Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Dev Cell ; 57(22): 2584-2598.e11, 2022 11 21.
Article in English | MEDLINE | ID: mdl-36413951

ABSTRACT

Autophagy is an essential catabolic process that promotes the clearance of surplus or damaged intracellular components. Loss of autophagy in age-related human pathologies contributes to tissue degeneration through a poorly understood mechanism. Here, we identify an evolutionarily conserved role of autophagy from yeast to humans in the preservation of nicotinamide adenine dinucleotide (NAD) levels, which are critical for cell survival. In respiring mouse fibroblasts with autophagy deficiency, loss of mitochondrial quality control was found to trigger hyperactivation of stress responses mediated by NADases of PARP and Sirtuin families. Uncontrolled depletion of the NAD(H) pool by these enzymes ultimately contributed to mitochondrial membrane depolarization and cell death. Pharmacological and genetic interventions targeting several key elements of this cascade improved the survival of autophagy-deficient yeast, mouse fibroblasts, and human neurons. Our study provides a mechanistic link between autophagy and NAD metabolism and identifies targets for interventions in human diseases associated with autophagic, lysosomal, and mitochondrial dysfunction.


Subject(s)
NAD , Saccharomyces cerevisiae , Animals , Mice , Humans , Cell Survival , Autophagy , Cell Death
2.
Geroscience ; 44(4): 1961-1974, 2022 08.
Article in English | MEDLINE | ID: mdl-35355221

ABSTRACT

Mitochondrial reactive oxygen species (mtROS) are cellular messengers essential for cellular homeostasis. In response to stress, reverse electron transport (RET) through respiratory complex I generates high levels of mtROS. Suppression of ROS production via RET (ROS-RET) reduces survival under stress, while activation of ROS-RET extends lifespan in basal conditions. Here, we demonstrate that ROS-RET signalling requires increased electron entry and uninterrupted electron flow through the electron transport chain (ETC). We find that in old fruit flies, ROS-RET is abolished when electron flux is decreased and that their mitochondria produce consistently high levels of mtROS. Finally, we demonstrate that in young flies, limiting electron exit, but not entry, from the ETC phenocopies mtROS generation observed in old individuals. Our results elucidate the mechanism by which ROS signalling is lost during ageing.


Subject(s)
Diptera , Electrons , Animals , Reactive Oxygen Species , Electron Transport , Aging
3.
Redox Biol ; 32: 101450, 2020 05.
Article in English | MEDLINE | ID: mdl-32146156

ABSTRACT

Reactive Oxygen Species (ROS) are essential cellular messengers required for cellular homeostasis and regulate the lifespan of several animal species. The main site of ROS production is the mitochondrion, and within it, respiratory complex I (CI) is the main ROS generator. ROS produced by CI trigger several physiological responses that are essential for the survival of neurons, cardiomyocytes and macrophages. Here, we show that CI produces ROS when electrons flow in either the forward (Forward Electron Transport, FET) or reverse direction (Reverse Electron Transport, RET). We demonstrate that ROS production via RET (ROS-RET) is activated under thermal stress conditions and that interruption of ROS-RET production, through ectopic expression of the alternative oxidase AOX, attenuates the activation of pro-survival pathways in response to stress. Accordingly, we find that both suppressing ROS-RET signalling or decreasing levels of mitochondrial H2O2 by overexpressing mitochondrial catalase (mtCAT), reduces survival dramatically in flies under stress. Our results uncover a specific ROS signalling pathway where hydrogen peroxide (H2O2) generated by CI via RET is required to activate adaptive mechanisms, maximising survival under stress conditions.


Subject(s)
Drosophila melanogaster , Electron Transport Complex I , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Electron Transport , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Hydrogen Peroxide , Reactive Oxygen Species/metabolism
4.
J Gerontol A Biol Sci Med Sci ; 74(12): 1835-1843, 2019 11 13.
Article in English | MEDLINE | ID: mdl-29945183

ABSTRACT

Aging is a multifactorial process which affects all animals. Aging as a result of damage accumulation is the most accepted explanation but the proximal causes remain to be elucidated. There is also evidence indicating that aging has an important genetic component. Animal species age at different rates and specific signaling pathways, such as insulin/insulin-like growth factor, can regulate life span of individuals within a species by reprogramming cells in response to environmental changes. Here, we use an unbiased approach to identify novel factors that regulate life span in Drosophila melanogaster. We compare the transcriptome and metabolome of two wild-type strains used widely in aging research: short-lived Dahomey and long-lived Oregon R flies. We found that Dahomey flies carry several traits associated with short-lived individuals and species such as increased lipoxidative stress, decreased mitochondrial gene expression, and increased Target of Rapamycin signaling. Dahomey flies also have upregulated octopamine signaling known to stimulate foraging behavior. Accordingly, we present evidence that increased foraging behavior, under laboratory conditions where nutrients are in excess increases damage generation and accelerates aging. In summary, we have identified several new pathways, which influence longevity highlighting the contribution and importance of the genetic component of aging.


Subject(s)
Aging/genetics , Aging/physiology , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Animals , Gene Expression , Longevity/genetics , Longevity/physiology , Metabolome/genetics , Metabolome/physiology , Oxidative Stress/genetics , Oxidative Stress/physiology , Phenotype , Signal Transduction/genetics , Signal Transduction/physiology , Transcriptome/genetics , Transcriptome/physiology
5.
Nat Commun ; 9(1): 256, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29343728

ABSTRACT

Cellular homoeostatic pathways such as macroautophagy (hereinafter autophagy) are regulated by basic mechanisms that are conserved throughout the eukaryotic kingdom. However, it remains poorly understood how these mechanisms further evolved in higher organisms. Here we describe a modification in the autophagy pathway in vertebrates, which promotes its activity in response to oxidative stress. We have identified two oxidation-sensitive cysteine residues in a prototypic autophagy receptor SQSTM1/p62, which allow activation of pro-survival autophagy in stress conditions. The Drosophila p62 homologue, Ref(2)P, lacks these oxidation-sensitive cysteine residues and their introduction into the protein increases protein turnover and stress resistance of flies, whereas perturbation of p62 oxidation in humans may result in age-related pathology. We propose that the redox-sensitivity of p62 may have evolved in vertebrates as a mechanism that allows activation of autophagy in response to oxidative stress to maintain cellular homoeostasis and increase cell survival.


Subject(s)
Autophagy , Proteostasis , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein/metabolism , Amino Acid Sequence , Animals , Cell Survival/drug effects , Cells, Cultured , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , HEK293 Cells , HeLa Cells , Humans , Hydrogen Peroxide/pharmacology , Mice, Knockout , Oxidants/pharmacology , Oxidation-Reduction , Sequence Homology, Amino Acid , Sequestosome-1 Protein/genetics
6.
FEBS Lett ; 592(5): 743-758, 2018 03.
Article in English | MEDLINE | ID: mdl-29106705

ABSTRACT

The brain is the most complex human organ, consuming more energy than any other tissue in proportion to its size. It relies heavily on mitochondria to produce energy and is made up of mitotic and postmitotic cells that need to closely coordinate their metabolism to maintain essential bodily functions. During aging, damaged mitochondria that produce less ATP and more reactive oxygen species (ROS) accumulate. The current consensus is that ROS cause oxidative stress, damaging mitochondria and resulting in an energetic crisis that triggers neurodegenerative diseases and accelerates aging. However, in model organisms, increasing mitochondrial ROS (mtROS) in the brain extends lifespan, suggesting that ROS may participate in signaling that protects the brain. Here, we summarize the mechanisms by which mtROS are produced at the molecular level, how different brain cells and regions produce different amounts of mtROS, and how mtROS levels change during aging. Finally, we critically discuss the possible roles of ROS in aging as signaling molecules and damaging agents, addressing whether age-associated increases in mtROS are a cause or a consequence of aging.


Subject(s)
Aging/metabolism , Brain/metabolism , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Aging/genetics , Aging/pathology , Animals , Brain/pathology , Energy Metabolism , Humans , Mitochondria/genetics , Mitochondria/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology
7.
Cell Stress ; 2(4): 91-93, 2018 Mar 23.
Article in English | MEDLINE | ID: mdl-31225472

ABSTRACT

Ageing and age-related diseases are characterised by increased oxidative and proteotoxic stress, which results in negative effects on cell function and survival. The cell possesses several mechanisms to deal with damaged proteins, including degradation via macroautophagy (hereafter called autophagy). This essential cellular pathway is conserved from yeast to humans and it is well established that its impairment reduces lifespan in multiple model organisms, including worms, flies and mice. In our study, recently published in Nature Communications, we asked if longer lifespan characteristic of higher organisms is the result of evolutionary adaptations to the autophagy machinery. We found that the autophagy receptor p62 can be oxidised leading to its oligomerisation which ultimately promotes autophagy. However this mechanism, present in vertebrates, has been acquired late in evolution. We propose that the ability of p62 to sense reactive oxygen species (ROS) via oxidation, and potentially other similar modifications, may have evolved in higher organisms and contributed to their increased lifespan. Indeed, impairment of this process could result in age-related neurodegeneration in humans.

8.
PLoS One ; 11(8): e0161817, 2016.
Article in English | MEDLINE | ID: mdl-27570965

ABSTRACT

Drosophila melanogaster is a popular research model organism thanks to its' powerful genetic tools that allow spatial and temporal control of gene expression. The inducible GeneSwitch Gal4 system (GS) system is a modified version of the classic UAS/GAL4 system which allows inducible regulation of gene expression and eliminates background effects. It is widely acknowledged that the GS system is leaky, with low level expression of UAS transgenes in absence of the inducer RU-486 (the progesterone analog that activates the modified GAL4 protein). However, in the course of our experiments, we have observed that the extent of this leak depends on the nature of the transgene being expressed. In the absence of RU-486, when strong drivers are used to express protein coding transgenes, leaky expression is low or negligible, however expression of RNA interference (RNAi) transgenes results in complete depletion of protein levels. The majority of published studies, using the GS system and RNAi transgenes validate knock-down efficiency by comparing target gene mRNA levels between induced and non-induced groups. Here, we demonstrate that this approach is lacking and that both additional control groups and further validation is required at the protein level. Unfortunately, this experimental limitation of the GS system eliminates "the background advantage", but does offer the possibility of performing more complex experiments (e.g. studying depletion and overexpression of different proteins in the same genetic background). The limitations and new possible applications of the GS system are discussed in detail.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression/drug effects , Mifepristone/pharmacology , RNA Interference/physiology , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics , Transgenes/genetics
9.
Cell Rep ; 6(5): 855-67, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24582964

ABSTRACT

High tumor burden is associated with increased levels of circulating inflammatory cytokines that influence the pathophysiology of the tumor and its environment. The cellular and molecular events mediating the organismal response to a growing tumor are poorly understood. Here, we report a bidirectional crosstalk between epithelial tumors and the fat body-a peripheral immune tissue-in Drosophila. Tumors trigger a systemic immune response through activation of Eiger/TNF signaling, which leads to Toll pathway upregulation in adipocytes. Reciprocally, Toll elicits a non-tissue-autonomous program in adipocytes, which drives tumor cell death. Hemocytes play a critical role in this system by producing the ligands Spätzle and Eiger, which are required for Toll activation in the fat body and tumor cell death. Altogether, our results provide a paradigm for a long-range tumor suppression function of adipocytes in Drosophila, which may represent an evolutionarily conserved mechanism in the organismal response to solid tumors.


Subject(s)
Adipocytes/metabolism , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Toll-Like Receptors/metabolism , Animals , Apoptosis/physiology , Carcinogenesis/metabolism , Cell Growth Processes/physiology , Drosophila melanogaster , Female , Hemocytes/cytology , Hemocytes/metabolism , Male , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
10.
Gut ; 63(3): 480-93, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23585469

ABSTRACT

OBJECTIVE: Colorectal cancer (CRC) is a major contributor to cancer mortality and morbidity. LIM kinase 2 (LIMK2) promotes tumour cell invasion and metastasis. The objectives of this study were to determine how LIMK2 expression is associated with CRC progression and patient outcome, and to use genetically modified Drosophila and mice to determine how LIMK2 deletion affects gastrointestinal stem cell regulation and tumour development. DESIGN: LIMK2 expression and activity were measured by immunostaining tumours from CRC-prone mice, human CRC cell lines and 650 human tumours. LIMK knockdown in Drosophila or Limk2 deletion in mice allowed for assessment of their contributions to gastrointestinal stem cell homeostasis and tumour development. RESULTS: LIMK2 expression was reduced in intestinal tumours of cancer-prone mice, as well as in human CRC cell lines and tumours. Reduced LIMK2 expression and substrate phosphorylation were associated with shorter patient survival. Genetic analysis in Drosophila midgut and intestinal epithelial cells isolated from genetically modified mice revealed a conserved role for LIMK2 in constraining gastrointestinal stem cell proliferation. Limk2 deletion increased colon tumour size in a colitis-associated colorectal mouse cancer model. CONCLUSIONS: This study revealed that LIMK2 expression and activity progressively decrease with advancing stage, and supports the hypothesis that there is selective pressure for reduced LIMK2 expression in CRC to relieve negative constraints imposed upon gastrointestinal stem cells.


Subject(s)
Biomarkers, Tumor/metabolism , Colon/enzymology , Colorectal Neoplasms/enzymology , Intestinal Mucosa/enzymology , Lim Kinases/metabolism , Neoplastic Stem Cells/enzymology , Animals , Biomarkers, Tumor/deficiency , Cell Line, Tumor , Cell Proliferation , Colon/pathology , Colon/physiopathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/physiopathology , DNA Methylation , Disease Progression , Down-Regulation , Drosophila melanogaster , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Intestinal Mucosa/pathology , Intestinal Mucosa/physiopathology , Lim Kinases/deficiency , Mice , Mice, Knockout , Neoplastic Stem Cells/physiology , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis
11.
PLoS One ; 8(12): e83942, 2013.
Article in English | MEDLINE | ID: mdl-24349561

ABSTRACT

p120ctn is a ubiquitously expressed core component of cadherin junctions and essential for vertebrate development. Surprisingly, Drosophila p120ctn (dp120ctn) is dispensable for adherens junctions and development, which has discouraged Drosophila researchers from further pursuing the biological role of dp120ctn. Here we demonstrate that dp120ctn loss results in increased heat shock sensitivity and reduced animal lifespan, which are completely rescued by ectopic expression of a dp120ctn-GFP transgene. Transcriptomic analysis revealed multiple relish/NF-κB target genes differentially expressed upon loss of dp120ctn. Importantly, this aberrant gene expression was rescued by overexpression of dp120ctn-GFP or heterozygosity for relish. Our results uncover a novel role for dp120ctn in the regulation of animal stress response and immune signalling. This may represent an ancient role of p120ctn and can influence further studies in Drosophila and mammals.


Subject(s)
Catenins , Heat-Shock Response/physiology , Signal Transduction/physiology , Animals , Catenins/genetics , Catenins/immunology , Catenins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/immunology , Drosophila Proteins/metabolism , Drosophila melanogaster , Longevity/physiology , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism , Transcriptome/physiology , Delta Catenin
12.
Antioxid Redox Signal ; 19(16): 1953-69, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-22938137

ABSTRACT

SIGNIFICANCE: Aging is a consequence of the accumulation of cellular damage that impairs the capacity of an aging organism to adapt to stress. The Mitochondrial Free Radical Theory of Aging (MFRTA) has been one of the most influential ideas over the past 50 years. The MFRTA is supported by the accumulation of oxidative damage during aging along with comparative studies demonstrating that long-lived species or individuals produce fewer mitochondrial reactive oxygen species and have lower levels of oxidative damage. RECENT ADVANCES: Recently, however, species that combine high oxidative damage with a longer lifespan (i.e., naked mole rats) have been described. Moreover, most of the interventions based on antioxidant supplementation do not increase longevity, as would be predicted by the MFRTA. Studies to date provide a clear understanding that mitochondrial function regulates the rate of aging, but the underlying mechanisms remain unclear. CRITICAL ISSUES: Here, we review the reactive oxygen species (ROS)-dependent and ROS-independent mechanisms by which mitochondria can affect longevity. We discuss the role of different ROS (superoxide, hydrogen peroxide, and hydroxyl radical), both as oxidants as well as signaling molecules. We also describe how mitochondria can regulate longevity by ROS-independent mechanisms. We discuss alterations in mitochondrial DNA, accumulation of cellular waste as a consequence of glyco- and lipoxidative damage, and the regulation of DNA maintenance enzymes as mechanisms that can determine longevity without involving ROS. FUTURE DIRECTIONS: We also show how the regulation of longevity is a complex process whereby ROS-dependent and ROS-independent mechanisms interact to determine the maximum lifespan of species and individuals.


Subject(s)
Longevity/physiology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Animals , Electron Transport , Humans
13.
Development ; 139(24): 4524-35, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23172913

ABSTRACT

Inactivating mutations within adenomatous polyposis coli (APC), a negative regulator of Wnt signaling, are responsible for most sporadic and hereditary forms of colorectal cancer (CRC). Here, we use the adult Drosophila midgut as a model system to investigate the molecular events that mediate intestinal hyperplasia following loss of Apc in the intestine. Our results indicate that the conserved Wnt target Myc and its binding partner Max are required for the initiation and maintenance of intestinal stem cell (ISC) hyperproliferation following Apc1 loss. Importantly, we find that loss of Apc1 leads to the production of the interleukin-like ligands Upd2/3 and the EGF-like Spitz in a Myc-dependent manner. Loss of Apc1 or high Wg in ISCs results in non-cell-autonomous upregulation of upd3 in enterocytes and subsequent activation of Jak/Stat signaling in ISCs. Crucially, knocking down Jak/Stat or Spitz/Egfr signaling suppresses Apc1-dependent ISC hyperproliferation. In summary, our results uncover a novel non-cell-autonomous interplay between Wnt/Myc, Egfr and Jak/Stat signaling in the regulation of intestinal hyperproliferation. Furthermore, we present evidence suggesting potential conservation in mouse models and human CRC. Therefore, the Drosophila adult midgut proves to be a powerful genetic system to identify novel mediators of APC phenotypes in the intestine.


Subject(s)
Drosophila Proteins/physiology , Drosophila , ErbB Receptors/physiology , Intestines/pathology , Janus Kinases/physiology , Receptors, Invertebrate Peptide/physiology , STAT Transcription Factors/physiology , Transcription Factors/physiology , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Adult Stem Cells/physiology , Age Factors , Animals , Animals, Genetically Modified , Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome , DNA Replication/genetics , DNA Replication/physiology , Drosophila/genetics , Drosophila/growth & development , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Enterocytes/metabolism , Enterocytes/pathology , Enterocytes/physiology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Hyperplasia/genetics , Intestinal Mucosa/metabolism , Janus Kinases/genetics , Janus Kinases/metabolism , Receptor Cross-Talk/physiology , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
14.
EMBO J ; 31(19): 3901-17, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-22948071

ABSTRACT

The ability to regenerate following stress is a hallmark of self-renewing tissues. However, little is known about how regeneration differs from homeostatic tissue maintenance. Here, we study the role and regulation of Wingless (Wg)/Wnt signalling during intestinal regeneration using the Drosophila adult midgut. We show that Wg is produced by the intestinal epithelial compartment upon damage or stress and it is exclusively required for intestinal stem cell (ISC) proliferation during tissue regeneration. Reducing Wg or downstream signalling components from the intestinal epithelium blocked tissue regeneration. Importantly, we demonstrate that Wg from the undifferentiated progenitor cell, the enteroblast, is required for Myc-dependent ISC proliferation during regeneration. Similar to young regenerating tissues, ageing intestines required Wg and Myc for ISC hyperproliferation. Unexpectedly, our results demonstrate that epithelial but not mesenchymal Wg is essential for ISC proliferation in response to damage, while neither source of the ligand is solely responsible for ISC maintenance and tissue self-renewal in unchallenged tissues. Therefore, fine-tuning Wnt results in optimal balance between the ability to respond to stress without negatively affecting organismal viability.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/physiology , Intestines/physiology , Regeneration/physiology , Stem Cells/physiology , Wnt1 Protein/physiology , Animals , Cell Proliferation , Female , Signal Transduction/physiology
15.
Cell Cycle ; 11(20): 3876-86, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22983063

ABSTRACT

DJ-1 (or PARK-7) is a multifunctional protein implicated in numerous pathologies including cancer, sterility and Parkinson disease (PD). The popular genetic model Drosophila melanogaster has two orthologs, dj-1: α and ß. Dysfunction of dj-1ß strongly impairs fly mobility in an age-dependent manner. In this study, we analyze in detail the molecular mechanism underlying the dj-1ß mutant phenotype. Mitochondrial hydrogen peroxide production, but not superoxide production, was increased in mutant flies. An increase in peroxide leak from mitochondria causes oxidative damage elsewhere and explains the strong reduction in mobility caused by dj-1ß mutation. However, at the same time, increased levels of hydrogen peroxide activated a pro-survival program characterized by (1) an alteration in insulin-like signaling, (2) an increase in mitochondrial biogenesis and (3) an increase in the de-acetylase activity of sirtuins. The activation of this pro-survival program was associated with increased longevity under conditions of moderate oxidative stress. Additionally, the dj-1ß mutation unexpectedly accelerated development, a phenotype not previously associated with this mutation. Our results reveal an important role of dj-1ß in oxidative stress handling, insulin-like signaling and development in Drosophila melanogaster.


Subject(s)
Aging/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Hydrogen Peroxide/metabolism , Insulin/metabolism , Nerve Tissue Proteins/metabolism , Aging/genetics , Animals , Drosophila Proteins/genetics , Female , Gene Expression Regulation, Developmental , Longevity , Male , Mitochondria/metabolism , Mitochondrial Turnover/physiology , Motor Activity , Mutation , Nerve Tissue Proteins/genetics , Oxidation-Reduction , Oxidative Stress , Phenotype , Protein Deglycase DJ-1 , Signal Transduction/genetics , Sirtuins/genetics , Sirtuins/metabolism
16.
J Genet Genomics ; 38(10): 431-8, 2011 Oct 20.
Article in English | MEDLINE | ID: mdl-22035864

ABSTRACT

Invasion and metastasis are the most deadly hallmarks of cancer. Once a cancer has acquired the ability to colonize new sites in the body it becomes dramatically more difficult to treat. This has made it a focus of much of cancer research. The humble fruit fly, Drosophila melanogaster, has despite its relative simplicity, made significant contributions to the understanding of tumor progression. In this review we outline and highlight those with an emphasis on modeling the genetic and epigenetic changes required for invasion and metastasis. We will revisit the early years of cancer modeling in Drosophila where the first parallels were drawn between Drosophila and vertebrate neoplasms and highlight recent advances using genetic screens and interactions with the epithelial microenvironment and innate immune system. We focus on the power and limitations of current fly models of metastasis.


Subject(s)
Drosophila/genetics , Gene Expression Regulation, Neoplastic , Immunity, Innate/genetics , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Tumor Microenvironment/genetics , Animals , DNA Methylation , Disease Models, Animal , Epigenesis, Genetic/genetics , Humans
17.
Cell Cycle ; 10(10): 1528-32, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21471732

ABSTRACT

Mitochondria are considered major regulators of longevity, although their exact role in aging is not fully understood. Data from different laboratories show a negative correlation between reactive oxygen species (ROS) generated by complex I and lifespan. This suggests that complex I has a central role in the regulation of longevity. Here, we review data that both support and refute the role of complex I as a pacemaker of aging. We include data from our laboratory, where we have manipulated ROS production by the electron transport chain (ETC) in Drosophila melanogaster. The bypass of complex I increases the lifespan of the fruit fly, but it is not clear if this is caused by a reduction in ROS or by a change in the NAD+ to NADH ratio. We propose that complex I regulates aging through at least two mechanisms: (1) an ROS-dependent mechanism that leads to mitochondrial DNA damage and (2) an ROS-independent mechanism through the control of the NAD+ to NADH ratio. Control of the relative levels of NAD+ and NADH would allow the regulation of (1) glyco- and (2) lipoxidative-damage and (3) the activation of sirtuins.


Subject(s)
Aging , Electron Transport Complex I/metabolism , Mitochondria/metabolism , Animals , DNA Damage , Drosophila melanogaster/metabolism , NAD/metabolism , Reactive Oxygen Species/metabolism
18.
Dev Cell ; 18(6): 999-1011, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20627081

ABSTRACT

The roles of inflammatory cytokines and the immune response in cancer remain paradoxical. In the case of tumor necrosis factor (TNF), there is undisputed evidence indicating both protumor and antitumor activities. Recent work in Drosophila indicated that a TNF-dependent mechanism eliminates cells deficient for the polarity tumor suppressors dlg or scrib. In this study, however, we show that in tumors deficient for scrib that also expressed the Ras oncoprotein, the TNF signal was diverted into a protumor signal that enhanced tumor growth through larval arrest and stimulated invasive migration. In this case, TNF promoted malignancy and was detrimental to host survival. TNF was expressed at high levels by tumor-associated hemocytes recruited from the circulation. The expression of TNF by hemocytes was both necessary and sufficient to trigger TNF signaling in tumor cells. Our evidence suggests that tumors can evolve into malignancy through oncogenic Ras activation and the hijacking of TNF signaling.


Subject(s)
Carcinogens/metabolism , Neoplasms/metabolism , Oncogenes/physiology , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Proteins/metabolism , ras Proteins/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Gene Expression Regulation, Neoplastic/immunology , Hemocytes/cytology , Hemocytes/immunology , Hemocytes/metabolism , Membrane Proteins , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/immunology , Neoplasms/genetics , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/genetics , Tumor Suppressor Proteins/genetics , ras Proteins/genetics
19.
Aging (Albany NY) ; 2(4): 200-23, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20453260

ABSTRACT

The Mitochondrial Free Radical Theory of Aging (MFRTA) is currently one of the most widely accepted theories used to explain aging. From MFRTA three basic predictions can be made: long-lived individuals or species should produce fewer mitochondrial Reactive Oxygen Species (mtROS) than short-lived individuals or species; a decrease in mtROS production will increase lifespan; and an increase in mtROS production will decrease lifespan. It is possible to add a further fourth prediction: if ROS is controlling longevity separating these parameters through selection would be impossible. These predictions have been tested in Drosophila melanogaster. Firstly, we studied levels of mtROS production and lifespan of three wild-type strains of Drosophila, Oregon R, Canton S and Dahomey. Oregon R flies live the longest and produce significantly fewer mtROS than both Canton S and Dahomey. These results are therefore in accordance with the first prediction. A new transgenic Drosophila model expressing the Ciona intestinalis Alternative Oxidase (AOX) was used to test the second prediction. In fungi and plants, AOX expression regulates both free radical production and lifespan. In Drosophila, AOX expression decreases mtROS production, but does not increase lifespan. This result contradicts the second prediction of MFRTA. The third prediction was tested in flies mutant for the gene dj-1beta. These flies are characterized by an age-associated decline in locomotor function and increased levels of mtROS production. Nevertheless, dj-1beta mutant flies do not display decreased lifespan, which again is in contradiction with MFRTA. In our final experiment we utilized flies with DAH mitochondrial DNA in an OR nuclear background, and OR mitochondrial DNA in DAH nuclear background. From this, Mitochondrial DNA does not control free radical production, but it does determine longevity of females independently of mtROS production. In summary, these results do not systematically support the predictions of the MFRTA. Accordingly, MFRTA should be revised to accommodate these findings.


Subject(s)
Aging/metabolism , Drosophila melanogaster/metabolism , Longevity , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Animals , Female , Male , Oxygen Consumption , Species Specificity
20.
Proc Natl Acad Sci U S A ; 107(20): 9105-10, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20435911

ABSTRACT

Mutations in mitochondrial oxidative phosphorylation complex I are associated with multiple pathologies, and complex I has been proposed as a crucial regulator of animal longevity. In yeast, the single-subunit NADH dehydrogenase Ndi1 serves as a non-proton-translocating alternative enzyme that replaces complex I, bringing about the reoxidation of intramitochondrial NADH. We have created transgenic strains of Drosophila that express yeast NDI1 ubiquitously. Mitochondrial extracts from NDI1-expressing flies displayed a rotenone-insensitive NADH dehydrogenase activity, and functionality of the enzyme in vivo was confirmed by the rescue of lethality resulting from RNAi knockdown of complex I. NDI1 expression increased median, mean, and maximum lifespan independently of dietary restriction, and with no change in sirtuin activity. NDI1 expression mitigated the aging associated decline in respiratory capacity and the accompanying increase in mitochondrial reactive oxygen species production, and resulted in decreased accumulation of markers of oxidative damage in aged flies. Our results support a central role of mitochondrial oxidative phosphorylation complex I in influencing longevity via oxidative stress, independently of pathways connected to nutrition and growth signaling.


Subject(s)
Aging/metabolism , Drosophila melanogaster/physiology , Electron Transport Complex I/metabolism , Longevity/physiology , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Animals , Blotting, Western , Caloric Restriction , Drosophila melanogaster/enzymology , Electron Transport Complex I/genetics , Histocytochemistry , Longevity/genetics , Mitochondria/metabolism , Oxidative Stress/genetics , Oxidative Stress/physiology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...