apoptosis. Pharmacological activation of AMPK using aminoimidazole carboxamide ribonucleotide (AICAR) was sufficient to induce apoptosis in naive cells. Conversely, inhibition of AMPK with compound C or AMPKα gene silencing protected against DN-HNF1A-induced apoptosis. Interestingly, AMPK mediated the induction of the pro-apoptotic Bcl-2 homology domain-3-only protein Bmf (Bcl-2-modifying factor). Bmf expression was also elevated in islets of DN-HNF1A transgenic mice. Furthermore, knockdown of Bmf expression in INS-1 cells using siRNA was sufficient to protect against DN-HNF1A-induced apoptosis. Our study suggests that overexpression of DN-HNF1A induces bioenergetic stress and activation of AMPK. This in turn mediates the transcriptional activation of the pro-apoptotic Bcl-2-homology protein BMF, coupling prolonged energy stress to apoptosis activation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/physiology , Energy Metabolism/physiology , AMP-Activated Protein Kinases/genetics , Adaptor Proteins, Signal Transducing/genetics , Adenosine Triphosphate/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Doxycycline/pharmacology , Energy Metabolism/drug effects , Enzyme Activation/drug effects , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hypoglycemic Agents/pharmacology , Insulinoma/genetics , Insulinoma/metabolism , Insulinoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Interference , Rats , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleotides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...