Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Biol ; 34(7): 1426-1437.e6, 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38484734

RESUMO

7An efficient immune system must provide protection against a broad range of pathogens without causing excessive collateral tissue damage. While immune effectors have been well characterized, we know less about the resilience mechanisms protecting the host from its own immune response. Antimicrobial peptides (AMPs) are small, cationic peptides that contribute to innate defenses by targeting negatively charged membranes of microbes. While protective against pathogens, AMPs can be cytotoxic to host cells. Here, we reveal that a family of stress-induced proteins, the Turandots, protect the Drosophila respiratory system from AMPs, increasing resilience to stress. Flies lacking Turandot genes are susceptible to environmental stresses due to AMP-induced tracheal apoptosis. Turandot proteins bind to host cell membranes and mask negatively charged phospholipids, protecting them from cationic pore-forming AMPs. Collectively, these data demonstrate that Turandot stress proteins mitigate AMP cytotoxicity to host tissues and therefore improve their efficacy.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/metabolismo , Peptídeos Antimicrobianos , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/metabolismo , Peptídeos Catiônicos Antimicrobianos/farmacologia , Imunidade Inata/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(30): e2208461119, 2022 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-35858432

RESUMO

Insects frequently harbor endosymbionts, which are bacteria housed within host tissues. These associations are stably maintained over evolutionary timescales through vertical transmission of endosymbionts from host mothers to their offspring. Some endosymbionts manipulate host reproduction to facilitate spread within natural populations. Consequently, such infections have major impacts on insect physiology and evolution. However, technical hurdles have limited our understanding of the molecular mechanisms underlying such insect-endosymbiont interactions. Here, we investigate the nutritional interactions between endosymbiotic partners using the tractable insect Drosophila melanogaster and its natural endosymbiont Spiroplasma poulsonii. Using a combination of functional assays, metabolomics, and proteomics, we show that the abundance and amino acid composition of a single Spiroplasma membrane lectin, Spiralin B (SpiB), dictates the amino acid requirements of the endosymbiont and determines its proliferation within host tissues. Ectopically increasing SpiB levels in host tissues disrupts localization of endosymbionts in the fly egg chambers and decreases vertical transmission. We find that SpiB is likely to be required by the endosymbiont to enter host oocytes, which may explain the massive investment of S. poulsonii in SpiB synthesis. SpiB both permits vertical transmission of the symbiont and limits its growth in nutrient-limiting conditions for the host; therefore, a single protein plays a pivotal role in ensuring durability of the interaction in a variable environment.


Assuntos
Proteínas da Membrana Bacteriana Externa , Drosophila melanogaster , Interações entre Hospedeiro e Microrganismos , Spiroplasma , Simbiose , Aminoácidos/metabolismo , Animais , Proteínas da Membrana Bacteriana Externa/metabolismo , Drosophila melanogaster/microbiologia , Drosophila melanogaster/fisiologia , Spiroplasma/metabolismo
3.
EMBO J ; 40(4): e104347, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33372708

RESUMO

Adult stem cells must continuously fine-tune their behavior to regenerate damaged organs and avoid tumors. While several signaling pathways are well known to regulate somatic stem cells, the underlying mechanisms remain largely unexplored. Here, we demonstrate a cell-intrinsic role for the OvoL family transcription factor, Shavenbaby (Svb), in balancing self-renewal and differentiation of Drosophila intestinal stem cells. We find that svb is a downstream target of Wnt and EGFR pathways, mediating their activity for stem cell survival and proliferation. This requires post-translational processing of Svb into a transcriptional activator, whose upregulation induces tumor-like stem cell hyperproliferation. In contrast, the unprocessed form of Svb acts as a repressor that imposes differentiation into enterocytes, and suppresses tumors induced by altered signaling. We show that the switch between Svb repressor and activator is triggered in response to systemic steroid hormone, which is produced by ovaries. Therefore, the Svb axis allows intrinsic integration of local signaling cues and inter-organ communication to adjust stem cell proliferation versus differentiation, suggesting a broad role of OvoL/Svb in adult and cancer stem cells.


Assuntos
Diferenciação Celular , Autorrenovação Celular , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Intestinos/fisiologia , Células-Tronco/citologia , Esteroides/farmacologia , Fatores de Transcrição/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Drosophila , Proteínas de Drosophila/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Células-Tronco/metabolismo , Fatores de Transcrição/genética
4.
Proc Natl Acad Sci U S A ; 117(13): 7317-7325, 2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32188787

RESUMO

Iron sequestration is a recognized innate immune mechanism against invading pathogens mediated by iron-binding proteins called transferrins. Despite many studies on antimicrobial activity of transferrins in vitro, their specific in vivo functions are poorly understood. Here we use Drosophila melanogaster as an in vivo model to investigate the role of transferrins in host defense. We find that systemic infections with a variety of pathogens trigger a hypoferremic response in flies, namely, iron withdrawal from the hemolymph and accumulation in the fat body. Notably, this hypoferremia to infection requires Drosophila nuclear factor κB (NF-κB) immune pathways, Toll and Imd, revealing that these pathways also mediate nutritional immunity in flies. Next, we show that the iron transporter Tsf1 is induced by infections downstream of the Toll and Imd pathways and is necessary for iron relocation from the hemolymph to the fat body. Consistent with elevated iron levels in the hemolymph, Tsf1 mutants exhibited increased susceptibility to Pseudomonas bacteria and Mucorales fungi, which could be rescued by chemical chelation of iron. Furthermore, using siderophore-deficient Pseudomonas aeruginosa, we discover that the siderophore pyoverdine is necessary for pathogenesis in wild-type flies, but it becomes dispensable in Tsf1 mutants due to excessive iron present in the hemolymph of these flies. As such, our study reveals that, similar to mammals, Drosophila uses iron limitation as an immune defense mechanism mediated by conserved iron-transporting proteins transferrins. Our in vivo work, together with accumulating in vitro studies, supports the immune role of insect transferrins against infections via an iron withholding strategy.


Assuntos
Proteínas de Drosophila/metabolismo , Ferro/metabolismo , Transferrina/metabolismo , Animais , Proteínas de Drosophila/imunologia , Drosophila melanogaster , Hemolinfa/imunologia , Hemolinfa/metabolismo , Imunidade Inata , Ferro/imunologia , NF-kappa B/metabolismo , Pseudomonas aeruginosa/metabolismo , Sideróforos/metabolismo , Transferrina/imunologia
5.
FEBS J ; 287(16): 3399-3426, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32009293

RESUMO

In animals, growth is regulated by the complex interplay between paracrine and endocrine signals. When food is scarce, tissues compete for nutrients, leading to critical resource allocation and prioritization. Little is known about how the immune system maturation is coordinated with the growth of other tissues. Here, we describe a signaling mechanism that regulates the number of hemocytes (blood cells) according to the nutritional state of the Drosophila larva. Specifically, we found that a secreted protein, NimB5, is produced in the fat body upon nutrient scarcity downstream of metabolic sensors and ecdysone signaling. NimB5 is then secreted and binds to hemocytes to down-regulate their proliferation and adhesion. Blocking this signaling loop results in conditional lethality when larvae are raised on a poor diet, due to excessive hemocyte numbers and insufficient energy storage. Similar regulatory mechanisms shaping the immune system in response to nutrient availability are likely to be widespread in animals.


Assuntos
Adipocinas/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Hematopoese/genética , Adipocinas/metabolismo , Animais , Animais Geneticamente Modificados , Adesão Celular/genética , Proliferação de Células/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Corpo Adiposo/metabolismo , Hemócitos/citologia , Hemócitos/metabolismo , Larva/citologia , Larva/genética , Larva/metabolismo , Mutação , Fagocitose/genética , Transdução de Sinais/genética
6.
Cell Rep ; 27(3): 886-899.e6, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30995484

RESUMO

In ectotherms, increased ambient temperature requires the organism to consume substantial amounts of energy to sustain a higher metabolic rate, prevent cellular damage, and respond to heat stress. Here, we identify a heat-inducible apolipoprotein required for thermal acclimation in Drosophila. Neuropeptide-like precursor 2 (Nplp2) is an abundant hemolymphatic protein thought to be a neuropeptide. In contrast, we show that Nplp2 contributes to lipid transport, functioning as an exchangeable apolipoprotein. More precisely, Nplp2-deficient flies accumulate lipids in their gut, have reduced fat stores, and display a dyslipoproteinemia, showing that Nplp2 is required for dietary lipid assimilation. Importantly, Nplp2 is induced upon thermal stress and contributes to survival upon heat stress. We propose that Nplp2 associates with lipoprotein particles under homeostatic and high energy-demand conditions to optimize fat transport and storage. Our study also shows that modulation of the lipid uptake and transport machinery is part of an integrated cytoprotective response.


Assuntos
Apolipoproteínas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Metabolismo dos Lipídeos/fisiologia , Neuropeptídeos/metabolismo , Aclimatação , Sequência de Aminoácidos , Animais , Apolipoproteínas/química , Apolipoproteínas/genética , Drosophila/crescimento & desenvolvimento , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Corpo Adiposo/metabolismo , Corpo Adiposo/patologia , Resposta ao Choque Térmico , Mucosa Intestinal/metabolismo , Larva/metabolismo , Lipoproteínas/metabolismo , Mutagênese , Neuropeptídeos/química , Neuropeptídeos/genética , Ligação Proteica , Alinhamento de Sequência , Temperatura
7.
Immunity ; 49(5): 929-942.e5, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30446385

RESUMO

Commensal microbes colonize the gut epithelia of virtually all animals and provide several benefits to their hosts. Changes in commensal populations can lead to dysbiosis, which is associated with numerous pathologies and decreased lifespan. Peptidoglycan recognition proteins (PGRPs) are important regulators of the commensal microbiota and intestinal homeostasis. Here, we found that a null mutation in Drosophila PGRP-SD was associated with overgrowth of Lactobacillus plantarum in the fly gut and a shortened lifespan. L. plantarum-derived lactic acid triggered the activation of the intestinal NADPH oxidase Nox and the generation of reactive oxygen species (ROS). In turn, ROS production promoted intestinal damage, increased proliferation of intestinal stem cells, and dysplasia. Nox-mediated ROS production required lactate oxidation by the host intestinal lactate dehydrogenase, revealing a host-commensal metabolic crosstalk that is probably broadly conserved. Our findings outline a mechanism whereby host immune dysfunction leads to commensal dysbiosis that in turn promotes age-related pathologies.


Assuntos
Drosophila/fisiologia , Ácido Láctico/metabolismo , Longevidade , Microbiota , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Disbiose , Expressão Gênica , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , L-Lactato Desidrogenase/genética , L-Lactato Desidrogenase/metabolismo , Mutação , NADPH Oxidases/genética , Transdução de Sinais , Simbiose
8.
Immunity ; 48(5): 897-910.e7, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29752064

RESUMO

Intestinal infection triggers potent immune responses to combat pathogens and concomitantly drives epithelial renewal to maintain barrier integrity. Current models propose that epithelial renewal is primarily driven by damage caused by reactive oxygen species (ROS). Here we found that in Drosophila, the Imd-NF-κB pathway controlled enterocyte (EC) shedding upon infection, via a mechanism independent of ROS-associated apoptosis. Mechanistically, the Imd pathway synergized with JNK signaling to induce epithelial cell shedding specifically in the context of bacterial infection, requiring also the reduced expression of the transcription factor GATAe. Furthermore, cell-specific NF-κB responses enabled simultaneous production of antimicrobial peptides (AMPs) and epithelial shedding in different EC populations. Thus, the Imd-NF-κB pathway is central to the intestinal antibacterial response by mediating both AMP production and the maintenance of barrier integrity. Considering the similarities between Drosophila Imd signaling and mammalian TNFR pathway, our findings suggest the existence of an evolutionarily conserved genetic program in immunity-induced epithelial shedding.


Assuntos
Peptídeos Catiônicos Antimicrobianos/imunologia , Bactérias/imunologia , Infecções Bacterianas/imunologia , Proteínas de Drosophila/imunologia , Células Epiteliais/imunologia , NF-kappa B/imunologia , Animais , Animais Geneticamente Modificados , Peptídeos Catiônicos Antimicrobianos/metabolismo , Bactérias/crescimento & desenvolvimento , Infecções Bacterianas/metabolismo , Infecções Bacterianas/microbiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/imunologia , Drosophila melanogaster/metabolismo , Drosophila melanogaster/microbiologia , Enterócitos/imunologia , Enterócitos/metabolismo , Enterócitos/microbiologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Fatores de Transcrição GATA/genética , Fatores de Transcrição GATA/imunologia , Fatores de Transcrição GATA/metabolismo , Regulação da Expressão Gênica/imunologia , Mucosa Intestinal/citologia , NF-kappa B/metabolismo , Transdução de Sinais/imunologia
9.
PLoS Genet ; 13(6): e1006854, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28662029

RESUMO

The speed of stem cell differentiation has to be properly coupled with self-renewal, both under basal conditions for tissue maintenance and during regeneration for tissue repair. Using the Drosophila midgut model, we analyze at the cellular and molecular levels the differentiation program required for robust regeneration. We observe that the intestinal stem cell (ISC) and its differentiating daughter, the enteroblast (EB), form extended cell-cell contacts in regenerating intestines. The contact between progenitors is stabilized by cell adhesion molecules, and can be dynamically remodeled to elicit optimal juxtacrine Notch signaling to determine the speed of progenitor differentiation. Notably, increasing the adhesion property of progenitors by expressing Connectin is sufficient to induce rapid progenitor differentiation. We further demonstrate that JAK/STAT signaling, Sox21a and GATAe form a functional relay to orchestrate EB differentiation. Thus, our study provides new insights into the complex and sequential events that are required for rapid differentiation following stem cell division during tissue replenishment.


Assuntos
Diferenciação Celular/genética , Proteínas de Drosophila/genética , Fatores de Transcrição GATA/genética , Intestinos/crescimento & desenvolvimento , Regeneração/genética , Fatores de Transcrição SOXB2/genética , Animais , Adesão Celular/genética , Comunicação Celular/genética , Proliferação de Células/genética , Autorrenovação Celular , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Intestinos/citologia , Janus Quinases/genética , Fatores de Transcrição STAT/genética , Transdução de Sinais , Células-Tronco/citologia
10.
Nat Commun ; 6: 10219, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26690827

RESUMO

Stem cell self-renewal and differentiation are coordinated to maintain tissue homeostasis and prevent cancer. Mutations causing stem cell proliferation are traditionally the focus of cancer studies. However, the contribution of the differentiating stem cell progenies in tumorigenesis is poorly characterized. Here we report that loss of the SOX transcription factor, Sox21a, blocks the differentiation programme of enteroblast (EB), the intestinal stem cell progeny in the adult Drosophila midgut. This results in EB accumulation and formation of tumours. Sox21a tumour initiation and growth involve stem cell proliferation induced by the unpaired 2 mitogen released from accumulating EBs generating a feed-forward loop. EBs found in the tumours are heterogeneous and grow towards the intestinal lumen. Sox21a tumours modulate their environment by secreting matrix metalloproteinase and reactive oxygen species. Enterocytes surrounding the tumours are eliminated through delamination allowing tumour progression, a process requiring JNK activation. Our data highlight the tumorigenic properties of transit differentiating cells.


Assuntos
Carcinogênese/metabolismo , Proteínas de Drosophila/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Intestinos/citologia , Fatores de Transcrição SOXB2/metabolismo , Células-Tronco/fisiologia , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster , Janus Quinases/genética , Janus Quinases/metabolismo , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Mutação , Fatores de Transcrição SOXB2/genética , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
Cell Rep ; 3(5): 1725-38, 2013 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-23643535

RESUMO

Although the gut is a central organ of Eumetazoans and is essential for organismal health, our understanding of its morphological and molecular determinants remains rudimentary. Here, we provide a comprehensive atlas of Drosophila adult midgut. Specifically, we uncover a fine-grained regional organization consisting of 14 subregions with distinct morphological, histological, and genetic properties. We also show that Drosophila intestinal regionalization is defined after adult emergence, remains stable throughout life, and reestablishes following acute tissue damage. Additionally, we show that this midgut compartmentalization is achieved through the interplay between pan-midgut and regionalized transcription factors, in concert with spatial activities of morphogens. Interestingly, disruption of the midgut compartmentalization leads to a loss of intestinal homeostasis characterized by an increase in stem cell proliferation and aberrant immune responses. Our integrative analysis of Drosophila midgut compartmentalization provides insights into the conserved mechanisms underlying intestinal regionalization in metazoans.


Assuntos
Drosophila/anatomia & histologia , Intestinos/anatomia & histologia , Animais , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/crescimento & desenvolvimento , Interferência de RNA , Transdução de Sinais , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma , Proteínas Wnt/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...