Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33593901

RESUMO

Reduced activity of insulin/insulin-like growth factor signaling (IIS) increases healthy lifespan among diverse animal species. Downstream of IIS, multiple evolutionarily conserved transcription factors (TFs) are required; however, distinct TFs are likely responsible for these effects in different tissues. Here we have asked which TFs can extend healthy lifespan within distinct cell types of the adult nervous system in Drosophila Starting from published single-cell transcriptomic data, we report that forkhead (FKH) is endogenously expressed in neurons, whereas forkhead-box-O (FOXO) is expressed in glial cells. Accordingly, we find that neuronal FKH and glial FOXO exert independent prolongevity effects. We have further explored the role of neuronal FKH in a model of Alzheimer's disease-associated neuronal dysfunction, where we find that increased neuronal FKH preserves behavioral function and reduces ubiquitinated protein aggregation. Finally, using transcriptomic profiling, we identify Atg17, a member of the Atg1 autophagy initiation family, as one FKH-dependent target whose neuronal overexpression is sufficient to extend healthy lifespan. Taken together, our results underscore the importance of cell type-specific mapping of TF activity to preserve healthy function with age.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Longevidade , Neuroglia/metabolismo , Neurônios/metabolismo , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Feminino , Fatores de Transcrição Forkhead/genética , Perfilação da Expressão Gênica , Masculino , Neuroglia/citologia , Neurônios/citologia , Transcriptoma
2.
PLoS Genet ; 16(11): e1009083, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33253201

RESUMO

Increased cellular degradation by autophagy is a feature of many interventions that delay ageing. We report here that increased autophagy is necessary for reduced insulin-like signalling (IIS) to extend lifespan in Drosophila and is sufficient on its own to increase lifespan. We first established that the well-characterised lifespan extension associated with deletion of the insulin receptor substrate chico was completely abrogated by downregulation of the essential autophagy gene Atg5. We next directly induced autophagy by over-expressing the major autophagy kinase Atg1 and found that a mild increase in autophagy extended lifespan. Interestingly, strong Atg1 up-regulation was detrimental to lifespan. Transcriptomic and metabolomic approaches identified specific signatures mediated by varying levels of autophagy in flies. Transcriptional upregulation of mitochondrial-related genes was the signature most specifically associated with mild Atg1 upregulation and extended lifespan, whereas short-lived flies, possessing strong Atg1 overexpression, showed reduced mitochondrial metabolism and up-regulated immune system pathways. Increased proteasomal activity and reduced triacylglycerol levels were features shared by both moderate and high Atg1 overexpression conditions. These contrasting effects of autophagy on ageing and differential metabolic profiles highlight the importance of fine-tuning autophagy levels to achieve optimal healthspan and disease prevention.


Assuntos
Autofagia/genética , Longevidade/genética , Mitocôndrias/genética , Envelhecimento/genética , Animais , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Genes Mitocondriais/genética , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Proteínas Serina-Treonina Quinases/genética , Receptor de Insulina/genética , Transdução de Sinais
3.
Hum Mol Genet ; 29(14): 2420-2434, 2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32592479

RESUMO

Alzheimer's disease (AD) is the most common form of dementia and the most prevalent neurodegenerative disease. Genome-wide association studies have linked PICALM to AD risk. PICALM has been implicated in Aß42 production and turnover, but whether it plays a direct role in modulating Aß42 toxicity remains unclear. We found that increased expression of the Drosophila PICALM orthologue lap could rescue Aß42 toxicity in an adult-onset model of AD, without affecting Aß42 level. Imbalances in the glutamatergic system, leading to excessive, toxic stimulation, have been associated with AD. We found that Aß42 caused the accumulation of presynaptic vesicular glutamate transporter (VGlut) and increased spontaneous glutamate release. Increased lap expression reversed these phenotypes back to control levels, suggesting that lap may modulate glutamatergic transmission. We also found that lap modulated the localization of amphiphysin (Amph), the homologue of another AD risk factor BIN1, and that Amph itself modulated postsynaptic glutamate receptor (GluRII) localization. We propose a model where PICALM modulates glutamatergic transmission, together with BIN1, to ameliorate synaptic dysfunction and disease progression.


Assuntos
Doença de Alzheimer/genética , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteínas de Drosophila/genética , Receptores Ionotrópicos de Glutamato/genética , Fatores de Transcrição/genética , Proteínas Vesiculares de Transporte de Glutamato/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Comportamento Animal , Drosophila melanogaster/genética , Fármacos Atuantes sobre Aminoácidos Excitatórios , Humanos , Proteínas Monoméricas de Montagem de Clatrina/genética , Proteínas do Tecido Nervoso/genética , Fragmentos de Peptídeos/genética , Transmissão Sináptica/genética
4.
BMC Biol ; 18(1): 124, 2020 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-32928209

RESUMO

BACKGROUND: The increasing age of global populations highlights the urgent need to understand the biological underpinnings of ageing. To this end, inhibition of the insulin/insulin-like signalling (IIS) pathway can extend healthy lifespan in diverse animal species, but with trade-offs including delayed development. It is possible that distinct cell types underlie effects on development and ageing; cell-type-specific strategies could therefore potentially avoid negative trade-offs when targeting diseases of ageing, including prevalent neurodegenerative diseases. The highly conserved diversity of neuronal and non-neuronal (glial) cell types in the Drosophila nervous system makes it an attractive system to address this possibility. We have thus investigated whether IIS in distinct glial cell populations differentially modulates development and lifespan in Drosophila. RESULTS: We report here that glia-specific IIS inhibition, using several genetic means, delays development while extending healthy lifespan. The effects on lifespan can be recapitulated by adult-onset IIS inhibition, whereas developmental IIS inhibition is dispensable for modulation of lifespan. Notably, the effects we observe on both lifespan and development act through the PI3K branch of the IIS pathway and are dependent on the transcription factor FOXO. Finally, IIS inhibition in several glial subtypes can delay development without extending lifespan, whereas the same manipulations in astrocyte-like glia alone are sufficient to extend lifespan without altering developmental timing. CONCLUSIONS: These findings reveal a role for distinct glial subpopulations in the organism-wide modulation of development and lifespan, with IIS in astrocyte-like glia contributing to lifespan modulation but not to developmental timing. Our results enable a more complete picture of the cell-type-specific effects of the IIS network, a pathway whose evolutionary conservation in humans make it tractable for therapeutic interventions. Our findings therefore underscore the necessity for cell-type-specific strategies to optimise interventions for the diseases of ageing.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Insulina/fisiologia , Longevidade , Fosfatidilinositol 3-Quinases/fisiologia , Animais , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Neuroglia/classificação , Neuroglia/fisiologia , Transdução de Sinais
5.
J Neurosci ; 37(17): 4584-4592, 2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-28363982

RESUMO

Transforming growth factor-ß (TGF-ß) plays an important role in the development and maintenance of embryonic dopaminergic (DA) neurons in the midbrain. To study the function of TGF-ß signaling in the adult nigrostriatal system, we generated transgenic mice with reduced TGF-ß signaling in mature neurons. These mice display age-related motor deficits and degeneration of the nigrostriatal system. Increasing TGF-ß signaling in the substantia nigra through adeno-associated virus expressing a constitutively active type I receptor significantly reduces 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurodegeneration and motor deficits. These results suggest that TGF-ß signaling is critical for adult DA neuron survival and that modulating this signaling pathway has therapeutic potential in Parkinson disease.SIGNIFICANCE STATEMENT We show that reducing Transforming growth factor-ß (TGF-ß) signaling promotes Parkinson disease-related pathologies and motor deficits, and increasing TGF-ß signaling reduces neurotoxicity of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, a parkinsonism-inducing agent. Our results provide a rationale to pursue a means of increasing TGF-ß signaling as a potential therapy for Parkinson's disease.


Assuntos
Intoxicação por MPTP/fisiopatologia , Neostriado/fisiopatologia , Doenças Neurodegenerativas/fisiopatologia , Transdução de Sinais , Substância Negra/fisiopatologia , Fator de Crescimento Transformador beta/deficiência , Animais , Sobrevivência Celular/genética , Transtornos Neurológicos da Marcha/induzido quimicamente , Transtornos Neurológicos da Marcha/fisiopatologia , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Doenças Neurodegenerativas/induzido quimicamente , Equilíbrio Postural , Proteínas Serina-Treonina Quinases/genética , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/genética
6.
J Neurosci ; 36(46): 11654-11670, 2016 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-27852774

RESUMO

Glucocerebrosidase (GBA1) mutations are associated with Gaucher disease (GD), an autosomal recessive disorder caused by functional deficiency of glucocerebrosidase (GBA), a lysosomal enzyme that hydrolyzes glucosylceramide to ceramide and glucose. Neuronopathic forms of GD can be associated with rapid neurological decline (Type II) or manifest as a chronic form (Type III) with a wide spectrum of neurological signs. Furthermore, there is now a well-established link between GBA1 mutations and Parkinson's disease (PD), with heterozygote mutations in GBA1 considered the commonest genetic defect in PD. Here we describe a novel Drosophila model of GD that lacks the two fly GBA1 orthologs. This knock-out model recapitulates the main features of GD at the cellular level with severe lysosomal defects and accumulation of glucosylceramide in the fly brain. We also demonstrate a block in autophagy flux in association with reduced lifespan, age-dependent locomotor deficits and accumulation of autophagy substrates in dGBA-deficient fly brains. Furthermore, mechanistic target of rapamycin (mTOR) signaling is downregulated in dGBA knock-out flies, with a concomitant upregulation of Mitf gene expression, the fly ortholog of mammalian TFEB, likely as a compensatory response to the autophagy block. Moreover, the mTOR inhibitor rapamycin is able to partially ameliorate the lifespan, locomotor, and oxidative stress phenotypes. Together, our results demonstrate that this dGBA1-deficient fly model is a useful platform for the further study of the role of lysosomal-autophagic impairment and the potential therapeutic benefits of rapamycin in neuronopathic GD. These results also have important implications for the role of autophagy and mTOR signaling in GBA1-associated PD SIGNIFICANCE STATEMENT: We developed a Drosophila model of neuronopathic GD by knocking-out the fly orthologs of the GBA1 gene, demonstrating abnormal lysosomal pathology in the fly brain. Functioning lysosomes are required for autophagosome-lysosomal fusion in the autophagy pathway. We show in vivo that autophagy is impaired in dGBA-deficient fly brains. In response, mechanistic target of rapamycin (mTOR) activity is downregulated in dGBA-deficient flies and rapamycin ameliorates the lifespan, locomotor, and oxidative stress phenotypes. dGBA knock-out flies also display an upregulation of the Drosophila ortholog of mammalian TFEB, Mitf, a response that is unable to overcome the autophagy block. Together, our results suggest that rapamycin may have potential benefits in the treatment of GD, as well as PD linked to GBA1 mutations.


Assuntos
Modelos Animais de Doenças , Doença de Gaucher/metabolismo , Doença de Gaucher/prevenção & controle , Glucosilceramidase/genética , Lisossomos/metabolismo , Sirolimo/administração & dosagem , Serina-Treonina Quinases TOR/metabolismo , Animais , Animais Geneticamente Modificados , Autofagia/efeitos dos fármacos , Drosophila , Doença de Gaucher/patologia , Técnicas de Inativação de Genes , Transdução de Sinais/efeitos dos fármacos
7.
Brain ; 139(Pt 7): 2063-81, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27190010

RESUMO

Identifying preventive targets for Alzheimer's disease is a central challenge of modern medicine. Non-steroidal anti-inflammatory drugs, which inhibit the cyclooxygenase enzymes COX-1 and COX-2, reduce the risk of developing Alzheimer's disease in normal ageing populations. This preventive effect coincides with an extended preclinical phase that spans years to decades before onset of cognitive decline. In the brain, COX-2 is induced in neurons in response to excitatory synaptic activity and in glial cells in response to inflammation. To identify mechanisms underlying prevention of cognitive decline by anti-inflammatory drugs, we first identified an early object memory deficit in APPSwe-PS1ΔE9 mice that preceded previously identified spatial memory deficits in this model. We modelled prevention of this memory deficit with ibuprofen, and found that ibuprofen prevented memory impairment without producing any measurable changes in amyloid-ß accumulation or glial inflammation. Instead, ibuprofen modulated hippocampal gene expression in pathways involved in neuronal plasticity and increased levels of norepinephrine and dopamine. The gene most highly downregulated by ibuprofen was neuronal tryptophan 2,3-dioxygenase (Tdo2), which encodes an enzyme that metabolizes tryptophan to kynurenine. TDO2 expression was increased by neuronal COX-2 activity, and overexpression of hippocampal TDO2 produced behavioural deficits. Moreover, pharmacological TDO2 inhibition prevented behavioural deficits in APPSwe-PS1ΔE9 mice. Taken together, these data demonstrate broad effects of cyclooxygenase inhibition on multiple neuronal pathways that counteract the neurotoxic effects of early accumulating amyloid-ß oligomers.


Assuntos
Doença de Alzheimer/prevenção & controle , Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Transtornos da Memória/prevenção & controle , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase , Modelos Animais de Doenças , Regulação para Baixo , Eletroencefalografia , Ibuprofeno , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Reconhecimento Psicológico/efeitos dos fármacos , Triptofano Oxigenase/efeitos dos fármacos
8.
J Neurosci ; 34(17): 5882-94, 2014 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-24760848

RESUMO

A persistent and nonresolving inflammatory response to accumulating Aß peptide species is a cardinal feature in the development of Alzheimer's disease (AD). In response to accumulating Aß peptide species, microglia, the innate immune cells of the brain, generate a toxic inflammatory response that accelerates synaptic and neuronal injury. Many proinflammatory signaling pathways are linked to progression of neurodegeneration. However, endogenous anti-inflammatory pathways capable of suppressing Aß-induced inflammation represent a relatively unexplored area. Here we report that signaling through the prostaglandin-E2 (PGE2) EP4 receptor potently suppresses microglial inflammatory responses to Aß42 peptides. In cultured microglial cells, EP4 stimulation attenuated levels of Aß42-induced inflammatory factors and potentiated phagocytosis of Aß42. Microarray analysis demonstrated that EP4 stimulation broadly opposed Aß42-driven gene expression changes in microglia, with enrichment for targets of IRF1, IRF7, and NF-κB transcription factors. In vivo, conditional deletion of microglial EP4 in APPSwe-PS1ΔE9 (APP-PS1) mice conversely increased inflammatory gene expression, oxidative protein modification, and Aß deposition in brain at early stages of pathology, but not at later stages, suggesting an early anti-inflammatory function of microglial EP4 signaling in the APP-PS1 model. Finally, EP4 receptor levels decreased significantly in human cortex with progression from normal to AD states, suggesting that early loss of this beneficial signaling system in preclinical AD development may contribute to subsequent progression of pathology.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/farmacologia , Inflamação/metabolismo , Microglia/metabolismo , Fragmentos de Peptídeos/farmacologia , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Transdução de Sinais/fisiologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Humanos , Inflamação/patologia , Éteres Metílicos/farmacologia , Microglia/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP4/agonistas , Transdução de Sinais/efeitos dos fármacos
9.
J Neurosci ; 33(40): 16016-32, 2013 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-24089506

RESUMO

Prostaglandin E2 (PGE2), a potent lipid signaling molecule, modulates inflammatory responses through activation of downstream G-protein coupled EP(1-4) receptors. Here, we investigated the cell-specific in vivo function of PGE2 signaling through its E-prostanoid 2 (EP2) receptor in murine innate immune responses systemically and in the CNS. In vivo, systemic administration of lipopolysaccharide (LPS) resulted in a broad induction of cytokines and chemokines in plasma that was significantly attenuated in EP2-deficient mice. Ex vivo stimulation of peritoneal macrophages with LPS elicited proinflammatory responses that were dependent on EP2 signaling and that overlapped with in vivo plasma findings, suggesting that myeloid-lineage EP2 signaling is a major effector of innate immune responses. Conditional deletion of the EP2 receptor in myeloid lineage cells in Cd11bCre;EP2(lox/lox) mice attenuated plasma inflammatory responses and transmission of systemic inflammation to the brain was inhibited, with decreased hippocampal inflammatory gene expression and cerebral cortical levels of IL-6. Conditional deletion of EP2 significantly blunted microglial and astrocytic inflammatory responses to the neurotoxin MPTP and reduced striatal dopamine turnover. Suppression of microglial EP2 signaling also increased numbers of dopaminergic (DA) neurons in the substantia nigra independent of MPTP treatment, suggesting that microglial EP2 may influence development or survival of DA neurons. Unbiased microarray analysis of microglia isolated from adult Cd11bCre;EP2(lox/lox) and control mice demonstrated a broad downregulation of inflammatory pathways with ablation of microglial EP2 receptor. Together, these data identify a cell-specific proinflammatory role for macrophage/microglial EP2 signaling in innate immune responses systemically and in brain.


Assuntos
Encéfalo/metabolismo , Inflamação/metabolismo , Macrófagos/metabolismo , Microglia/metabolismo , Receptores de Prostaglandina E Subtipo EP2/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Inflamação/induzido quimicamente , Inflamação/genética , Lipopolissacarídeos , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP2/genética
10.
Ann Neurol ; 72(5): 788-98, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22915243

RESUMO

OBJECTIVE: There is significant evidence for a central role of inflammation in the development of Alzheimer disease (AD). Epidemiological studies indicate that chronic use of nonsteroidal anti-inflammatory drugs (NSAIDs) reduces the risk of developing AD in healthy aging populations. As NSAIDs inhibit the enzymatic activity of the inflammatory cyclooxygenases COX-1 and COX-2, these findings suggest that downstream prostaglandin signaling pathways function in the preclinical development of AD. Here, we investigate the function of prostaglandin E(2) (PGE(2) ) signaling through its EP3 receptor in the neuroinflammatory response to Aß peptide. METHODS: The function of PGE(2) signaling through its EP3 receptor was examined in vivo in a model of subacute neuroinflammation induced by administration of Aß(42) peptides. Our findings were then confirmed in young adult APPSwe-PS1ΔE9 transgenic mice. RESULTS: Deletion of the PGE(2) EP3 receptor in a model of Aß(42) peptide-induced neuroinflammation reduced proinflammatory gene expression, cytokine production, and oxidative stress. In the APPSwe-PS1ΔE9 model of familial AD, deletion of the EP3 receptor blocked induction of proinflammatory gene and protein expression and lipid peroxidation. In addition, levels of Aß peptides were significantly decreased, as were ß-secretase and ß C-terminal fragment levels, suggesting that generation of Aß peptides may be increased as a result of proinflammatory EP3 signaling. Finally, deletion of EP3 receptor significantly reversed the decline in presynaptic proteins seen in APPSwe-PS1ΔE9 mice. INTERPRETATION: Our findings identify the PGE(2) EP3 receptor as a novel proinflammatory, proamyloidogenic, and synaptotoxic signaling pathway, and suggest a role for COX-PGE(2) -EP3 signaling in the development of AD.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/metabolismo , Dinoprostona/metabolismo , Encefalite/metabolismo , Regulação da Expressão Gênica/genética , Transdução de Sinais/fisiologia , Fatores Etários , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/farmacologia , Precursor de Proteína beta-Amiloide/genética , Análise de Variância , Animais , Animais Recém-Nascidos , Ácido Aspártico Endopeptidases/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Proteínas de Ligação ao Cálcio , Células Cultivadas , Disfunção Cognitiva/patologia , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Encefalite/induzido quimicamente , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Fragmentos de Peptídeos/farmacologia , Prostaglandina-Endoperóxido Sintases/metabolismo , RNA Mensageiro/metabolismo , Receptores de Prostaglandina E Subtipo EP3/deficiência , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteína 25 Associada a Sinaptossoma/metabolismo , Proteína 2 Associada à Membrana da Vesícula/metabolismo
11.
Open Biol ; 13(9): 230171, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37699519

RESUMO

Alterations in the neuromuscular system underlie several neuromuscular diseases and play critical roles in the development of sarcopenia, the age-related loss of muscle mass and function. Mammalian Myostatin (MST) and GDF11, members of the TGF-ß superfamily of growth factors, are powerful regulators of muscle size in both model organisms and humans. Myoglianin (MYO), the Drosophila homologue of MST and GDF11, is a strong inhibitor of synaptic function and structure at the neuromuscular junction in flies. Here, we identified Plum, a transmembrane cell surface protein, as a modulator of MYO function in the larval neuromuscular system. Reduction of Plum in the larval body-wall muscles abolishes the previously demonstrated positive effect of attenuated MYO signalling on both muscle size and neuromuscular junction structure and function. In addition, downregulation of Plum on its own results in decreased synaptic strength and body weight, classifying Plum as a (novel) regulator of neuromuscular function and body (muscle) size. These findings offer new insights into possible regulatory mechanisms behind ageing- and disease-related neuromuscular dysfunctions in humans and identify potential targets for therapeutic interventions.


Assuntos
Drosophila melanogaster , Prunus domestica , Animais , Humanos , Envelhecimento , Proteínas Morfogenéticas Ósseas , Regulação para Baixo , Drosophila , Fatores de Diferenciação de Crescimento , Larva , Mamíferos
12.
Neurobiol Aging ; 132: 154-174, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37837732

RESUMO

Amyloid ß (Aß) accumulation is a hallmark of Alzheimer's disease. In adult Drosophila brains, human Aß overexpression harms climbing and lifespan. It's uncertain whether Aß is intrinsically toxic or activates downstream neurodegeneration pathways. Our study uncovers a novel protective role against Aß toxicity: intra-endoplasmic reticulum (ER) protein accumulation with a focus on laminin and collagen subunits. Despite high Aß, laminin B1 (LanB1) overexpression robustly counters toxicity, suggesting a potential Aß resistance mechanism. Other laminin subunits and collagen IV also alleviate Aß toxicity; combining them with LanB1 augments the effect. Imaging reveals ER retention of LanB1 without altering Aß secretion. LanB1's rescue function operates independently of the IRE1α/XBP1 ER stress response. ER-targeted GFP overexpression also mitigates Aß toxicity, highlighting broader ER protein retention advantages. Proof-of-principle tests in murine hippocampal slices using mouse Lamb1 demonstrate ER retention in transduced cells, indicating a conserved mechanism. Though ER protein retention generally harms, it could paradoxically counter neuronal Aß toxicity, offering a new therapeutic avenue for Alzheimer's disease.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Animais , Camundongos , Humanos , Peptídeos beta-Amiloides/toxicidade , Peptídeos beta-Amiloides/metabolismo , Drosophila , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Endorribonucleases/metabolismo , Laminina/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Estresse do Retículo Endoplasmático , Retículo Endoplasmático/metabolismo , Colágeno/metabolismo
13.
J Immunol ; 184(12): 7207-18, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20483760

RESUMO

Peripheral inflammation leads to immune responses in brain characterized by microglial activation, elaboration of proinflammatory cytokines and reactive oxygen species, and secondary neuronal injury. The inducible cyclooxygenase (COX), COX-2, mediates a significant component of this response in brain via downstream proinflammatory PG signaling. In this study, we investigated the function of the PGE2 E-prostanoid (EP) 4 receptor in the CNS innate immune response to the bacterial endotoxin LPS. We report that PGE2 EP4 signaling mediates an anti-inflammatory effect in brain by blocking LPS-induced proinflammatory gene expression in mice. This was associated in cultured murine microglial cells with decreased Akt and I-kappaB kinase phosphorylation and decreased nuclear translocation of p65 and p50 NF-kappaB subunits. In vivo, conditional deletion of EP4 in macrophages and microglia increased lipid peroxidation and proinflammatory gene expression in brain and in isolated adult microglia following peripheral LPS administration. Conversely, EP4 selective agonist decreased LPS-induced proinflammatory gene expression in hippocampus and in isolated adult microglia. In plasma, EP4 agonist significantly reduced levels of proinflammatory cytokines and chemokines, indicating that peripheral EP4 activation protects the brain from systemic inflammation. The innate immune response is an important component of disease progression in a number of neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. In addition, recent studies demonstrated adverse vascular effects with chronic administration of COX-2 inhibitors, indicating that specific PG signaling pathways may be protective in vascular function. This study supports an analogous and beneficial effect of PGE2 EP4 receptor signaling in suppressing brain inflammation.


Assuntos
Encéfalo/imunologia , Imunidade Inata/imunologia , Mediadores da Inflamação/imunologia , Receptores de Prostaglandina E/imunologia , Transdução de Sinais/imunologia , Animais , Western Blotting , Encéfalo/metabolismo , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Regulação da Expressão Gênica/imunologia , Imuno-Histoquímica , Inflamação/imunologia , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/imunologia , Microglia/metabolismo , Microscopia Confocal , Receptores de Prostaglandina E/metabolismo , Receptores de Prostaglandina E Subtipo EP4 , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Front Neurosci ; 15: 786076, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899176

RESUMO

Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are neurodegenerative disorders characterized by declining motor and cognitive functions. Even though these diseases present with distinct sets of symptoms, FTD and ALS are two extremes of the same disease spectrum, as they show considerable overlap in genetic, clinical and neuropathological features. Among these overlapping features, mitochondrial dysfunction is associated with both FTD and ALS. Recent studies have shown that cells derived from patients' induced pluripotent stem cells (iPSC)s display mitochondrial abnormalities, and similar abnormalities have been observed in a number of animal disease models. Drosophila models have been widely used to study FTD and ALS because of their rapid generation time and extensive set of genetic tools. A wide array of fly models have been developed to elucidate the molecular mechanisms of toxicity for mutations associated with FTD/ALS. Fly models have been often instrumental in understanding the role of disease associated mutations in mitochondria biology. In this review, we discuss how mutations associated with FTD/ALS disrupt mitochondrial function, and we review how the use of Drosophila models has been pivotal to our current knowledge in this field.

15.
Neural Regen Res ; 15(7): 1208-1219, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31960800

RESUMO

Alzheimer's disease is the leading cause of dementia. Its increased prevalence in developed countries, due to the sharp rise in ageing populations, presents one of the costliest challenges to modern medicine. In order to find disease-modifying therapies to confront this challenge, a more complete understanding of the pathogenesis of Alzheimer's disease is necessary. Recent studies have revealed increasing evidence for the roles played by microglia, the resident innate immune system cells of the brain. Reflecting the well-established roles of microglia in reacting to pathogens and inflammatory stimuli, there is now a growing literature describing both protective and detrimental effects for individual cytokines and chemokines produced by microglia in Alzheimer's disease. A smaller but increasing number of studies have also addressed the divergent roles played by microglial neurotrophic and neurogenic factors, and how their perturbation may play a key role in the pathogenesis of Alzheimer's disease. Here we review recent findings on the roles played by microglia in neuroinflammation, neuronal survival and neurogenesis in Alzheimer's disease. In each case, landmark studies have provided evidence for the divergent ways in which microglia can either promote neuronal function and survival, or perturb neuronal function, leading to cell death. In many cases, the secreted molecules of microglia can lead to divergent effects depending on the magnitude and context of microglial activation. This suggests that microglial functions must be maintained in a fine equilibrium, in order to support healthy neuronal function, and that the cellular microenvironment in the Alzheimer's disease brain disrupts this fine balance, leading to neurodegeneration. Thus, an understanding of microglial homeostasis, both in health and across the trajectory of the disease state, will improve our understanding of the pathogenic mechanisms underlying Alzheimer's disease, and will hopefully lead to the development of microglial-based therapeutic strategies to restore equilibrium in the Alzheimer's disease brain.

16.
Aging Cell ; 19(5): e13137, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32291952

RESUMO

Inhibition of signalling through several receptor tyrosine kinases (RTKs), including the insulin-like growth factor receptor and its orthologues, extends healthy lifespan in organisms from diverse evolutionary taxa. This raises the possibility that other RTKs, including those already well studied for their roles in cancer and developmental biology, could be promising targets for extending healthy lifespan. Here, we focus on anaplastic lymphoma kinase (Alk), an RTK with established roles in nervous system development and in multiple cancers, but whose effects on aging remain unclear. We find that several means of reducing Alk signalling, including mutation of its ligand jelly belly (jeb), RNAi knock-down of Alk, or expression of dominant-negative Alk in adult neurons, can extend healthy lifespan in female, but not male, Drosophila. Moreover, reduced Alk signalling preserves neuromuscular function with age, promotes resistance to starvation and xenobiotic stress, and improves night sleep consolidation. We find further that inhibition of Alk signalling in adult neurons modulates the expression of several insulin-like peptides, providing a potential mechanistic link between neuronal Alk signalling and organism-wide insulin-like signalling. Finally, we show that TAE-684, a small molecule inhibitor of Alk, can extend healthy lifespan in Drosophila, suggesting that the repurposing of Alk inhibitors may be a promising direction for strategies to promote healthy aging.


Assuntos
Quinase do Linfoma Anaplásico/metabolismo , Longevidade , Quinase do Linfoma Anaplásico/antagonistas & inibidores , Animais , Senescência Celular/efeitos dos fármacos , Drosophila , Feminino , Longevidade/efeitos dos fármacos , Masculino , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos
17.
Neuron ; 98(6): 1059-1061, 2018 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-29953866

RESUMO

Parkinson's disease patients report sleep disturbances well ahead of motor symptoms. In this issue of Neuron, Valadas et al. (2018) report that the disease genes pink1 and parkin exert novel, cell-type-specific effects to modulate ER-mitochondria contacts, neuropeptidergic transmission, and sleep patterns.


Assuntos
Doença de Parkinson , Humanos , Lipídeos , Mitocôndrias , Mutação , Neurônios , Ubiquitina-Proteína Ligases/genética
18.
Gene ; 368: 46-52, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16326033

RESUMO

We have utilized a transposable P element construct to scan the genome for modifiers of Drosophila Notch pathway phenotypes. From a collection of 2000 inserts we obtained two enhancers and one suppressor. Sequence analysis of the insertion regions demonstrated that two modifiers affect known components of the Notch pathway, whereas the third is an insert in the gene encoding ribosomal protein L13a at cytogenetic region 83B6-7. The insert in the RpL13A coding region creates a classic Minute mutation which enhances Notch pathway wing phenotypes. This report adds RpL13A to the list of Drosophila ribosomal protein genes that cause Minute phenotypes when mutated.


Assuntos
Proteínas de Drosophila/genética , Inativação Gênica , Receptores Notch/genética , Proteínas Ribossômicas/genética , Sequência de Aminoácidos , Animais , Elementos de DNA Transponíveis , Drosophila melanogaster , Elementos Facilitadores Genéticos , Feminino , Genes Supressores , Masculino , Dados de Sequência Molecular , Morfogênese/genética , Mutagênese Insercional , Mutação , Fenótipo , Proteínas Ribossômicas/antagonistas & inibidores , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Asas de Animais/embriologia
19.
ACS Chem Neurosci ; 7(4): 454-63, 2016 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-26979823

RESUMO

The neuroinflammatory response has received increasing attention as a key factor in the pathogenesis of Alzheimer's disease (AD). Microglia, the innate immune cells and resident phagocytes of the brain, respond to accumulating Aß peptides by generating a nonresolving inflammatory response. While this response can clear Aß peptides from the nervous system in some settings, its failure to do so in AD accelerates synaptic injury, neuronal loss, and cognitive decline. The complex molecular components of this response are beginning to be unraveled, with identification of both damaging and protective roles for individual components of the neuroinflammatory response. Even within one molecular pathway, contrasting effects are often present. As one example, recent studies of the inflammatory cyclooxygenase-prostaglandin pathway have revealed both beneficial and detrimental effects dependent on the disease context, cell type, and downstream signaling pathway. Nonsteroidal anti-inflammatory drugs (NSAIDs), which inhibit cyclooxygenases, are associated with reduced AD risk when taken by cognitively normal populations, but additional clinical and mouse model studies have added complexities and caveats to this finding. Downstream of cyclooxygenase activity, prostaglandin E2 signaling exerts both damaging pro-inflammatory and protective anti-inflammatory effects through actions of specific E-prostanoid G-protein coupled receptors on specific cell types. These complexities underscore the need for careful study of individual components of the neuroinflammatory response to better understand their contribution to AD pathogenesis and progression.


Assuntos
Doença de Alzheimer/complicações , Dinoprosta/metabolismo , Encefalite/etiologia , Transdução de Sinais/fisiologia , Animais , Dinoprosta/genética , Encefalite/prevenção & controle , Humanos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Transdução de Sinais/efeitos dos fármacos
20.
Curr Immunol Rev ; 11(2): 125-131, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28413375

RESUMO

The inflammatory response is a fundamental driving force in the pathogenesis of Alzheimer's disease (AD). In the setting of accumulating immunogenic Aß peptide assemblies, microglia, the innate immune cells of the brain, generate a non-resolving immune response and fail to adequately clear accumulating Aß peptides, accelerating neuronal and synaptic injury. Pathological, biomarker, and imaging studies point to a prominent role of the innate immune response in AD development, and the molecular components of this response are beginning to be unraveled. The inflammatory cyclooxygenase-PGE2 pathway is implicated in pre-clinical development of AD, both in epidemiology of normal aging populations and in transgenic mouse models of Familial AD. The cyclooxygenase-PGE2 pathway modulates the inflammatory response to accumulating Aß peptides through actions of specific E-prostanoid G-protein coupled receptors.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA