Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Cell Biol ; 25(4): 550-564, 2023 04.
Article in English | MEDLINE | ID: mdl-36894671

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the RNA virus responsible for the coronavirus disease 2019 (COVID-19) pandemic. Although SARS-CoV-2 was reported to alter several cellular pathways, its impact on DNA integrity and the mechanisms involved remain unknown. Here we show that SARS-CoV-2 causes DNA damage and elicits an altered DNA damage response. Mechanistically, SARS-CoV-2 proteins ORF6 and NSP13 cause degradation of the DNA damage response kinase CHK1 through proteasome and autophagy, respectively. CHK1 loss leads to deoxynucleoside triphosphate (dNTP) shortage, causing impaired S-phase progression, DNA damage, pro-inflammatory pathways activation and cellular senescence. Supplementation of deoxynucleosides reduces that. Furthermore, SARS-CoV-2 N-protein impairs 53BP1 focal recruitment by interfering with damage-induced long non-coding RNAs, thus reducing DNA repair. Key observations are recapitulated in SARS-CoV-2-infected mice and patients with COVID-19. We propose that SARS-CoV-2, by boosting ribonucleoside triphosphate levels to promote its replication at the expense of dNTPs and by hijacking damage-induced long non-coding RNAs' biology, threatens genome integrity and causes altered DNA damage response activation, induction of inflammation and cellular senescence.


Subject(s)
COVID-19 , Animals , Mice , SARS-CoV-2 , Cellular Senescence , DNA Damage
2.
EMBO Rep ; 23(2): e53658, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34854526

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes the coronavirus disease 2019 (COVID-19), known to be more common in the elderly, who also show more severe symptoms and are at higher risk of hospitalization and death. Here, we show that the expression of the angiotensin converting enzyme 2 (ACE2), the SARS-CoV-2 cell receptor, increases during aging in mouse and human lungs. ACE2 expression increases upon telomere shortening or dysfunction in both cultured mammalian cells and in vivo in mice. This increase is controlled at the transcriptional level, and Ace2 promoter activity is DNA damage response (DDR)-dependent. Both pharmacological global DDR inhibition of ATM kinase activity and selective telomeric DDR inhibition by the use of antisense oligonucleotides prevent Ace2 upregulation following telomere damage in cultured cells and in mice. We propose that during aging telomere dysfunction due to telomeric shortening or damage triggers DDR activation and this causes the upregulation of ACE2, the SARS-CoV-2 cell receptor, thus contributing to make the elderly more susceptible to the infection.


Subject(s)
Aging , Angiotensin-Converting Enzyme 2/genetics , COVID-19 , DNA Damage , Telomere , Aged , Aging/genetics , Animals , Humans , Mice , SARS-CoV-2 , Telomere/genetics
3.
Nat Commun ; 10(1): 4887, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31653834

ABSTRACT

Accumulation of DNA lesions causing transcription stress is associated with natural and accelerated aging and culminates with profound metabolic alterations. Our understanding of the mechanisms governing metabolic redesign upon genomic instability, however, is highly rudimentary. Using Ercc1-defective mice and Xpg knock-out mice, we demonstrate that combined defects in transcription-coupled DNA repair (TCR) and in nucleotide excision repair (NER) directly affect bioenergetics due to declined transcription, leading to increased ATP levels. This in turn inhibits glycolysis allosterically and favors glucose rerouting through the pentose phosphate shunt, eventually enhancing production of NADPH-reducing equivalents. In NER/TCR-defective mutants, augmented NADPH is not counterbalanced by increased production of pro-oxidants and thus pentose phosphate potentiation culminates in an over-reduced redox state. Skin fibroblasts from the TCR disease Cockayne syndrome confirm results in animal models. Overall, these findings unravel a mechanism connecting DNA damage and transcriptional stress to metabolic redesign and protective antioxidant defenses.


Subject(s)
Adenosine Triphosphate/metabolism , Antioxidants/metabolism , DNA Damage/genetics , DNA Repair/genetics , Glycolysis/physiology , NADP/metabolism , Pentose Phosphate Pathway/physiology , Transcription, Genetic/genetics , Allosteric Regulation , Animals , Cockayne Syndrome/metabolism , DNA-Binding Proteins/genetics , Endonucleases/genetics , Fibroblasts/metabolism , Genomic Instability , Metabolomics , Mice , Mice, Knockout , Nuclear Proteins/genetics , Oxidation-Reduction , Skin/cytology , Transcription Factors/genetics
5.
Nat Commun ; 8: 13980, 2017 02 27.
Article in English | MEDLINE | ID: mdl-28239143

ABSTRACT

The DNA damage response (DDR) is a set of cellular events that follows the generation of DNA damage. Recently, site-specific small non-coding RNAs, also termed DNA damage response RNAs (DDRNAs), have been shown to play a role in DDR signalling and DNA repair. Dysfunctional telomeres activate DDR in ageing, cancer and an increasing number of identified pathological conditions. Here we show that, in mammals, telomere dysfunction induces the transcription of telomeric DDRNAs (tDDRNAs) and their longer precursors from both DNA strands. DDR activation and maintenance at telomeres depend on the biogenesis and functions of tDDRNAs. Their functional inhibition by sequence-specific antisense oligonucleotides allows the unprecedented telomere-specific DDR inactivation in cultured cells and in vivo in mouse tissues. In summary, these results demonstrate that tDDRNAs are induced at dysfunctional telomeres and are necessary for DDR activation and they validate the viability of locus-specific DDR inhibition by targeting DDRNAs.


Subject(s)
DNA Damage , RNA/metabolism , Telomere/metabolism , Animals , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotides, Antisense/pharmacology , RNA/genetics , RNA Processing, Post-Transcriptional/drug effects , RNA Processing, Post-Transcriptional/genetics , Ribonuclease III/metabolism , Transcription, Genetic/drug effects
6.
Cell Rep ; 15(9): 1866-75, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27210754

ABSTRACT

The underlying relation between Parkinson's disease (PD) etiopathology and its major risk factor, aging, is largely unknown. In light of the causative link between genome stability and aging, we investigate a possible nexus between DNA damage accumulation, aging, and PD by assessing aging-related DNA repair pathways in laboratory animal models and humans. We demonstrate that dermal fibroblasts from PD patients display flawed nucleotide excision repair (NER) capacity and that Ercc1 mutant mice with mildly compromised NER exhibit typical PD-like pathological alterations, including decreased striatal dopaminergic innervation, increased phospho-synuclein levels, and defects in mitochondrial respiration. Ercc1 mouse mutants are also more sensitive to the prototypical PD toxin MPTP, and their transcriptomic landscape shares important similarities with that of PD patients. Our results demonstrate that specific defects in DNA repair impact the dopaminergic system and are associated with human PD pathology and might therefore constitute an age-related risk factor for PD.


Subject(s)
Aging/pathology , DNA Repair , Parkinson Disease/pathology , Animals , Corpus Striatum/pathology , Corpus Striatum/ultrastructure , DNA-Binding Proteins/metabolism , Dopaminergic Neurons/pathology , Dopaminergic Neurons/ultrastructure , Endonucleases/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Mice
7.
J Proteome Res ; 14(11): 4674-86, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26420666

ABSTRACT

Recent epidemiological data indicate that radiation doses as low as those used in computer tomography may result in long-term neurocognitive side effects. The aim of this study was to elucidate long-term molecular alterations related to memory formation in the brain after low and moderate doses of γ radiation. Female C57BL/6J mice were irradiated on postnatal day 10 with total body doses of 0.1, 0.5, or 2.0 Gy; the control group was sham-irradiated. The proteome analysis of hippocampus, cortex, and synaptosomes isolated from these brain regions indicated changes in ephrin-related, RhoGDI, and axonal guidance signaling. Immunoblotting and miRNA-quantification demonstrated an imbalance in the synapse morphology-related Rac1-Cofilin pathway and long-term potentiation-related cAMP response element-binding protein (CREB) signaling. Proteome profiling also showed impaired oxidative phosphorylation, especially in the synaptic mitochondria. This was accompanied by an early (4 weeks) reduction of mitochondrial respiration capacity in the hippocampus. Although the respiratory capacity was restored by 24 weeks, the number of deregulated mitochondrial complex proteins was increased at this time. All observed changes were significant at doses of 0.5 and 2.0 Gy but not at 0.1 Gy. This study strongly suggests that ionizing radiation at the neonatal state triggers persistent proteomic alterations associated with synaptic impairment.


Subject(s)
Cerebral Cortex/radiation effects , Gamma Rays/adverse effects , Hippocampus/radiation effects , Long-Term Potentiation/radiation effects , Proteome/genetics , Synaptic Transmission/radiation effects , Actin Depolymerizing Factors/genetics , Actin Depolymerizing Factors/metabolism , Animals , Animals, Newborn , Axons/metabolism , Axons/radiation effects , Axons/ultrastructure , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Electron Transport Chain Complex Proteins/genetics , Electron Transport Chain Complex Proteins/metabolism , Ephrins/genetics , Ephrins/metabolism , Female , Hippocampus/metabolism , Hippocampus/physiopathology , Memory/drug effects , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/metabolism , Mitochondria/radiation effects , Neuropeptides/genetics , Neuropeptides/metabolism , Oxidative Phosphorylation/radiation effects , Proteome/metabolism , Synaptosomes/metabolism , Synaptosomes/radiation effects , Whole-Body Irradiation , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rho-Specific Guanine Nucleotide Dissociation Inhibitors/genetics , rho-Specific Guanine Nucleotide Dissociation Inhibitors/metabolism
8.
Oxid Med Cell Longev ; 2015: 967203, 2015.
Article in English | MEDLINE | ID: mdl-25973140

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia, characterized by progressive neurodegeneration. Pathogenetic mechanisms, triggered by ß-amyloid (Aß) accumulation, include oxidative stress, derived from energy homeostasis deregulation and involving mitochondria and peroxisomes. We here addressed the oxidative stress status and the elicited cellular response at the onset and during the progression of Aß pathology, studying the neocortex of Tg2576 model of AD. Age-dependent changes of oxidative damage markers, antioxidant enzymes, and related transcription factors were analysed in relation to the distribution of Aß peptide and oligomers, by a combined molecular/morphological approach. Nucleic acid oxidative damage, accompanied by defective antioxidant defences, and decreased PGC1α expression are already detected in 3-month-old Tg2576 neurons. Conversely, PPARα is increased in these cells, with its cytoplasmic localization suggesting nongenomic, anti-inflammatory actions. At 6 months, when intracellular Aß accumulates, PMP70 is downregulated, indicating impairment of fatty acids peroxisomal translocation and their consequent harmful accumulation. In 9-month-old Tg2576 neocortex, Aß oligomers and acrolein deposition correlate with GFAP, GPX1, and PMP70 increases, supporting a compensatory response, involving astroglial peroxisomes. At severe pathological stages, when senile plaques disrupt cortical cytoarchitecture, antioxidant capacity is gradually lost. Overall, our data suggest early therapeutic intervention in AD, also targeting peroxisomes.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Neocortex/metabolism , Oxidative Stress , ATP-Binding Cassette Transporters/metabolism , Acrolein/metabolism , Aging , Alzheimer Disease/metabolism , Animals , Disease Models, Animal , Down-Regulation , Female , Genotype , Glial Fibrillary Acidic Protein , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Neocortex/pathology , Neocortex/ultrastructure , Nerve Tissue Proteins/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Peroxisomes/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Transcription Factors/genetics , Transcription Factors/metabolism , Glutathione Peroxidase GPX1
9.
J Proteome Res ; 14(5): 2055-64, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25807253

ABSTRACT

The increased use of radiation-based medical imaging methods such as computer tomography is a matter of concern due to potential radiation-induced adverse effects. Efficient protection against such detrimental effects has not been possible due to inadequate understanding of radiation-induced alterations in signaling pathways. The aim of this study was to elucidate the molecular mechanisms behind learning and memory deficits after acute low and moderate doses of ionizing radiation. Female C57BL/6J mice were irradiated on postnatal day 10 (PND10) with gamma doses of 0.1 or 0.5 Gy. This was followed by evaluation of the cellular proteome, pathway-focused transcriptome, and neurological development/disease-focused miRNAome of hippocampus and cortex 24 h postirradiation. Our analysis showed that signaling pathways related to mitochondrial and synaptic functions were changed by acute irradiation. This may lead to reduced mitochondrial function paralleled by enhanced number of dendritic spines and neurite outgrowth due to elevated long-term potentiation, triggered by increased phosphorylated CREB. This was predominately observed in the cortex at 0.1 and 0.5 Gy and in the hippocampus only at 0.5 Gy. Moreover, a radiation-induced increase in the expression of several neural miRNAs associated with synaptic plasticity was found. The early changes in signaling pathways related to memory formation may be associated with the acute neurocognitive side effects in patients after brain radiotherapy but might also contribute to late radiation-induced cognitive injury.


Subject(s)
Cerebral Cortex/radiation effects , Hippocampus/radiation effects , Long-Term Potentiation/radiation effects , Memory/radiation effects , Mitochondria/radiation effects , Synapses/radiation effects , Animals , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Cerebral Cortex/physiology , Dose-Response Relationship, Radiation , Female , Gamma Rays , Gene Expression , Hippocampus/physiology , Humans , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Phosphorylation , Proteome/metabolism , Signal Transduction/radiation effects , Synapses/physiology , Transcriptome
10.
Biochim Biophys Acta ; 1842(9): 1385-94, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24854107

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a complex disease and the current interest and focus of scientific research is both investigating the variety of causes that underlie PD pathogenesis, and identifying reliable biomarkers to diagnose and monitor the progression of pathology. Investigation on pathogenic mechanisms in peripheral cells, such as fibroblasts derived from patients with sporadic PD and age/gender matched controls, might generate deeper understanding of the deficits affecting dopaminergic neurons and, possibly, new tools applicable to clinical practice. METHODS: Primary fibroblast cultures were established from skin biopsies. Increased susceptibility to the PD-related toxin rotenone was determined with apoptosis- and necrosis-specific cell death assays. Protein quality control was evaluated assessing the efficiency of the Ubiquitin Proteasome System (UPS) and protein levels of autophagic markers. Changes in cellular bioenergetics were monitored by measuring oxygen consumption and glycolysis-dependent medium acidification. The oxido-reductive status was determined by detecting mitochondrial superoxide production and oxidation levels in proteins and lipids. RESULTS: PD fibroblasts showed higher vulnerability to necrotic cell death induced by complex I inhibitor rotenone, reduced UPS function and decreased maximal and rotenone-sensitive mitochondrial respiration. No changes in autophagy and redox markers were detected. CONCLUSIONS: Our study shows that increased susceptibility to rotenone and the presence of proteolytic and bioenergetic deficits that typically sustain the neurodegenerative process of PD can be detected in fibroblasts from idiopathic PD patients. Fibroblasts might therefore represent a powerful and minimally invasive tool to investigate PD pathogenic mechanisms, which might translate into considerable advances in clinical management of the disease.


Subject(s)
Energy Metabolism , Fibroblasts/pathology , Mitochondria/metabolism , Parkinson Disease/pathology , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Adenosine Triphosphate/metabolism , Apoptosis , Autophagy , Case-Control Studies , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Homeostasis , Humans , Male , Middle Aged , Oxygen Consumption/drug effects , Parkinson Disease/metabolism , Rotenone/pharmacology , Superoxides/metabolism , Uncoupling Agents/pharmacology
11.
Free Radic Biol Med ; 75 Suppl 1: S10, 2014 Oct.
Article in English | MEDLINE | ID: mdl-26461279

ABSTRACT

Parkinson׳s disease (PD) is a complex disease and the current interest and focus of scientific research is both investigating the variety of causes that underlie PD pathogenesis, and identifying reliable biomarkers to diagnose and monitor the progression of pathology. Investigation on pathogenic mechanisms in peripheral cells, such as fibroblasts derived from patients with sporadic PD and age/gender matched controls, might generate deeper understanding of the deficits affecting dopaminergic neurons and, possibly, new tools applicable to clinical practice. The chronic and slow progressing nature of PD may result from subtle yet persistent alterations in biological mechanisms, which might be undetectable in basal, unchallenged conditions. Unlike body fluids, dermal fibroblasts can be exposed to different challenges while in culture and can therefore generate information about the dynamic cellular responses to exogenous stressors. These studies may ultimately generate indicators highlighting the biological defects intrinsic to PD. In fact, fibroblasts from idiopathic PD patients' exhibit deficits typically sustaining the neurodegenerative process of PD, such as increased susceptibility to rotenone as well as deficits in protein homeostasis and mitochondrial bioenergetics Fibroblasts therefore represent a powerful and minimally invasive tool to investigate PD pathogenic mechanisms, which might translate into considerable advances in clinical management of the disease.

12.
Neurobiol Aging ; 35(1): 96-108, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23910655

ABSTRACT

Autophagy is a major protein degradation pathway, essential for stress-induced and constitutive protein turnover. In nervous tissue, autophagy is constitutively active and crucial to neuronal survival. The efficiency of the autophagic pathway reportedly undergoes age-related decline, and autophagy defects are observed in neurodegenerative diseases. Since Ambra1 plays a fundamental role in regulating the autophagic process in developing nervous tissue, we investigated the expression of this protein in mature mouse brain and during physiological and Alzheimer type aging. The present study accomplished the first complete map of Ambra1 protein distribution in the various brain areas, and highlights differential expression in neuronal/glial cell populations. Differences in Ambra1 content are possibly related to specific neuronal features and properties, particularly concerning susceptibility to neurodegeneration. Furthermore, the analysis of Ambra1 expression in physiological and pathological brain aging supports important, though conflicting, functions of autophagy in neurodegenerative processes. Thus, novel therapeutic approaches, based on autophagy modulation, should also take into account the age-dependent roles of this mechanism in establishing, promoting, or counteracting neurodegeneration.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Aging/genetics , Alzheimer Disease/genetics , Autophagy/genetics , Brain/metabolism , Brain/physiology , Gene Expression Regulation, Developmental/genetics , Aging/pathology , Aging/physiology , Alzheimer Disease/pathology , Animals , Autophagy/physiology , Genetic Predisposition to Disease/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology
13.
DNA Repair (Amst) ; 12(8): 568-77, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23726220

ABSTRACT

Impaired DNA repair involving the nucleotide excision repair (NER)/transcription-coupled repair (TCR) pathway cause human pathologies associated with severe neurological symptoms. These clinical observations suggest that defective NER/TCR might also play a critical role in chronic neurodegenerative disorders (ND), such as Alzheimer's and Parkinson's disease. Involvement of NER/TCR in these disorders is also substantiated by the evidence that aging constitutes the principal risk factor for chronic ND and that this DNA repair mechanism is very relevant for the aging process itself. Our understanding of the exact role of NER/TCR in chronic ND, however, is extremely rudimentary; while there is no doubt that defective NER/TCR can lead to neuronal death, evidence for its participation in the etiopathogenesis of ND is inconclusive thus far. Here we summarize the experimental observations supporting a role for NER/TCR in chronic ND and suggest questions and lines of investigation that might help in addressing this important issue. We also present a preliminary yet unprecedented meta-analysis on human brain microarray data to understand the expression levels of the various NER factors in the anatomical areas relevant for chronic ND pathogenesis. In summary, this review intends to highlight elements supporting a role of NER/TCR in these devastating disorders and to propose potential strategies of investigation.


Subject(s)
Aging/genetics , DNA Repair , Neurodegenerative Diseases/genetics , Animals , Brain/metabolism , Brain/pathology , Chronic Disease , DNA Damage , Disease Models, Animal , Humans , Neurodegenerative Diseases/pathology , Neurons/cytology , Neurons/pathology
14.
Mol Neurodegener ; 8: 8, 2013 Feb 02.
Article in English | MEDLINE | ID: mdl-23374228

ABSTRACT

BACKGROUND: Alzheimer's Disease (AD) is a progressive neurodegenerative disease, especially affecting the hippocampus. Impairment of cognitive and memory functions is associated with amyloid ß-peptide-induced oxidative stress and alterations in lipid metabolism. In this scenario, the dual role of peroxisomes in producing and removing ROS, and their function in fatty acids ß-oxidation, may be critical. This work aims to investigating the possible involvement of peroxisomes in AD onset and progression, as studied in a transgenic mouse model, harboring the human Swedish familial AD mutation. We therefore characterized the peroxisomal population in the hippocampus, focusing on early, advanced, and late stages of the disease (3, 6, 9, 12, 18 months of age). Several peroxisome-related markers in transgenic and wild-type hippocampal formation were comparatively studied, by a combined molecular/immunohistochemical/ultrastructural approach. RESULTS: Our results demonstrate early and significant peroxisomal modifications in AD mice, compared to wild-type. Indeed, the peroxisomal membrane protein of 70 kDa and acyl-CoA oxidase 1 are induced at 3 months, possibly reflecting the need for efficient fatty acid ß-oxidation, as a compensatory response to mitochondrial dysfunction. The concomitant presence of oxidative damage markers and the altered expression of antioxidant enzymes argue for early oxidative stress in AD. During physiological and pathological brain aging, important changes in the expression of peroxisome-related proteins, also correlating with ongoing gliosis, occur in the hippocampus. These age- and genotype-based alterations, strongly dependent on the specific marker considered, indicate metabolic and/or numerical remodeling of peroxisomal population. CONCLUSIONS: Overall, our data support functional and biogenetic relationships linking peroxisomes to mitochondria and suggest peroxisomal proteins as biomarkers/therapeutic targets in pre-symptomatic AD.


Subject(s)
Aging/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Hippocampus/metabolism , Peroxisomes/metabolism , Amyloid beta-Peptides/genetics , Animals , Energy Metabolism/physiology , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, Transgenic , Microscopy, Immunoelectron , Oxidative Stress/physiology
15.
Antioxid Redox Signal ; 15(4): 855-71, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21395478

ABSTRACT

AIMS: The study of the intracellular oxido-reductive (redox) state is of extreme relevance to the dopamine (DA) neurons of the substantia nigra pars compacta. These cells possess a distinct physiology intrinsically associated with elevated reactive oxygen species production, and they selectively degenerate in Parkinson's disease under oxidative stress conditions. To test the hypothesis that these cells display a unique redox response to mild, physiologically relevant oxidative insults when compared with other neuronal populations, we sought to develop a novel method for quantitatively assessing mild variations in intracellular redox state. RESULTS: We have developed a new imaging strategy to study redox variations in single cells, which is sensitive enough to detect changes within the physiological range. We studied DA neurons' physiological redox response in biological systems of increasing complexity--from primary cultures to zebrafish larvae, to mammalian brains-and identified a redox response that is distinctive for substantia nigra pars compacta DA neurons. We studied simultaneously, and in the same cells, redox state and signaling activation and found that these phenomena are synchronized. INNOVATION: The redox histochemistry method we have developed allows for sensitive quantification of intracellular redox state in situ. As this method is compatible with traditional immunohistochemical techniques, it can be applied to diverse settings to investigate, in theory, any cell type of interest. CONCLUSION: Although the technique we have developed is of general interest, these findings provide insights into the biology of DA neurons in health and disease and may have implications for therapeutic intervention.


Subject(s)
Brain Mapping/methods , Dopamine/physiology , Immunohistochemistry/methods , Neurons/physiology , Single-Cell Analysis , Substantia Nigra/physiology , Animals , Astrocytes/cytology , Cells, Cultured , Neurons/cytology , Oxidation-Reduction , Parkinson Disease/pathology , Rats , Reactive Oxygen Species/metabolism , Zebrafish/physiology
16.
Mod Pathol ; 24(6): 855-65, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21441900

ABSTRACT

The demonstration that type 2 transglutaminase (TG2) can incorporate polyamine into the E7 oncoprotein of human papillomavirus (HPV) type 18 has led to the hypothesis that TG2 can have a role in the host cellular response to HPV infection. The aim of this study was to investigate whether HPV-related pathology, in infected human cervical epithelium, was associated with modulation of TG2 expression. Normal controls and HPV-infected cervical biopsies were analyzed for the expression of TG2, and the findings were compared with lesion grade. The correlation between TG2 expression and p16, a marker for HPV-induced dysplasia, and the retinoblastoma protein (Rb), a target of the E7 protein of HPV, was also investigated. Results obtained showed that TG2 was absent in normal squamous mucosa, whereas it was present in 100% CIN I lesions. Low-grade lesions showed significantly higher TG2 expression than high-grade lesions (P<0.0001). In 94% of CIN I more than 50% of the cells were positive for TG2, with a strong staining intensity (+3), whereas a decreased staining intensity and a low number of positive cells were found in CIN II/III. In CIN I cases, both nuclear and cytoplasmic staining were found in cells exhibiting classical morphological features of HPV infection. In addition, during progression from low-grade squamous intraepithelial lesions to severe dysplasia, TG2 expression was inversely correlated with p16 (Pearson: -0.930), whereas a positive correlation was observed between the expression of TG2 and pRb (Pearson: 0.997). TG2 is expressed in HPV infection as an early phenomenon, not restricted to high-risk genotypes. TG2 upregulation is probably part of host cell reaction against HPV-induced tissue modification. It may act as a cellular antioxidant defense factor, playing an important role in counteracting oxidative damage in neoplastic disease.


Subject(s)
GTP-Binding Proteins/metabolism , Precancerous Conditions/diagnosis , Transglutaminases/metabolism , Uterine Cervical Dysplasia/diagnosis , Uterine Cervical Neoplasms/diagnosis , Adult , Biomarkers, Tumor/metabolism , Cervix Uteri/enzymology , Cervix Uteri/pathology , Cervix Uteri/virology , Cyclin-Dependent Kinase Inhibitor p16 , Female , Humans , Middle Aged , Neoplasm Proteins/metabolism , Papillomavirus Infections/complications , Papillomavirus Infections/enzymology , Papillomavirus Infections/pathology , Precancerous Conditions/enzymology , Precancerous Conditions/virology , Predictive Value of Tests , Protein Glutamine gamma Glutamyltransferase 2 , Retinoblastoma Protein/metabolism , Retrospective Studies , Uterine Cervical Neoplasms/enzymology , Young Adult , Uterine Cervical Dysplasia/enzymology , Uterine Cervical Dysplasia/virology
17.
Hum Mol Genet ; 19(22): 4529-42, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20829229

ABSTRACT

Vulnerability of motoneurons in amyotrophic lateral sclerosis (ALS) arises from a combination of several mechanisms, including protein misfolding and aggregation, mitochondrial dysfunction and oxidative damage. Protein aggregates are found in motoneurons in models for ALS linked to a mutation in the gene coding for Cu,Zn superoxide dismutase (SOD1) and in ALS patients as well. Aggregation of mutant SOD1 in the cytoplasm and/or into mitochondria has been repeatedly proposed as a main culprit for the degeneration of motoneurons. It is, however, still debated whether SOD1 aggregates represent a cause, a correlate or a consequence of processes leading to cell death. We have exploited the ability of glutaredoxins (Grxs) to reduce mixed disulfides to protein thiols either in the cytoplasm and in the IMS (Grx1) or in the mitochondrial matrix (Grx2) as a tool for restoring a correct redox environment and preventing the aggregation of mutant SOD1. Here we show that the overexpression of Grx1 increases the solubility of mutant SOD1 in the cytosol but does not inhibit mitochondrial damage and apoptosis induced by mutant SOD1 in neuronal cells (SH-SY5Y) or in immortalized motoneurons (NSC-34). Conversely, the overexpression of Grx2 increases the solubility of mutant SOD1 in mitochondria, interferes with mitochondrial fragmentation by modifying the expression pattern of proteins involved in mitochondrial dynamics, preserves mitochondrial function and strongly protects neuronal cells from apoptosis. The toxicity of mutant SOD1, therefore, mostly arises from mitochondrial dysfunction and rescue of mitochondrial damage may represent a promising therapeutic strategy.


Subject(s)
Glutaredoxins/metabolism , Mitochondria/metabolism , Superoxide Dismutase/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Apoptosis/genetics , Cell Death/genetics , Cell Line, Transformed , Cell Line, Tumor , Humans , Mice , Mitochondria/genetics , Mitochondria/ultrastructure , Motor Neurons/cytology , Motor Neurons/metabolism , Mutation , Neuroblastoma/pathology , Neurons/metabolism , Oxidation-Reduction , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Superoxide Dismutase-1
18.
Neurobiol Dis ; 40(3): 634-44, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20708685

ABSTRACT

The endoplasmic reticulum (ER) stress-mediated pathway is involved in a wide range of human neurodegenerative disorders. Hence, molecules that regulate the ER stress response represent potential candidates as drug targets to tackle these diseases. In previous studies we demonstrated that upon acetylation the reticulon-1C (RTN-1C) variant of the reticulon family leads to inhibition of histone deacetylase (HDAC) enzymatic activity and endoplasmic reticulum stress-dependent apoptosis. Here, by microarray analysis of the whole human genome we found that RTN-1C is able to specifically regulate gene expression, modulating transcript clusters which have been implicated in the onset of neurodegenerative disorders. Interestingly, we show that some of the identified genes were also modulated in vivo in a brain-specific mouse model overexpressing RTN-1C. These data provide a basis for further investigation of RTN-1C as a potential molecular target for use in therapy and as a specific marker for neurological diseases.


Subject(s)
Brain/metabolism , Gene Expression Regulation , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/genetics , Neurons/metabolism , Animals , Apoptosis/physiology , Blotting, Western , Brain/pathology , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Fluorescent Antibody Technique , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Nerve Tissue Proteins/metabolism , Neuroblastoma , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Oligonucleotide Array Sequence Analysis , Patch-Clamp Techniques , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...