Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Psychiatry ; 28(11): 4902-4914, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37779111

RESUMO

In the field of neurodegenerative diseases, especially sporadic Parkinson's disease (sPD) with dementia (sPDD), the question of how the disease starts and spreads in the brain remains central. While prion-like proteins have been designated as a culprit, recent studies suggest the involvement of additional factors. We found that oxidative stress, damaged DNA binding, cytosolic DNA sensing, and Toll-Like Receptor (TLR)4/9 activation pathways are strongly associated with the sPDD transcriptome, which has dysregulated type I Interferon (IFN) signaling. In sPD patients, we confirmed deletions of mitochondrial (mt)DNA in the medial frontal gyrus, suggesting a potential role of damaged mtDNA in the disease pathophysiology. To explore its contribution to pathology, we used spontaneous models of sPDD caused by deletion of type I IFN signaling (Ifnb-/-/Ifnar-/- mice). We found that the lack of neuronal IFNß/IFNAR leads to oxidization, mutation, and deletion in mtDNA, which is subsequently released outside the neurons. Injecting damaged mtDNA into mouse brain induced PDD-like behavioral symptoms, including neuropsychiatric, motor, and cognitive impairments. Furthermore, it caused neurodegeneration in brain regions distant from the injection site, suggesting that damaged mtDNA triggers spread of PDD characteristics in an "infectious-like" manner. We also discovered that the mechanism through which damaged mtDNA causes pathology in healthy neurons is independent of Cyclic GMP-AMP synthase and IFNß/IFNAR, but rather involves the dual activation of TLR9/4 pathways, resulting in increased oxidative stress and neuronal cell death, respectively. Our proteomic analysis of extracellular vesicles containing damaged mtDNA identified the TLR4 activator, Ribosomal Protein S3 as a key protein involved in recognizing and extruding damaged mtDNA. These findings might shed light on new molecular pathways through which damaged mtDNA initiates and spreads PD-like disease, potentially opening new avenues for therapeutic interventions or disease monitoring.


Assuntos
DNA Mitocondrial , Doença de Parkinson , Humanos , Camundongos , Animais , DNA Mitocondrial/genética , Doença de Parkinson/genética , Doença de Parkinson/patologia , Proteômica , Mitocôndrias/metabolismo , Neurônios/metabolismo
2.
Front Oncol ; 12: 1028959, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36531051

RESUMO

The PDZ-LIM domain-containing protein 2 (PDLIM2) regulates cell polarity and the protein stability of key transcription factors in epithelial and hemopoietic cells. We previously reported that PDLIM2 is more highly expressed in Triple Negative Breast Cancer (TNBC) than in other breast cancer types or normal breast tissue. In the course of the TNBC study, it was noted that PDLIM2 was highly expressed in the stroma of PDLIM2-expressing tumours. Here, we investigated the phenotype of these stromal cells and whether any infiltrating immune population was linked to PDLIM2 expression. We found that high PDLIM2 expression in breast tumours was associated with higher levels of infiltrating M2 macrophages, but was not associated with infiltrating T cell sub-populations. We then tested whether PDLIM2 contributes to macrophage differentiation or function by using cultures of bone marrow-derived macrophages from wildtype and Pdlim2 knockout mice. This demonstrated that PDLIM2 is required for naïve macrophage migration and for the full adoption of IL-4-induced M2 polarization, including expression of M2 phenotypic markers, cell adhesion and cell migration. TLR4-, TLR3- or IFNγ-induced M1 macrophage activity was less dependent on PDLIM2. Finally, analysis of publicly available breast cancer datasets showed that high PDLIM2 expression is associated with increased M2 macrophage infiltration. We conclude that PDLIM2 expression influences the tumour associated stroma and, in particular, M2 macrophage infiltration that may contribute to the progression of TNBC or other subsets of breast cancer.

3.
Ann Neurol ; 90(5): 789-807, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34476836

RESUMO

OBJECTIVE: Parkinson's disease (PD) manifests in motor dysfunction, non-motor symptoms, and eventual dementia (PDD). Neuropathological hallmarks include nigrostriatal neurodegeneration, Lewy body (LB) pathology, and neuroinflammation. Alpha-synuclein (α-syn), a primary component of LBs, is implicated in PD pathogenesis, accumulating, and aggregating in both familial and sporadic PD. However, as α-syn pathology is often comorbid with amyloid-beta (Aß) plaques and phosphorylated tau (pTau) tangles in PDD, it is still unclear whether α-syn is the primary cause of neurodegeneration in sporadic PDD. We aimed to determine how the absence of α-syn would affect PDD manifestation. METHODS: IFN-ß knockout (Ifnb-/- ) mice spontaneously develop progressive behavior abnormalities and neuropathology resembling PDD, notably with α-syn+ LBs. We generated Ifnb/Snca double knockout (DKO) mice and evaluated their behavior and neuropathology compared with wild-type (Wt), Ifnb-/- , and Snca-/- mice using immunohistochemistry, electron microscopy, immunoblots, qPCR, and modification of neuronal signaling. RESULTS: Ifnb/Snca DKO mice developed all clinical PDD-like behavioral manifestations induced by IFN-ß loss. Independently of α-syn expression, lack of IFN-ß alone induced Aß plaques, pTau tangles, and LB-like Aß+ /pTau+ inclusion bodies and neuroinflammation. IFN-ß loss caused significant elevated glial and neuronal TNF-α and neuronal TNFR1, associated with neurodegeneration. Restoring neuronal IFN-ß signaling or blocking TNFR1 rescued caspase 3/t-BID-mediated neuronal-death through upregulation of c-FLIPS and lowered intraneuronal Aß and pTau accumulation. INTERPRETATION: These findings increase our understanding of PD pathology and suggest that targeting α-syn alone is not sufficient to mitigate disease. Targeting specific aspects of neuroinflammation, such as aberrant neuronal TNF-α/TNFR1 or IFN-ß/IFNAR signaling, may attenuate disease. ANN NEUROL 2021;90:789-807.


Assuntos
Doenças Neuroinflamatórias/metabolismo , Neurônios/metabolismo , Doença de Parkinson/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Progressão da Doença , Corpos de Lewy/patologia , Doença por Corpos de Lewy/metabolismo , Camundongos Knockout , Neuroglia/patologia , Doença de Parkinson/genética , Placa Amiloide/metabolismo , Fator de Necrose Tumoral alfa/deficiência
4.
Mol Psychiatry ; 26(10): 6083-6099, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34234281

RESUMO

Familial Parkinson disease (PD) is associated with rare genetic mutations, but the etiology in most patients with sporadic (s)PD is largely unknown, and the basis for its progression to dementia (sPDD) is poorly characterized. We have identified that loss of IFNß or IFNAR1, the receptor for IFNα/ß, causes pathological and behavioral changes resembling PDD, prompting us to hypothesize that dysregulated genes in IFNß-IFNAR signaling pathway predispose one to sPD. By transcriptomic analysis, we found defective neuronal IFNß-IFNAR signaling, including particularly elevated PIAS2 associated with sPDD. With meta-analysis of GWASs, we identified sequence variants in IFNß-IFNAR-related genes in sPD patients. Furthermore, sPDD patients expressed higher levels of PIAS2 mRNA and protein in neurons. To determine its function in brain, we overexpressed PIAS2 under a neuronal promoter, alone or with human α-synuclein, in the brains of mice, which caused motor and cognitive impairments and correlated with intraneuronal phosphorylated (p)α-synuclein accumulation and dopaminergic neuron loss. Ectopic expression of neuronal PIAS2 blocked mitophagy, increased the accumulation of senescent mitochondrial and oxidative stress, as evidenced by excessive oxDJ1 and 8OHdG, by inactivating ERK1/2-P53 signaling. Conversely, PIAS2 knockdown rescued the clinicopathological manifestations of PDD in Ifnb-/- mice on restoring mitochondrial homeostasis, oxidative stress, and pERK1/2-pP53 signaling. The regulation of JAK-STAT2-PIAS2 signaling was crucial for neurite outgrowth and neuronal survival and excitability and thus might prevent cognitive impairments. Our findings provide insights into the progression of sPD and dementia and have implications for new therapeutic approaches.


Assuntos
Demência , Interferon beta/metabolismo , Doença de Parkinson , Proteínas Inibidoras de STAT Ativados , Transdução de Sinais , Animais , Demência/genética , Neurônios Dopaminérgicos/metabolismo , Humanos , Camundongos , Camundongos Knockout , Degeneração Neural , Doença de Parkinson/genética , Proteínas Inibidoras de STAT Ativados/genética , alfa-Sinucleína/metabolismo
5.
EMBO J ; 40(11): e106868, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33913175

RESUMO

Mitochondrial homeostasis is essential for providing cellular energy, particularly in resource-demanding neurons, defects in which cause neurodegeneration, but the function of interferons (IFNs) in regulating neuronal mitochondrial homeostasis is unknown. We found that neuronal IFN-ß is indispensable for mitochondrial homeostasis and metabolism, sustaining ATP levels and preventing excessive ROS by controlling mitochondrial fission. IFN-ß induces events that are required for mitochondrial fission, phosphorylating STAT5 and upregulating PGAM5, which phosphorylates serine 622 of Drp1. IFN-ß signaling then recruits Drp1 to mitochondria, oligomerizes it, and engages INF2 to stabilize mitochondria-endoplasmic reticulum (ER) platforms. This process tethers damaged mitochondria to the ER to separate them via fission. Lack of neuronal IFN-ß in the Ifnb-/- model of Parkinson disease (PD) disrupts STAT5-PGAM5-Drp1 signaling, impairing fission and causing large multibranched, damaged mitochondria with insufficient ATP production and excessive oxidative stress to accumulate. In other PD models, IFN-ß rescues dopaminergic neuronal cell death and pathology, associated with preserved mitochondrial homeostasis. Thus, IFN-ß activates mitochondrial fission in neurons through the pSTAT5/PGAM5/S622 Drp1 pathway to stabilize mitochondria/ER platforms, constituting an essential neuroprotective mechanism.


Assuntos
Interferon beta/metabolismo , Dinâmica Mitocondrial , Doença de Parkinson/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Dinaminas/metabolismo , Forminas/metabolismo , Interferon beta/genética , Camundongos , Mitocôndrias/metabolismo , Neurônios/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Fator de Transcrição STAT5/metabolismo
6.
EBioMedicine ; 57: 102868, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32629384

RESUMO

BACKGROUND: Point mutations and structural variations (SVs) in mitochondrial DNA (mtDNA) contribute to many neurodegenerative diseases. Technical limitations and heteroplasmy, however, have impeded their identification, preventing these changes from being examined in neurons in healthy and disease states. METHODS: We have developed a high-resolution technique-Mitochondrial DNA Structural Variation Sequencing (MitoSV-seq)-that identifies all types of mtDNA SVs and single-nucleotide variations (SNVs) in single neurons and novel variations that have been undetectable with conventional techniques. FINDINGS: Using MitoSV-seq, we discovered SVs/SNVs in dopaminergic neurons in the Ifnar1-/- murine model of Parkinson disease. Further, MitoSV-seq was found to have broad applicability, delivering high-quality, full-length mtDNA sequences in a species-independent manner from human PBMCs, haematological cancers, and tumour cell lines, regardless of heteroplasmy. We characterised several common SVs in haematological cancers (AML and MDS) that were linked to the same mtDNA region, MT-ND5, using only 10 cells, indicating the power of MitoSV-seq in determining single-cancer-cell ontologies. Notably, the MT-ND5 hotspot, shared between all examined cancers and Ifnar1-/- dopaminergic neurons, suggests that its mutations have clinical value as disease biomarkers. INTERPRETATION: MitoSV-seq identifies disease-relevant mtDNA mutations in single cells with high resolution, rendering it a potential drug screening platform in neurodegenerative diseases and cancers. FUNDING: The Lundbeck Foundation, Danish Council for Independent Research-Medicine, and European Union Horizon 2020 Research and Innovation Programme.


Assuntos
DNA Mitocondrial/genética , Genoma Mitocondrial/genética , Variação Estrutural do Genoma/genética , Neoplasias/genética , Animais , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Camundongos , Mutação/genética , Neoplasias/patologia , Degeneração Neural/genética , Degeneração Neural/patologia , Polimorfismo de Nucleotídeo Único/genética , Análise de Sequência de DNA , Análise de Célula Única
7.
Artigo em Inglês | MEDLINE | ID: mdl-26191041

RESUMO

IGF-1R expression and activation levels generally cannot be correlated in cancer cells, suggesting that cellular proteins may modulate IGF-1R activity. Strong candidates for such modulation are found in cell-matrix and cell-cell adhesion signaling complexes. Activated IGF-1R is present at focal adhesions, where it can stabilize ß1 integrin and participate in signaling complexes that promote invasiveness associated with epithelial mesenchymal transition (EMT) and resistance to therapy. Whether IGF-1R contributes to EMT or to non-invasive tumor growth may be strongly influenced by the degree of extracellular matrix engagement and the presence or absence of key proteins in IGF-1R-cell adhesion complexes. One such protein is PDLIM2, which promotes both cell polarization and EMT by regulating the stability of transcription factors including NFκB, STATs, and beta catenin. PDLIM2 exhibits tumor suppressor activity, but is also highly expressed in certain invasive cancers. It is likely that distinct adhesion complex proteins modulate IGF-1R signaling during cancer progression or adaptive responses to therapy. Thus, identifying the key modulators will be important for developing effective therapeutic strategies and predictive biomarkers.

8.
Mol Biol Cell ; 25(1): 184-95, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24196835

RESUMO

Epithelial cell differentiation and polarized migration associated with epithelial-to-mesenchymal transition (EMT) in cancer requires integration of gene expression with cytoskeletal dynamics. Here we show that the PDZ-LIM domain protein PDLIM2 (Mystique/SLIM), a known cytoskeletal protein and promoter of nuclear nuclear factor κB (NFκB) and signal transducer and activator of transcription (STAT) degradation, regulates transcription factor activity and gene expression through the COP9 signalosome (CSN). Although repressed in certain cancers, PDLIM2 is highly expressed in invasive cancer cells. Here we show that PDLIM2 suppression causes loss of directional migration, inability to polarize the cytoskeleton, and reversal of the EMT phenotype. This is accompanied by altered activity of several transcription factor families, including ß-catenin, Ap-1, NFκB, interferon regulatory factors, STATs, JUN, and p53. We also show that PDLIM2 associates with CSN5, and cells with suppressed PDLIM2 exhibit reduced nuclear accumulation and deneddylation activity of the CSN toward the cullin 1 and cullin 3 subunits of cullin-RING ubiquitin ligases. Thus PDLIM2 integrates cytoskeleton signaling with gene expression in epithelial differentiation by controlling the stability of key transcription factors and CSN activity.


Assuntos
Transição Epitelial-Mesenquimal , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas com Domínio LIM/fisiologia , Proteínas dos Microfilamentos/fisiologia , Complexo do Signalossomo COP9 , Diferenciação Celular , Movimento Celular , Polaridade Celular , Células Epiteliais/fisiologia , Humanos , Células MCF-7 , Complexos Multiproteicos/metabolismo , NF-kappa B/metabolismo , Peptídeo Hidrolases/metabolismo , Transporte Proteico , beta Catenina
9.
Neuron ; 78(1): 65-80, 2013 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-23498974

RESUMO

Mutations in VCP cause multisystem degeneration impacting the nervous system, muscle, and/or bone. Patients may present with ALS, Parkinsonism, frontotemporal dementia, myopathy, Paget's disease, or a combination of these. The disease mechanism is unknown. We developed a Drosophila model of VCP mutation-dependent degeneration. The phenotype is reminiscent of PINK1 and parkin mutants, including a pronounced mitochondrial defect. Indeed, VCP interacts genetically with the PINK1/parkin pathway in vivo. Paradoxically, VCP complements PINK1 deficiency but not parkin deficiency. The basis of this paradox is resolved by mechanistic studies in vitro showing that VCP recruitment to damaged mitochondria requires Parkin-mediated ubiquitination of mitochondrial targets. VCP recruitment coincides temporally with mitochondrial fission, and VCP is required for proteasome-dependent degradation of Mitofusins in vitro and in vivo. Further, VCP and its adaptor Npl4/Ufd1 are required for clearance of damaged mitochondria via the PINK1/Parkin pathway, and this is impaired by pathogenic mutations in VCP.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Mitocôndrias/genética , Neurônios/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transporte Vesicular , Adenosina Trifosfatases/genética , Animais , Animais Geneticamente Modificados , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Proteínas de Ciclo Celular/genética , Células Cultivadas , Drosophila , Proteínas de Drosophila/genética , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , GTP Fosfo-Hidrolases/metabolismo , Gânglios Espinais/citologia , Regulação da Expressão Gênica/genética , Proteínas de Choque Térmico HSP72/genética , Humanos , Imunoprecipitação , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular , Leupeptinas/farmacologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microscopia Eletrônica de Transmissão , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Mutação/genética , Junção Neuromuscular/genética , Junção Neuromuscular/metabolismo , Neurônios/ultraestrutura , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Fosfatases/genética , Proteínas/metabolismo , Ionóforos de Próton/farmacologia , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/farmacologia , Fatores de Tempo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção , Ubiquitina-Proteína Ligases/genética , Ubiquitinação/genética , Proteína com Valosina
10.
J Neurosci ; 32(12): 4240-6, 2012 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-22442086

RESUMO

The autophagy-lysosomal pathway plays an important role in the clearance of long-lived proteins and dysfunctional organelles. Lysosomal dysfunction has been implicated in several neurodegenerative disorders including Parkinson's disease and related synucleinopathies that are characterized by accumulations of α-synuclein in Lewy bodies. Recent identification of mutations in genes linked to lysosomal function and neurodegeneration has offered a unique opportunity to directly examine the role of lysosomes in disease pathogenesis. Mutations in lysosomal membrane protein ATP13A2 (PARK9) cause familial Kufor-Rakeb syndrome characterized by early-onset parkinsonism, pyramidal degeneration and dementia. While previous data suggested a role of ATP13A2 in α-synuclein misfolding and toxicity, the mechanistic link has not been established. Here we report that loss of ATP13A2 in human fibroblasts from patients with Kufor-Rakeb syndrome or in mouse primary neurons leads to impaired lysosomal degradation capacity. This lysosomal dysfunction results in accumulation of α-synuclein and toxicity in primary cortical neurons. Importantly, silencing of endogenous α-synuclein attenuated the toxicity in ATP13A2-depleted neurons, suggesting that loss of ATP13A2 mediates neurotoxicity at least in part via the accumulation of α-synuclein. Our findings implicate lysosomal dysfunction in the pathogenesis of Kufor-Rakeb syndrome and suggest that upregulation of lysosomal function and downregulation of α-synuclein represent important therapeutic strategies for this disorder.


Assuntos
Regulação da Expressão Gênica/genética , Lisossomos/metabolismo , Neurônios/metabolismo , ATPases Translocadoras de Prótons/deficiência , alfa-Sinucleína/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/citologia , Demência/patologia , Embrião de Mamíferos , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Fibroblastos/metabolismo , Proteínas de Fluorescência Verde/genética , Humanos , L-Lactato Desidrogenase/metabolismo , Leucina/metabolismo , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Lisossomos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação/genética , Proteínas de Neurofilamentos/metabolismo , Neurônios/efeitos dos fármacos , Transtornos Parkinsonianos/patologia , ATPases Translocadoras de Prótons/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Estatísticas não Paramétricas , Fatores de Tempo , Transfecção , Trítio/metabolismo
11.
Autophagy ; 6(2): 217-27, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20104022

RESUMO

VCP (VCP/p97) is a ubiquitously expressed member of the AAA(+)-ATPase family of chaperone-like proteins that regulates numerous cellular processes including chromatin decondensation, homotypic membrane fusion and ubiquitin-dependent protein degradation by the proteasome. Mutations in VCP cause a multisystem degenerative disease consisting of inclusion body myopathy, Paget disease of bone, and frontotemporal dementia (IBMPFD). Here we show that VCP is essential for autophagosome maturation. We generated cells stably expressing dual-tagged LC3 (mCherry-EGFP-LC3) which permit monitoring of autophagosome maturation. We determined that VCP deficiency by RNAi-mediated knockdown or overexpression of dominant-negative VCP results in significant accumulation of immature autophagic vesicles, some of which are abnormally large, acidified and exhibit cathepsin B activity. Furthermore, expression of disease-associated VCP mutants (R155H and A232E) also causes this autophagy defect. VCP was found to be essential to autophagosome maturation under basal conditions and in cells challenged by proteasome inhibition, but not in cells challenged by starvation, suggesting that VCP might be selectively required for autophagic degradation of ubiquitinated substrates. Indeed, a high percentage of the accumulated autophagic vesicles contain ubiquitin-positive contents, a feature that is not observed in autophagic vesicles that accumulate following starvation or treatment with Bafilomycin A. Finally, we show accumulation of numerous, large LAMP-1 and LAMP-2-positive vacuoles and accumulation of LC3-II in myoblasts derived from patients with IBMPFD. We conclude that VCP is essential for maturation of ubiquitin-containing autophagosomes and that defect in this function may contribute to IBMPFD pathogenesis.


Assuntos
Adenosina Trifosfatases , Proteínas de Ciclo Celular , Demência Frontotemporal , Mutação , Miosite de Corpos de Inclusão , Osteíte Deformante , Fagossomos/metabolismo , Ubiquitina/metabolismo , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Catepsina B/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Demência Frontotemporal/genética , Demência Frontotemporal/fisiopatologia , Humanos , Proteína 1 de Membrana Associada ao Lisossomo/genética , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Proteína 2 de Membrana Associada ao Lisossomo/genética , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Miosite de Corpos de Inclusão/genética , Miosite de Corpos de Inclusão/fisiopatologia , Osteíte Deformante/genética , Osteíte Deformante/fisiopatologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Interferência de RNA , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Síndrome , Vacúolos/metabolismo , Proteína com Valosina
12.
Biochim Biophys Acta ; 1793(9): 1422-31, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19133302

RESUMO

Autophagic cell death (ACD) can be operationally described as cell death with an autophagic component. While most molecular bases of this autophagic component are known, in ACD the mechanism of cell death proper is not well defined, in particular because in animal cells there is poor experimental distinction between what triggers autophagy and what triggers ACD. Perhaps as a consequence, it is often thought that in animal cells a little autophagy is protective while a lot is destructive and leads to ACD, thus that the shift from autophagy to ACD is quantitative. The aim of this article is to review current knowledge on ACD in Dictyostelium, a very favorable model, with emphasis on (1) the qualitative, not quantitative nature of the shift from autophagy to ACD, in contrast to the above, and (2) random or targeted mutations of in particular the following genes: iplA (IP3R), TalB (talinB), DcsA (cellulose synthase), GbfA, ugpB, glcS (glycogen synthase) and atg1. These mutations allowed the genetic dissection of ACD features, dissociating in particular vacuolisation from cell death.


Assuntos
Autofagia , Dictyostelium/citologia , Animais , Dictyostelium/genética , Modelos Biológicos , Mutagênese , Proteínas de Protozoários/metabolismo
13.
Exp Cell Res ; 315(1): 26-38, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18951891

RESUMO

Dictyostelium atg1- mutant cells provide an experimentally and genetically favorable model to study necrotic cell death (NCD) with no interference from apoptosis or autophagy. In such cells subjected to starvation and cAMP, induction by the differentiation-inducing factor DIF or by classical uncouplers led within minutes to mitochondrial uncoupling, which causally initiated NCD. We now report that (1) in this model, NCD included a mitochondrial-lysosomal cascade of events, (2) mitochondrial uncoupling and therefore initial stages of death showed reversibility for a surprisingly long time, (3) subsequent lysosomal permeabilization could be demonstrated using Lysosensor blue, acridin orange, Texas red-dextran and cathepsin B substrate, (4) this lysosomal permeabilization was irreversible, and (5) the presence of the uncoupler was required to maintain mitochondrial lesions but also to induce lysosomal lesions, suggesting that signaling from mitochondria to lysosomes must be sustained by the continuous presence of the uncoupler. These results further characterized the NCD pathway in this priviledged model, contributed to a definition of NCD at the lysosomal level, and suggested that in mammalian NCD even late reversibility attempts by removal of the inducer may be of therapeutic interest.


Assuntos
Dictyostelium/citologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Necrose/patologia , Desacopladores/farmacologia , Laranja de Acridina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Catepsina B/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Dextranos/metabolismo , Dictyostelium/efeitos dos fármacos , Fluoresceínas/metabolismo , Fluorescência , Oxazóis/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Permeabilidade/efeitos dos fármacos
14.
Methods Enzymol ; 446: 1-15, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18603113

RESUMO

Non-apoptotic cell death types can be conveniently studied in Dictyostelium discoideum, an exceptionally favorable model not only because of its well-known genetic and experimental advantages, but also because in Dictyostelium there is no apoptosis machinery that could interfere with non-apoptotic cell death. We show here how to conveniently demonstrate, assess, and study these non-apoptotic cell death types. These can be generated by use of modifications of the monolayer technique of Rob Kay et al., and either wild-type HMX44A Dictyostelium cells, leading to autophagic cell death, or the corresponding atg1- autophagy gene mutant cells, leading to necrotic cell death. Methods to follow these non-apoptotic cell death types qualitatively and quantitatively will be reported.


Assuntos
Autofagia/fisiologia , Dictyostelium/citologia , Necrose/fisiopatologia , Animais , Dictyostelium/crescimento & desenvolvimento , Citometria de Fluxo , Microscopia Eletrônica , Microscopia de Contraste de Fase
15.
Autophagy ; 4(5): 680-91, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18424909

RESUMO

Autophagic cell death in Dictyostelium can be dissociated into a starvation-induced sensitization stage and a death induction stage. A UDP-glucose pyrophosphorylase (ugpB) mutant and a glycogen synthase (glcS) mutant shared the same abnormal phenotype. In vitro, upon starvation alone mutant cells showed altered contorted morphology, indicating that the mutations affected the pre-death sensitization stage. Upon induction of cell death, most of these mutant cells underwent death without vacuolization, distinct from either autophagic or necrotic cell death. Autophagy itself was not grossly altered as shown by conventional and electron microscopy. Exogenous glycogen or maltose could complement both ugpB(-) and glcS(-) mutations, leading back to autophagic cell death. The glcS(-) mutation could also be complemented by 2-deoxyglucose that cannot undergo glycolysis. In agreement with the in vitro data, upon development glcS(-) stalk cells died but most were not vacuolated. We conclude that a UDP-glucose derivative (such as glycogen or maltose) plays an essential energy-independent role in autophagic cell death.


Assuntos
Autofagia/fisiologia , Uridina Difosfato Glucose/análogos & derivados , Uridina Difosfato Glucose/fisiologia , Vacúolos/patologia , Animais , Autofagia/genética , Dictyostelium/citologia , Dictyostelium/enzimologia , Dictyostelium/genética , Glicogênio Sintase/genética , Glicogênio Sintase/fisiologia , Mutagênese Insercional , UTP-Glucose-1-Fosfato Uridililtransferase/genética , Vacúolos/enzimologia , Vacúolos/genética
16.
Methods Enzymol ; 451: 343-58, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19185731

RESUMO

Autophagic cell death can be conveniently studied in Dictyostelium discoideum, an exceptionally favorable model not only because of its well-known genetic and experimental advantages but also because in Dictyostelium there is no apoptosis machinery that could interfere with nonapoptotic cell death. Moreover, autophagic cell death in Dictyostelium can be dissociated into a starvation-induced sensitization stage, during which autophagy is induced, and a death induction stage. We show here how to demonstrate, assess and analyze this autophagic cell death. This can be studied in vivo during the development of Dictyostelium, and in vitro, using modifications of the monolayer technique of Rob Kay et al. Methods to follow this autophagic cell death qualitatively and quantitatively are reported.


Assuntos
Autofagia/fisiologia , Bioensaio/métodos , Morte Celular/fisiologia , Dictyostelium/fisiologia , Animais , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Dictyostelium/citologia , Mutagênese , Proteínas de Protozoários/metabolismo
17.
Semin Cancer Biol ; 17(2): 94-100, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17150370

RESUMO

Among unusual models to study cell death mechanisms, the protist Dictyostelium is remarkable because of its strategic phylogenetic position, with early emergence among eukaryotes and unicellular/multicellular transition, and its very favorable experimental and genetic flexibility. Dictyostelium shows developmental vacuolar cell death, and in vitro monolayer approaches revealed both an autophagic vacuolar and a necrotic type of cell death. These are described in some detail, as well as implications and future prospects.


Assuntos
Autofagia , Dictyostelium/fisiologia , Necrose , Animais , Morte Celular
18.
Methods Mol Biol ; 346: 535-50, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16957313

RESUMO

In this chapter, we describe how to conveniently demonstrate, assess, and study cell death in Dictyostelium through simple cell culture, clonogenic tests, and photonic (with the help of staining techniques) and electronic microscopy. Cell death can be convniently generated using minor modifications of the monolayer technique of Rob Kay et al., and either wild-type HMX44A Dictyostelium cells or the corresponding atg1- autophagy gene mutant cells. Methods to follow cell death qualitatively and quantitatively facilitate detailed studies of vacuolar death in wild-type cells and of nonvacuolar, "condensed" death in atg1- mutant cells.


Assuntos
Autofagia , Morte Celular , Dictyostelium , Animais , Ensaio de Unidades Formadoras de Colônias , Dictyostelium/genética , Dictyostelium/metabolismo , Dictyostelium/ultraestrutura , Citometria de Fluxo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...