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(24): 2679-2682, 2022 Dec 19.
Article in English | MEDLINE | ID: mdl-36538893

ABSTRACT

In this selection, we celebrate the art of science by highlighting some of the submitted cover images from the past year. In this collection, our authors share the stories behind their inspiration for how to portray their science to captivate a broader audience.

2.
Dev Cell ; 57(13): 1661-1675.e7, 2022 07 11.
Article in English | MEDLINE | ID: mdl-35716661

ABSTRACT

Recruitment of stem cells is crucial for tissue repair. Although stem cell niches can provide important signals, little is known about mechanisms that coordinate the engagement of disseminated stem cells across an injured tissue. In Drosophila, adult brain lesions trigger local recruitment of scattered dormant neural stem cells suggesting a mechanism for creating a transient stem cell activation zone. Here, we find that injury triggers a coordinated response in neuro-glial clusters that promotes the spread of a neuron-derived stem cell factor via glial secretion of the lipocalin-like transporter Swim. Strikingly, swim is induced in a Hif1-α-dependent manner in response to brain hypoxia. Mammalian Swim (Lcn7) is also upregulated in glia of the mouse hippocampus upon brain injury. Our results identify a central role of neuro-glial clusters in promoting neural stem cell activation at a distance, suggesting a conserved function of the HIF1-α/Swim/Wnt module in connecting injury-sensing and regenerative outcomes.


Subject(s)
Drosophila , Neural Stem Cells , Animals , Mammals , Mice , Neuroglia , Neurons , Stem Cell Niche
3.
Dev Cell ; 51(6): 787-803.e5, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31735669

ABSTRACT

The use of adult Drosophila melanogaster as a model for hematopoiesis or organismal immunity has been debated. Addressing this question, we identify an extensive reservoir of blood cells (hemocytes) at the respiratory epithelia (tracheal air sacs) of the thorax and head. Lineage tracing and functional analyses demonstrate that the majority of adult hemocytes are phagocytic macrophages (plasmatocytes) from the embryonic lineage that parallels vertebrate tissue macrophages. Surprisingly, we find no sign of adult hemocyte expansion. Instead, hemocytes play a role in relaying an innate immune response to the blood cell reservoir: through Imd signaling and the Jak/Stat pathway ligand Upd3, hemocytes act as sentinels of bacterial infection, inducing expression of the antimicrobial peptide Drosocin in respiratory epithelia and colocalizing fat body domains. Drosocin expression in turn promotes animal survival after infection. Our work identifies a multi-signal relay of organismal humoral immunity, establishing adult Drosophila as model for inter-organ immunity.


Subject(s)
Blood Cells/metabolism , Hematopoiesis/physiology , Macrophages/metabolism , Respiratory Mucosa/metabolism , Animals , Drosophila/metabolism , Hemocytes/metabolism , Immunity, Cellular/immunology , Immunity, Innate/immunology , Janus Kinases/metabolism , Transcription Factors/metabolism
4.
Nature ; 572(7768): 260-264, 2019 08.
Article in English | MEDLINE | ID: mdl-31341286

ABSTRACT

In humans, the adaptive immune system uses the exchange of information between cells to detect and eliminate foreign or damaged cells; however, the removal of unwanted cells does not always require an adaptive immune system1,2. For example, cell selection in Drosophila uses a cell selection mechanism based on 'fitness fingerprints', which allow it to delay ageing3, prevent developmental malformations3,4 and replace old tissues during regeneration5. At the molecular level, these fitness fingerprints consist of combinations of Flower membrane proteins3,4,6. Proteins that indicate reduced fitness are called Flower-Lose, because they are expressed in cells marked to be eliminated6. However, the presence of Flower-Lose isoforms at a cell's membrane does not always lead to elimination, because if neighbouring cells have similar levels of Lose proteins, the cell will not be killed4,6,7. Humans could benefit from the capability to recognize unfit cells, because accumulation of damaged but viable cells during development and ageing causes organ dysfunction and disease8-17. However, in Drosophila this mechanism is hijacked by premalignant cells to gain a competitive growth advantage18. This would be undesirable for humans because it might make tumours more aggressive19-21. It is unknown whether a similar mechanism of cell-fitness comparison is present in humans. Here we show that two human Flower isoforms (hFWE1 and hFWE3) behave as Flower-Lose proteins, whereas the other two isoforms (hFWE2 and hFWE4) behave as Flower-Win proteins. The latter give cells a competitive advantage over cells expressing Lose isoforms, but Lose-expressing cells are not eliminated if their neighbours express similar levels of Lose isoforms; these proteins therefore act as fitness fingerprints. Moreover, human cancer cells show increased Win isoform expression and proliferate in the presence of Lose-expressing stroma, which confers a competitive growth advantage on the cancer cells. Inhibition of the expression of Flower proteins reduces tumour growth and metastasis, and induces sensitivity to chemotherapy. Our results show that ancient mechanisms of cell recognition and selection are active in humans and affect oncogenic growth.


Subject(s)
Calcium Channels/metabolism , Cell Proliferation , Drosophila Proteins/metabolism , Neoplasms/pathology , Protein Isoforms/metabolism , Animals , Calcium Channels/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Drosophila melanogaster , Female , Gene Knockdown Techniques , Humans , Male , Neoplasm Metastasis , Neoplasms/drug therapy , Protein Isoforms/genetics
5.
Cell Rep ; 25(13): 3661-3673.e3, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30590040

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia, impairing cognitive and motor functions. One of the pathological hallmarks of AD is neuronal loss, which is not reflected in mouse models of AD. Therefore, the role of neuronal death is still uncertain. Here, we used a Drosophila AD model expressing a secreted form of human amyloid-ß42 peptide and showed that it recapitulates key aspects of AD pathology, including neuronal death and impaired long-term memory. We found that neuronal apoptosis is mediated by cell fitness-driven neuronal culling, which selectively eliminates impaired neurons from brain circuits. We demonstrated that removal of less fit neurons delays ß-amyloid-induced brain damage and protects against cognitive and motor decline, suggesting that contrary to common knowledge, neuronal death may have a beneficial effect in AD.


Subject(s)
Amyloid beta-Peptides/toxicity , Brain/pathology , Brain/physiopathology , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Motor Activity , Neurons/pathology , Neuroprotection , Peptide Fragments/toxicity , Animals , Brain/drug effects , Cell Death , Courtship , Disease Models, Animal , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Epithelium/drug effects , Epithelium/metabolism , Epithelium/pathology , Female , Humans , Huntington Disease/pathology , Longevity , Male , Memory, Long-Term/drug effects , Motor Activity/drug effects , Mutant Proteins/metabolism , Neurons/drug effects , Neuroprotection/drug effects , Parkinson Disease/pathology , Vacuoles/metabolism
6.
Int J Dev Biol ; 62(6-7-8): 465-471, 2018.
Article in English | MEDLINE | ID: mdl-29938758

ABSTRACT

Adult stem cells in mammals are important for normal tissue renewal (homeostasis) and regeneration after injury. In the past ten years, different types of homeostatic adult stem cells have also been identified in the genetically accessible fruit fly (Drosophila melanogaster), among which intestinal stem cells have taken centre stage. Recent studies provide evidence that adult fly tissues may also harbor quiescent stem cells, which can enter cell cycle upon injury to regenerate compromised tissue. Such damage-responsive stem cells have been described in flight muscles, the adult brain and in a narrow region of the fly hindgut. Strikingly, many mammalian tissues have also been shown to maintain quiescent, but regeneration-competent, stem cells. However, little is known about the injury-induced signals that lead to their activation. Here, we provide a brief overview of active and damage-responsive adult stem cells in the fruit fly and focus on injury-dependent signalling events. We highlight the potential of Drosophila to model damage-induced stem cell activation to deepen our molecular understanding of how dormant stem cells can be efficiently recruited for tissue repair after injury.


Subject(s)
Drosophila melanogaster/physiology , Homeostasis/physiology , Intestines/physiology , Regeneration/physiology , Stem Cells/physiology , Animals , Cell Cycle/physiology , Cell Differentiation/physiology , Cell Proliferation/physiology , Drosophila melanogaster/cytology , Intestines/cytology , Signal Transduction/physiology , Stem Cells/cytology
7.
Front Neurosci ; 11: 327, 2017.
Article in English | MEDLINE | ID: mdl-28642678

ABSTRACT

During brain development, highly complex and interconnected neural circuits are established. This intricate wiring needs to be robust to faithfully perform adult brain function throughout life, but at the same time offer room for plasticity to integrate new information. In the mammalian brain, adult-born neurons are produced in restricted niches harboring neural stem cells. In the fruit fly Drosophila, low-level adult neurogenesis arising from a dispersed population of neural progenitors has recently been detected in the optic lobes. Strikingly, these normally quiescent neural stem cells proliferate upon brain injury and produce new neurons for brain regeneration. Here, we review adult neurogenesis in crustaceans and insects and highlight that neurogenesis in the visual system is prominent in arthropods, but its role and underlying mechanisms are unclear. Moreover, we discuss how the study of damage-responsive progenitor cells in Drosophila may help to understand robust regenerative neurogenesis and open new avenues to enhance brain repair after injury or stroke in humans.

8.
Neurosci Biobehav Rev ; 56: 62-72, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26118647

ABSTRACT

Neuronal circuits in the adult brain have long been viewed as static and stable. However, research in the past 20 years has shown that specialized regions of the adult brain, which harbor adult neural stem cells, continue to produce new neurons in a wide range of species. Brain plasticity is also observed after injury. Depending on the extent and permissive environment of neurogenic regions, different organisms show great variability in their capacity to replace lost neurons by endogenous neurogenesis. In Zebrafish and Drosophila, the formation of new neurons from progenitor cells in the adult brain was only discovered recently. Here, we compare properties of adult neural stem cells, their niches and regenerative responses from mammals to flies. Current models of brain injury have revealed that specific injury-induced genetic programs and comparison of neuronal fitness are implicated in brain repair. We highlight the potential of these recently implemented models of brain regeneration to identify novel regulators of stem cell activation and regenerative neurogenesis.


Subject(s)
Brain Injuries/physiopathology , Models, Genetic , Neurogenesis/genetics , Neurons/physiology , Regeneration/genetics , Animals , Humans
9.
Curr Biol ; 25(7): 955-63, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25754635

ABSTRACT

Darwinian-like cell selection has been studied during development and cancer [1-11]. Cell selection is often mediated by direct intercellular comparison of cell fitness, using "fitness fingerprints" [12-14]. In Drosophila, cells compare their fitness via several isoforms of the transmembrane protein Flower [12, 13]. Here, we studied the role of intercellular fitness comparisons during regeneration. Regeneration-competent organisms are traditionally injured by amputation [15, 16], whereas in clinically relevant injuries such as local ischemia or traumatic injury, damaged tissue remains within the organ [17-19]. We reasoned that "Darwinian" interactions between old and newly formed tissues may be important in the elimination of damaged cells. We used a model of adult brain regeneration in Drosophila in which mechanical puncture activates regenerative neurogenesis based on damage-responsive stem cells [20]. We found that apoptosis after brain injury occurs in damage-exposed tissue located adjacent to zones of de novo neurogenesis. Injury-affected neurons start to express isoforms of the Flower cell fitness indicator protein not found on intact neurons. We show that this change in the neuronal fitness fingerprint is required to recognize and eliminate such neurons. Moreover, apoptosis is inhibited if all neurons express "low-fitness" markers, showing that the availability of new and healthy cells drives tissue replacement. In summary, we found that elimination of impaired tissue during brain regeneration requires comparison of neuronal fitness and that tissue replacement after brain damage is coordinated by injury-modulated fitness fingerprints. Intercellular fitness comparisons between old and newly formed tissues could be a general mechanism of regenerative tissue replacement.


Subject(s)
Brain Injuries/physiopathology , Brain/physiopathology , Genetic Fitness , Neurons/metabolism , Regeneration , Animals , Apoptosis , Brain Injuries/pathology , Drosophila , Neurons/cytology , Neurons/pathology
10.
Cell ; 160(3): 461-76, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25601460

ABSTRACT

Viable yet damaged cells can accumulate during development and aging. Although eliminating those cells may benefit organ function, identification of this less fit cell population remains challenging. Previously, we identified a molecular mechanism, based on "fitness fingerprints" displayed on cell membranes, which allows direct fitness comparison among cells in Drosophila. Here, we study the physiological consequences of efficient cell selection for the whole organism. We find that fitness-based cell culling is naturally used to maintain tissue health, delay aging, and extend lifespan in Drosophila. We identify a gene, azot, which ensures the elimination of less fit cells. Lack of azot increases morphological malformations and susceptibility to random mutations and accelerates tissue degeneration. On the contrary, improving the efficiency of cell selection is beneficial for tissue health and extends lifespan.


Subject(s)
Calcium-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Aging , Amino Acid Sequence , Animals , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Longevity , Molecular Sequence Data , Neurons/cytology , Neurons/metabolism , Promoter Regions, Genetic , Sequence Alignment
11.
Curr Opin Cell Biol ; 31: 16-22, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25022356

ABSTRACT

Metazoans have evolved ways to engage only the most appropriate cells for long-term tissue development and homeostasis. In many cases, competitive interactions have been shown to guide such cell selection events. In Drosophila, a process termed cell competition eliminates slow proliferating cells from growing epithelia. Recent studies show that cell competition is conserved in mammals with crucial functions like the elimination of suboptimal stem cells from the early embryo and the replacement of old T-cell progenitors in the thymus to prevent tumor formation. Moreover, new data in Drosophila has revealed that fitness indicator proteins, required for cell competition, are also involved in the culling of retinal neurons suggesting that 'fitness fingerprints' may play a general role in cell selection.


Subject(s)
Cell Physiological Phenomena , Aging/pathology , Aging/physiology , Animals , Cell Proliferation , Drosophila/cytology , Humans , Mammals/physiology , Neoplasms/pathology , Stem Cells/pathology
12.
PLoS One ; 8(9): e74908, 2013.
Article in English | MEDLINE | ID: mdl-24066155

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) are proteins with long covalently attached sugar side chains of the heparan sulfate (HS) type. Depending on the cellular context HS chains carry multiple structural modifications such as sulfate residues or epimerized sugars allowing them to bind to a wide range of molecules. HSPGs have been found to play extremely diverse roles in animal development and were shown to interact with certain axon guidance molecules. In this study we describe the role of the Caenorhabditis elegans HSPG core proteins Syndecan (SDN-1) and Glypican (LON-2) and the HS modifying enzymes in the dorsal guidance of D-type motor axons, a process controlled mainly by the conserved axon guidance molecule UNC-6/Netrin. Our genetic analysis established the specific HS code relevant for this axon guidance event. Using two sensitized genetic backgrounds, we isolated novel components influencing D-type motor axon guidance with a link to HSPGs, as well as new alleles of several previously characterized axon guidance genes. Interestingly, the dorsal axon guidance defects induced by mutations in zfp-1 or lin-35 depended on the transgene oxIs12 used to visualize the D-type motor neurons. oxIs12 is a large multi-copy transgene that enlarges the X chromosome by approximately 20%. In a search for genes with a comparable phenotype we found that a mutation in the known dosage compensation gene dpy-21 showed similar axon guidance defects as zfp-1 or lin-35 mutants. Thus, derepression of genes on X, where many genes relevant for HS dependent axon guidance are located, might also influence axon guidance of D-type motor neurons.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Heparan Sulfate Proteoglycans/metabolism , Heparitin Sulfate/metabolism , Motor Neurons/cytology , Nerve Tissue Proteins/metabolism , Animals , Motor Neurons/metabolism , Netrins
13.
Curr Biol ; 23(14): 1300-9, 2013 Jul 22.
Article in English | MEDLINE | ID: mdl-23810538

ABSTRACT

BACKGROUND: The flower gene has been previously linked to the elimination of slow dividing epithelial cells during development in a process known as "cell competition." During cell competition, different isoforms of the Flower protein are displayed at the cell membrane and reveal the reduced fitness of slow proliferating cells, which are therefore recognized, eliminated, and replaced by their normally dividing neighbors. This mechanism acts as a "cell quality" control in proliferating tissues. RESULTS: Here, we use the Drosophila eye as a model to study how unwanted neurons are culled during retina development and find that flower is required and sufficient for the recognition and elimination of supernumerary postmitotic neurons, contained within incomplete ommatidia units. This constitutes the first description of the "Flower Code" functioning as a cell selection mechanism in postmitotic cells and is also the first report of a physiological role for this cell quality control machinery. CONCLUSIONS: Our results show that the "Flower Code" is a general system to reveal cell fitness and that it may play similar roles in creating optimal neural networks in higher organisms. The Flower Code seems to be a more general mechanism for cell monitoring and selection than previously recognized.


Subject(s)
Calcium Channels/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Gene Expression Regulation, Developmental , Neurons/physiology , Retina/growth & development , Animals , Calcium Channels/metabolism , Cell Death , Cell Proliferation , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , In Situ Nick-End Labeling , Polymerase Chain Reaction , RNA Interference
14.
Cell Rep ; 3(6): 1857-65, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23791523

ABSTRACT

Adult neurogenesis has been linked to several cognitive functions and neurological disorders. Description of adult neurogenesis in a model organism like Drosophila could facilitate the genetic study of normal and abnormal neurogenesis in the adult brain. So far, formation of new neurons has not been detected in adult fly brains and hence has been thought to be absent in Drosophila. Here, we used an improved lineage-labeling method to show that, surprisingly, adult neurogenesis occurs in the medulla cortex of the Drosophila optic lobes. We also find that acute brain damage to this region stimulates adult neurogenesis. Finally, we identify a factor induced by acute damage, which is sufficient to specifically activate the proliferation of a cell type with adult neuroblast characteristics. Our results reveal unexpected plasticity in the adult Drosophila brain and describe a unique model for the genetic analysis of adult neurogenesis, plasticity, and brain regeneration.


Subject(s)
Drosophila/growth & development , Animals , Brain/cytology , Brain/growth & development , Drosophila/metabolism , Neural Stem Cells/cytology , Neurogenesis
15.
Dis Model Mech ; 5(4): 553-61, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22362363

ABSTRACT

Skin papillomas arise as a result of clonal expansion of mutant cells. It has been proposed that the expansion of pretumoral cell clones is propelled not only by the increased proliferation capacity of mutant cells, but also by active cell selection. Previous studies in Drosophila describe a clonal selection process mediated by the Flower (Fwe) protein, whereby cells that express certain Fwe isoforms are recognized and forced to undergo apoptosis. It was further shown that knock down of fwe expression in Drosophila can prevent the clonal expansion of dMyc-overexpressing pretumoral cells. Here, we study the function of the single predicted mouse homolog of Drosophila Fwe, referred to as mFwe, by clonal overexpression of mFwe isoforms in Drosophila and by analyzing mFwe knock-out mice. We show that clonal overexpression of certain mFwe isoforms in Drosophila also triggers non-autonomous cell death, suggesting that Fwe function is evolutionarily conserved. Although mFwe-deficient mice display a normal phenotype, they develop a significantly lower number of skin papillomas upon exposure to DMBA/TPA two-stage skin carcinogenesis than do treated wild-type and mFwe heterozygous mice. Furthermore, mFwe expression is higher in papillomas and the papilloma-surrounding skin of treated wild-type mice compared with the skin of untreated wild-type mice. Thus, we propose that skin papilloma cells take advantage of mFwe activity to facilitate their clonal expansion.


Subject(s)
Calcium Channels/deficiency , Cell Transformation, Neoplastic/pathology , Disease Susceptibility/pathology , Papilloma/pathology , Skin Neoplasms/pathology , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genetic Loci , Imaginal Discs/metabolism , Mice , Mice, Knockout , Papilloma/genetics , Phenotype , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Nucleic Acid , Skin/metabolism , Skin/pathology , Skin Neoplasms/genetics , Wings, Animal/metabolism
16.
Dev Cell ; 19(4): 562-73, 2010 Oct 19.
Article in English | MEDLINE | ID: mdl-20951347

ABSTRACT

During development and aging, animals suffer insults that modify the fitness of individual cells. In Drosophila, the elimination of viable but suboptimal cells is mediated by cell competition, ensuring that these cells do not accumulate during development. In addition, certain genes such as the Drosophila homolog of human c-myc (dmyc) are able to transform cells into supercompetitors, which eliminate neighboring wild-type cells by apoptosis and overproliferate, leaving total cell numbers unchanged. Here we have identified Drosophila Sparc as an early marker transcriptionally upregulated in loser cells that provides a transient protection by inhibiting Caspase activation in outcompeted cells. Overall, we describe the unexpected existence of a physiological mechanism that counteracts cell competition during development.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Osteonectin/metabolism , Signal Transduction , Animals , Apoptosis , Biomarkers/metabolism , Cell Line , Cytoprotection , Drosophila Proteins/genetics , Epistasis, Genetic , Osteonectin/genetics , Up-Regulation/genetics
17.
Dev Cell ; 18(6): 985-98, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20627080

ABSTRACT

Cell competition promotes the elimination of weaker cells from a growing population. Here we investigate how cells of Drosophila wing imaginal discs distinguish "winners" from "losers" during cell competition. Using genomic and functional assays, we have identified several factors implicated in the process, including Flower (Fwe), a cell membrane protein conserved in multicellular animals. Our results suggest that Fwe is a component of the cell competition response that is required and sufficient to label cells as "winners" or "losers." In Drosophila, the fwe locus produces three isoforms, fwe(ubi), fwe(Lose-A), and fwe(Lose-B). Basal levels of fwe(ubi) are constantly produced. During competition, the fwe(Lose) isoforms are upregulated in prospective loser cells. Cell-cell comparison of relative fwe(Lose) and fwe(ubi) levels ultimately determines which cell undergoes apoptosis. This "extracellular code" may constitute an ancient mechanism to terminate competitive conflicts among cells.


Subject(s)
Apoptosis/physiology , Calcium Channels/metabolism , Cell Differentiation/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Extracellular Space/metabolism , Wings, Animal/embryology , Animals , Calcium Channels/genetics , Cell Communication/physiology , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Survival/physiology , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Extracellular Space/genetics , Gene Expression Regulation, Developmental/physiology , Organogenesis/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Up-Regulation/physiology , Wings, Animal/cytology
18.
Development ; 136(7): 1137-45, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19270172

ABSTRACT

The proper development of tissues requires morphogen activity that dictates the appropriate growth and differentiation of each cell according to its position within a developing field. Elimination of underperforming cells that are less efficient in receiving/transducing the morphogenetic signal is thought to provide a general fail-safe mechanism to avoid developmental misspecification. In the developing Drosophila wing, the morphogen Dpp provides cells with growth and survival cues. Much of the regulation of transcriptional output by Dpp is mediated through repression of the transcriptional repressor Brinker (Brk), and thus through the activation of target genes. Mutant cells impaired for Dpp reception or transduction are lost from the wing epithelium. At the molecular level, reduced Dpp signaling results in Brk upregulation that triggers apoptosis through activation of the JNK pathway. Here we show that the transcriptional co-regulator dNAB is a Dpp target in the developing wing that interacts with Brk to eliminate cells with reduced Dpp signaling through the JNK pathway. We further show that both dNAB and Brk are required for cell elimination induced by differential dMyc expression, a process that depends on reduced Dpp transduction in outcompeted cells. We propose a novel mechanism whereby the morphogen Dpp regulates the responsiveness to its own survival signal by inversely controlling the expression of a repressor, Brk, and its co-repressor, dNAB.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/growth & development , Drosophila/metabolism , Repressor Proteins/metabolism , Animals , Animals, Genetically Modified , Apoptosis/genetics , Apoptosis/physiology , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning/genetics , Body Patterning/physiology , DNA Primers/genetics , Drosophila/genetics , Drosophila Proteins/genetics , Epistasis, Genetic , Genes, Insect , In Vitro Techniques , JNK Mitogen-Activated Protein Kinases/metabolism , Repressor Proteins/genetics , Signal Transduction , Wings, Animal/growth & development , Wings, Animal/metabolism
19.
Development ; 136(6): 995-1006, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19211674

ABSTRACT

Cell competition is a short-range cell-cell interaction leading to the proliferation of winner cells at the expense of losers, although either cell type shows normal growth in homotypic environments. Drosophila Myc (dMyc; Dm-FlyBase) is a potent inducer of cell competition in wing epithelia, but its role in the ovary germline stem cell niche is unknown. Here, we show that germline stem cells (GSCs) with relative lower levels of dMyc are replaced by GSCs with higher levels of dMyc. By contrast, dMyc-overexpressing GSCs outcompete wild-type stem cells without affecting total stem cell numbers. We also provide evidence for a naturally occurring cell competition border formed by high dMyc-expressing stem cells and low dMyc-expressing progeny, which may facilitate the concentration of the niche-provided self-renewal factor BMP/Dpp in metabolically active high dMyc stem cells. Genetic manipulations that impose uniform dMyc levels across the germline produce an extended Dpp signaling domain and cause uncoordinated differentiation events. We propose that dMyc-induced competition plays a dual role in regulating optimal stem cell pools and sharp differentiation boundaries, but is potentially harmful in the case of emerging dmyc duplications that facilitate niche occupancy by pre-cancerous stem cells. Moreover, competitive interactions among stem cells may be relevant for the successful application of stem cell therapies in humans.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Ovary/cytology , Ovary/metabolism , Stem Cell Niche/cytology , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Differentiation , Cell Proliferation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Female , Gene Expression Regulation, Developmental , Mutation/genetics , Stem Cell Niche/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Carcinogenesis ; 30(5): 723-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19126656

ABSTRACT

Cancer is the result of sequential genetic changes over time that transform a cell into a malignant and ultimately invasive entity. The insight that cancerous cells arise from a series of mutations in oncogenes and tumor suppressors, commonly known as multistep carcinogenesis, has been conceptually elaborated and proven in the last 20 years. Although knowledge about late steps of cancerogenesis and disease progression has greatly advanced, the initial molecular events remain largely unknown. Basic research in Drosophila has started the quest to find early markers that detect initial clonal expansion of precancerous cells. These efforts were spurred by novel findings demonstrating that certain mutations transform cells into super-competitors that expand at the expense of the surrounding epithelial cells without inducing histological changes. This mechanism, discovered as super competition in the fly, might also lie at the heart of a clinical observation termed 'field cancerization'. This review aims to bring together current understanding from basic research on cell competition and clinical studies that have analyzed field characteristics to highlight parallels and possible connections.


Subject(s)
Cell Transformation, Neoplastic/genetics , Precancerous Conditions/genetics , Animals , Apoptosis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Division , Cyclin D1/genetics , Drosophila/genetics , Genes, Tumor Suppressor , Genetic Markers , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Loss of Heterozygosity , Models, Biological , Neoplasms/genetics , Neoplasms/pathology , Oncogenes , Precancerous Conditions/diagnosis , Precancerous Conditions/pathology
SELECTION OF CITATIONS
SEARCH DETAIL