Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 212
Filtrar
1.
BMC Biol ; 22(1): 94, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38664795

RESUMO

BACKGROUND: Spinal muscular atrophy (SMA) is a devastating neuromuscular disease caused by hypomorphic loss of function in the survival motor neuron (SMN) protein. SMA presents across a broad spectrum of disease severity. Unfortunately, genetic models of intermediate SMA have been difficult to generate in vertebrates and are thus unable to address key aspects of disease etiology. To address these issues, we developed a Drosophila model system that recapitulates the full range of SMA severity, allowing studies of pre-onset biology as well as late-stage disease processes. RESULTS: Here, we carried out transcriptomic and proteomic profiling of mild and intermediate Drosophila models of SMA to elucidate molecules and pathways that contribute to the disease. Using this approach, we elaborated a role for the SMN complex in the regulation of innate immune signaling. We find that mutation or tissue-specific depletion of SMN induces hyperactivation of the immune deficiency (IMD) and Toll pathways, leading to overexpression of antimicrobial peptides (AMPs) and ectopic formation of melanotic masses in the absence of an external challenge. Furthermore, the knockdown of downstream targets of these signaling pathways reduced melanotic mass formation caused by SMN loss. Importantly, we identify SMN as a negative regulator of a ubiquitylation complex that includes Traf6, Bendless, and Diap2 and plays a pivotal role in several signaling networks. CONCLUSIONS: In alignment with recent research on other neurodegenerative diseases, these findings suggest that hyperactivation of innate immunity contributes to SMA pathology. This work not only provides compelling evidence that hyperactive innate immune signaling is a primary effect of SMN depletion, but it also suggests that the SMN complex plays a regulatory role in this process in vivo. In summary, immune dysfunction in SMA is a consequence of reduced SMN levels and is driven by cellular and molecular mechanisms that are conserved between insects and mammals.


Assuntos
Modelos Animais de Doenças , Imunidade Inata , Atrofia Muscular Espinal , Transdução de Sinais , Animais , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/imunologia , Drosophila melanogaster/imunologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
2.
PLoS Biol ; 20(1): e3001494, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34990456

RESUMO

The infiltration of immune cells into tissues underlies the establishment of tissue-resident macrophages and responses to infections and tumors. Yet the mechanisms immune cells utilize to negotiate tissue barriers in living organisms are not well understood, and a role for cortical actin has not been examined. Here, we find that the tissue invasion of Drosophila macrophages, also known as plasmatocytes or hemocytes, utilizes enhanced cortical F-actin levels stimulated by the Drosophila member of the fos proto oncogene transcription factor family (Dfos, Kayak). RNA sequencing analysis and live imaging show that Dfos enhances F-actin levels around the entire macrophage surface by increasing mRNA levels of the membrane spanning molecular scaffold tetraspanin TM4SF, and the actin cross-linking filamin Cheerio, which are themselves required for invasion. Both the filamin and the tetraspanin enhance the cortical activity of Rho1 and the formin Diaphanous and thus the assembly of cortical actin, which is a critical function since expressing a dominant active form of Diaphanous can rescue the Dfos macrophage invasion defect. In vivo imaging shows that Dfos enhances the efficiency of the initial phases of macrophage tissue entry. Genetic evidence argues that this Dfos-induced program in macrophages counteracts the constraint produced by the tension of surrounding tissues and buffers the properties of the macrophage nucleus from affecting tissue entry. We thus identify strengthening the cortical actin cytoskeleton through Dfos as a key process allowing efficient forward movement of an immune cell into surrounding tissues.


Assuntos
Citoesqueleto de Actina/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/imunologia , Macrófagos/fisiologia , Animais , Movimento Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Genes de Insetos , Genes fos , Análise de Sequência de RNA , Tetraspaninas , Fatores de Transcrição/metabolismo
3.
Nat Rev Cancer ; 21(11): 687-700, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34389815

RESUMO

There is a large gap between the deep understanding of mechanisms driving tumour growth and the reasons why patients ultimately die of cancer. It is now appreciated that interactions between the tumour and surrounding non-tumour (sometimes referred to as host) cells play critical roles in mortality as well as tumour progression, but much remains unknown about the underlying molecular mechanisms, especially those that act beyond the tumour microenvironment. Drosophila has a track record of high-impact discoveries about cell-autonomous growth regulation, and is well suited to now probe mysteries of tumour - host interactions. Here, we review current knowledge about how fly tumours interact with microenvironmental stroma, circulating innate immune cells and distant organs to influence disease progression. We also discuss reciprocal regulation between tumours and host physiology, with a particular focus on paraneoplasias. The fly's simplicity along with the ability to study lethality directly provide an opportunity to shed new light on how cancer actually kills.


Assuntos
Modelos Animais de Doenças , Progressão da Doença , Drosophila melanogaster , Neoplasias/patologia , Microambiente Tumoral , Animais , Drosophila melanogaster/citologia , Drosophila melanogaster/imunologia , Humanos , Imunidade Inata , Neoplasias/imunologia , Neoplasias/mortalidade , Microambiente Tumoral/imunologia
4.
PLoS Pathog ; 17(8): e1009846, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34432851

RESUMO

The fruit fly Drosophila melanogaster combats microbial infection by producing a battery of effector peptides that are secreted into the haemolymph. Technical difficulties prevented the investigation of these short effector genes until the recent advent of the CRISPR/CAS era. As a consequence, many putative immune effectors remain to be formally described, and exactly how each of these effectors contribute to survival is not well characterized. Here we describe a novel Drosophila antifungal peptide gene that we name Baramicin A. We show that BaraA encodes a precursor protein cleaved into multiple peptides via furin cleavage sites. BaraA is strongly immune-induced in the fat body downstream of the Toll pathway, but also exhibits expression in other tissues. Importantly, we show that flies lacking BaraA are viable but susceptible to the entomopathogenic fungus Beauveria bassiana. Consistent with BaraA being directly antimicrobial, overexpression of BaraA promotes resistance to fungi and the IM10-like peptides produced by BaraA synergistically inhibit growth of fungi in vitro when combined with a membrane-disrupting antifungal. Surprisingly, BaraA mutant males but not females display an erect wing phenotype upon infection. Here, we characterize a new antifungal immune effector downstream of Toll signalling, and show it is a key contributor to the Drosophila antimicrobial response.


Assuntos
Antifúngicos/farmacologia , Beauveria/efeitos dos fármacos , Proteínas de Drosophila/farmacologia , Drosophila melanogaster/efeitos dos fármacos , Micoses/tratamento farmacológico , Peptídeos/farmacologia , Animais , Beauveria/crescimento & desenvolvimento , Beauveria/imunologia , Drosophila melanogaster/genética , Drosophila melanogaster/imunologia , Drosophila melanogaster/microbiologia , Feminino , Masculino , Micoses/imunologia , Micoses/microbiologia
5.
Dev Cell ; 56(13): 1884-1899.e5, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34197724

RESUMO

Tissue homeostasis is achieved by balancing stem cell maintenance, cell proliferation and differentiation, as well as the purging of damaged cells. Elimination of unfit cells maintains tissue health; however, the underlying mechanisms driving competitive growth when homeostasis fails, for example, during tumorigenesis, remain largely unresolved. Here, using a Drosophila intestinal model, we find that tumor cells outcompete nearby enterocytes (ECs) by influencing cell adhesion and contractility. This process relies on activating the immune-responsive Relish/NF-κB pathway to induce EC delamination and requires a JNK-dependent transcriptional upregulation of the peptidoglycan recognition protein PGRP-LA. Consequently, in organisms with impaired PGRP-LA function, tumor growth is delayed and lifespan extended. Our study identifies a non-cell-autonomous role for a JNK/PGRP-LA/Relish signaling axis in mediating death of neighboring normal cells to facilitate tumor growth. We propose that intestinal tumors "hijack" innate immune signaling to eliminate enterocytes in order to support their own growth.


Assuntos
Proteínas de Transporte/genética , Proteínas de Drosophila/genética , Imunidade Inata/genética , MAP Quinase Quinase 4/genética , Neoplasias/genética , Fatores de Transcrição/genética , Animais , Adesão Celular/genética , Proliferação de Células/genética , Modelos Animais de Doenças , Drosophila melanogaster/genética , Drosophila melanogaster/imunologia , Enterócitos/metabolismo , Enterócitos/patologia , Humanos , Intestinos/crescimento & desenvolvimento , Intestinos/patologia , Mecanotransdução Celular/genética , NF-kappa B/genética , Neoplasias/patologia , Transdução de Sinais/genética , Microambiente Tumoral/genética
6.
mBio ; 12(4): e0082421, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34253067

RESUMO

The gut microbiota affects the physiology and metabolism of animals and its alteration can lead to diseases such as gut dysplasia or metabolic disorders. Several reports have shown that the immune system plays an important role in shaping both bacterial community composition and abundance in Drosophila, and that immune deficit, especially during aging, negatively affects microbiota richness and diversity. However, there has been little study at the effector level to demonstrate how immune pathways regulate the microbiota. A key set of Drosophila immune effectors are the antimicrobial peptides (AMPs), which confer defense upon systemic infection. AMPs and lysozymes, a group of digestive enzymes with antimicrobial properties, are expressed in the gut and are good candidates for microbiota regulation. Here, we take advantage of the model organism Drosophila melanogaster to investigate the role of AMPs and lysozymes in regulation of gut microbiota structure and diversity. Using flies lacking AMPs and newly generated lysozyme mutants, we colonized gnotobiotic flies with a defined set of commensal bacteria and analyzed changes in microbiota composition and abundance in vertical transmission and aging contexts through 16S rRNA gene amplicon sequencing. Our study shows that AMPs and, to a lesser extent, lysozymes are necessary to regulate the total and relative abundance of bacteria in the gut microbiota. We also decouple the direct function of AMPs from the immune deficiency (IMD) signaling pathway that regulates AMPs but also many other processes, more narrowly defining the role of these effectors in the microbial dysbiosis observed in IMD-deficient flies upon aging. IMPORTANCE This study advances current knowledge in the field of host-microbe interactions by demonstrating that the two families of immune effectors, antimicrobial peptides and lysozymes, actively regulate the gut microbiota composition and abundance. Consequences of the loss of these antimicrobial peptides and lysozymes are exacerbated during aging, and their loss contributes to increased microbiota abundance and shifted composition in old flies. This work shows that immune effectors, typically associated with resistance to pathogenic infections, also help shape the beneficial gut community, consistent with the idea that host-symbiont interactions use the same "language" typically associated with pathogenesis.


Assuntos
Peptídeos Antimicrobianos/metabolismo , Drosophila melanogaster/química , Drosophila melanogaster/microbiologia , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiologia , Muramidase/metabolismo , Animais , Peptídeos Antimicrobianos/genética , Peptídeos Antimicrobianos/imunologia , Bactérias/classificação , Bactérias/genética , Drosophila melanogaster/genética , Drosophila melanogaster/imunologia , Feminino , Microbioma Gastrointestinal/imunologia , Interações entre Hospedeiro e Microrganismos , Sistema Imunitário , Muramidase/genética , Muramidase/imunologia , RNA Ribossômico 16S/genética , Simbiose
7.
Nature ; 597(7874): 109-113, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34261127

RESUMO

Cyclic GMP-AMP synthase (cGAS) is a cytosolic DNA sensor that produces the second messenger cG[2'-5']pA[3'-5']p (2'3'-cGAMP) and controls activation of innate immunity in mammalian cells1-5. Animal genomes typically encode multiple proteins with predicted homology to cGAS6-10, but the function of these uncharacterized enzymes is unknown. Here we show that cGAS-like receptors (cGLRs) are innate immune sensors that are capable of recognizing divergent molecular patterns and catalysing synthesis of distinct nucleotide second messenger signals. Crystal structures of human and insect cGLRs reveal a nucleotidyltransferase signalling core shared with cGAS and a diversified primary ligand-binding surface modified with notable insertions and deletions. We demonstrate that surface remodelling of cGLRs enables altered ligand specificity and used a forward biochemical screen to identify cGLR1 as a double-stranded RNA sensor in the model organism Drosophila melanogaster. We show that RNA recognition activates Drosophila cGLR1 to synthesize the novel product cG[3'-5']pA[2'-5']p (3'2'-cGAMP). A crystal structure of Drosophila stimulator of interferon genes (dSTING) in complex with 3'2'-cGAMP explains selective isomer recognition, and 3'2'-cGAMP induces an enhanced antiviral state in vivo that protects from viral infection. Similar to radiation of Toll-like receptors in pathogen immunity, our results establish cGLRs as a diverse family of metazoan pattern recognition receptors.


Assuntos
Drosophila melanogaster/metabolismo , Nucleotídeos Cíclicos/metabolismo , Nucleotidiltransferases/metabolismo , RNA de Cadeia Dupla/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo , Sistemas do Segundo Mensageiro , Sequência de Aminoácidos , Animais , Cristalografia por Raios X , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/imunologia , Drosophila melanogaster/virologia , Feminino , Humanos , Imunidade Inata , Masculino , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Modelos Moleculares , Nucleotidiltransferases/química , Nucleotidiltransferases/imunologia , RNA de Cadeia Dupla/análise , RNA de Cadeia Dupla/imunologia , Receptores de Reconhecimento de Padrão/química , Receptores de Reconhecimento de Padrão/imunologia , Vírus/imunologia
8.
Nature ; 597(7874): 114-118, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34261128

RESUMO

In mammals, cyclic GMP-AMP (cGAMP) synthase (cGAS) produces the cyclic dinucleotide 2'3'-cGAMP in response to cytosolic DNA and this triggers an antiviral immune response. cGAS belongs to a large family of cGAS/DncV-like nucleotidyltransferases that is present in both prokaryotes1 and eukaryotes2-5. In bacteria, these enzymes synthesize a range of cyclic oligonucleotides and have recently emerged as important regulators of phage infections6-8. Here we identify two cGAS-like receptors (cGLRs) in the insect Drosophila melanogaster. We show that cGLR1 and cGLR2 activate Sting- and NF-κB-dependent antiviral immunity in response to infection with RNA or DNA viruses. cGLR1 is activated by double-stranded RNA to produce the cyclic dinucleotide 3'2'-cGAMP, whereas cGLR2 produces a combination of 2'3'-cGAMP and 3'2'-cGAMP in response to an as-yet-unidentified stimulus. Our data establish cGAS as the founding member of a family of receptors that sense different types of nucleic acids and trigger immunity through the production of cyclic dinucleotides beyond 2'3'-cGAMP.


Assuntos
Drosophila melanogaster/imunologia , Nucleotidiltransferases/imunologia , Receptores de Reconhecimento de Padrão/metabolismo , Vírus/imunologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/virologia , Feminino , Humanos , Imunidade Inata/genética , Imunidade Inata/imunologia , Ligantes , Masculino , Proteínas de Membrana/metabolismo , Modelos Moleculares , NF-kappa B/metabolismo , Nucleotídeos Cíclicos/metabolismo , Nucleotidiltransferases/classificação , Nucleotidiltransferases/deficiência , Nucleotidiltransferases/metabolismo , RNA de Cadeia Dupla/análise , RNA de Cadeia Dupla/imunologia , RNA de Cadeia Dupla/metabolismo , Receptores de Reconhecimento de Padrão/classificação , Receptores de Reconhecimento de Padrão/deficiência , Receptores de Reconhecimento de Padrão/imunologia
9.
Dev Comp Immunol ; 123: 104161, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34107277

RESUMO

Drosophila is a valuable paradigm for studying tumorigenesis and cancer. Mutations causing hematopoietic aberrations and melanotic-blood-tumors found in Drosophila mutants are vastly studied. Clear understanding about the blood cells, signaling pathways and the tissues affected during hematopoietic tumor formation provide an opportunity to delineate the effects of cancer therapeutics. Using this simple hematopoietic archetype, we elucidated the effects of the anti-cancer drug, Methotrexate (MTX) on immune responses in two scenarios i.e. against wasp infection and in hematopoietic mutant, hopTum-l. Through this in vivo study we show that MTX impedes the immune responses against wasp infection including the encapsulation response. We further observed that MTX reduces the tumor penetrance in gain-of-function mutants of JAK/STAT pathway, hopTum-l. MTX is anti-inflammatory as it hinders not only the immune responses of acute inflammation as observed after wasp infestation, but also chronic inflammatory responses associated with constitutively activated JAK/STAT pathway mutant (hopTum-l) carrying blood tumors.


Assuntos
Drosophila melanogaster/imunologia , Hemócitos/fisiologia , Imunidade/efeitos dos fármacos , Metotrexato/farmacologia , Vespas/fisiologia , Animais , Animais Geneticamente Modificados , Carcinogênese , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/parasitologia , Sistema Hematopoético , Janus Quinases/metabolismo , Larva , Mutação/genética , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais
10.
J Med Food ; 24(1): 101-109, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33449862

RESUMO

Carrageenan oligosaccharide (CAO), the hydrolysate of carrageenan from marine red algae, is used as a prebiotic additive or medical material. In this study, male Drosophila melanogaster was used as an animal model to explore the possibility that CAO can extend the life span through its relationship with antioxidation, immunity, and gut microbiota in vivo. The results show that a certain amount of CAO effectively prolonged the average life span and improved the climbing vitality and fecundity of male Drosophila. In addition, 0.125% CAO in the diet significantly increased the activity of Cu,Zn-superoxide dismutase (Cu,Zn-SOD) and catalase (CAT), reduced the content of malondialdehyde (MDA), and significantly repressed the expression of nuclear factor kappa B (NF-κB) gene in old male Drosophila tissues. In the intestinal microbiota analysis, 0.125% CAO in the diet increased the diversity of gut microbiota and improved the abundance of Commensalibacter at the genus level in Drosophila on the 40th day. The above results indicated that CAO supplementation could extend the life span of male Drosophila by improving antioxidant activity, immunity, and by regulating intestinal microflora.


Assuntos
Antioxidantes/metabolismo , Carragenina/farmacologia , Drosophila melanogaster/efeitos dos fármacos , Microbioma Gastrointestinal , Longevidade/efeitos dos fármacos , Oligossacarídeos/farmacologia , Animais , Drosophila melanogaster/imunologia , Sistema Imunitário/efeitos dos fármacos , Masculino
11.
Elife ; 92020 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-33377870

RESUMO

Postmitotic tissues are incapable of replacing damaged cells through proliferation, but need to rely on buffering mechanisms to prevent tissue disintegration. By constitutively activating the Ras/MAPK-pathway via RasV12-overexpression in the postmitotic salivary glands (SGs) of Drosophila larvae, we overrode the glands adaptability to growth signals and induced hypertrophy. The accompanied loss of tissue integrity, recognition by cellular immunity, and cell death are all buffered by blocking stress signaling through a genuine tissue-autonomous immune response. This novel, spatio-temporally tightly regulated mechanism relies on the inhibition of a feedback-loop in the JNK-pathway by the immune effector and antimicrobial peptide Drosomycin. While this interaction might allow growing SGs to cope with temporary stress, continuous Drosomycin expression in RasV12-glands favors unrestricted hypertrophy. These findings indicate the necessity to refine therapeutic approaches that stimulate immune responses by acknowledging their possible, detrimental effects in damaged or stressed tissues.


Tissues and organs work hard to maintain balance in everything from taking up nutrients to controlling their growth. Ageing, wounding, sickness, and changes in the genetic code can all alter this balance, and cause the tissue or organ to lose some of its cells. Many tissues restore this loss by dividing their remaining cells to fill in the gaps. But some ­ like the salivary glands of fruit fly larvae ­ have lost this ability. Tissues like these rely on being able to sense and counteract problems as they arise so as to not lose their balance in the first place. The immune system and stress responses are crucial for this process. They trigger steps to correct the problem and interact with each other to find a common decision about the fate of the affected tissue. To better understand how the immune system and stress response work together, Krautz, Khalili and Theopold genetically manipulated cells in the salivary gland of fruit fly larvae. These modifications switched on signals that stimulate cells to keep growing, causing the salivary gland's tissue to slowly lose its balance and trigger the stress and immune response. The experiments showed that while the stress response instructed the cells in the gland to die, a peptide released by the immune system called Drosomycin blocked this response and prevented the tissue from collapsing. The cells in the part of the gland not producing this immune peptide were consequently killed by the stress response. When all the cells in the salivary gland were forced to produce Drosomycin, none of the cells died and the whole tissue survived. But it also allowed the cells in the gland to grow uncontrollably, like a tumor, threatening the health of the entire organism. Mapping the interactions between immune and stress pathways could help to fine-tune treatments that can prevent tissue damage. Fruit flies share many genetic features and molecular pathways with humans. So, the next step towards these kinds of treatments would be to screen for similar mechanisms that block stress activation in damaged human tissues. But this research carries a warning: careless activation of the immune system to protect stressed tissues could lead to uncontrolled tissue growth, and might cause more harm than good.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/imunologia , IMP Desidrogenase/metabolismo , Transdução de Sinais/fisiologia , Estresse Fisiológico/imunologia , Animais , Proteínas de Drosophila/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hipertrofia , IMP Desidrogenase/genética , Larva , Sistema de Sinalização das MAP Quinases
12.
Elife ; 92020 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-33372660

RESUMO

Studies in different animal model systems have revealed the impact of odors on immune cells; however, any understanding on why and how odors control cellular immunity remained unclear. We find that Drosophila employ an olfactory-immune cross-talk to tune a specific cell type, the lamellocytes, from hematopoietic-progenitor cells. We show that neuronally released GABA derived upon olfactory stimulation is utilized by blood-progenitor cells as a metabolite and through its catabolism, these cells stabilize Sima/HIFα protein. Sima capacitates blood-progenitor cells with the ability to initiate lamellocyte differentiation. This systemic axis becomes relevant for larvae dwelling in wasp-infested environments where chances of infection are high. By co-opting the olfactory route, the preconditioned animals elevate their systemic GABA levels leading to the upregulation of blood-progenitor cell Sima expression. This elevates their immune-potential and primes them to respond rapidly when infected with parasitic wasps. The present work highlights the importance of the olfaction in immunity and shows how odor detection during animal development is utilized to establish a long-range axis in the control of blood-progenitor competency and immune-priming.


Assuntos
Fenômenos Bioquímicos/imunologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Células-Tronco Hematopoéticas/citologia , Hemócitos/citologia , Animais , Drosophila/imunologia , Drosophila/metabolismo , Proteínas de Drosophila/imunologia , Drosophila melanogaster/imunologia , Hematopoese/imunologia , Larva/metabolismo , Vespas/imunologia
13.
PLoS Pathog ; 16(9): e1008328, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32936835

RESUMO

Candida albicans cells depend on the energy derived from amino acid catabolism to induce and sustain hyphal growth inside phagosomes of engulfing macrophages. The concomitant deamination of amino acids is thought to neutralize the acidic microenvironment of phagosomes, a presumed requisite for survival and initiation of hyphal growth. Here, in contrast to an existing model, we show that mitochondrial-localized NAD+-dependent glutamate dehydrogenase (GDH2) catalyzing the deamination of glutamate to α-ketoglutarate, and not the cytosolic urea amidolyase (DUR1,2), accounts for the observed alkalization of media when amino acids are the sole sources of carbon and nitrogen. C. albicans strains lacking GDH2 (gdh2-/-) are viable and do not extrude ammonia on amino acid-based media. Environmental alkalization does not occur under conditions of high glucose (2%), a finding attributable to glucose-repression of GDH2 expression and mitochondrial function. Consistently, inhibition of oxidative phosphorylation or mitochondrial translation by antimycin A or chloramphenicol, respectively, prevents alkalization. GDH2 expression and mitochondrial function are derepressed as glucose levels are lowered from 2% (~110 mM) to 0.2% (~11 mM), or when glycerol is used as primary carbon source. Using time-lapse microscopy, we document that gdh2-/- cells survive, filament and escape from primary murine macrophages at rates indistinguishable from wildtype. In intact hosts, such as in fly and murine models of systemic candidiasis, gdh2-/- mutants are as virulent as wildtype. Thus, although Gdh2 has a critical role in central nitrogen metabolism, Gdh2-catalyzed deamination of glutamate is surprisingly dispensable for escape from macrophages and virulence. Consistently, using the pH-sensitive dye (pHrodo), we observed no significant difference between wildtype and gdh2-/- mutants in phagosomal pH modulation. Following engulfment of fungal cells, the phagosomal compartment is rapidly acidified and hyphal growth initiates and sustained under consistently acidic conditions within phagosomes. Together, our results demonstrate that amino acid-dependent alkalization is not essential for hyphal growth, survival in macrophages and hosts. An accurate understanding of the microenvironment within macrophage phagosomes and the metabolic events underlying the survival of phagocytized C. albicans cells and their escape are critical to understanding the host-pathogen interactions that ultimately determine the pathogenic outcome.


Assuntos
Candida albicans/imunologia , Candidíase/imunologia , Drosophila melanogaster/imunologia , Glutamato Desidrogenase/metabolismo , Macrófagos/imunologia , Aminoácidos/genética , Aminoácidos/metabolismo , Animais , Candida albicans/patogenicidade , Candidíase/metabolismo , Candidíase/microbiologia , Drosophila melanogaster/metabolismo , Drosophila melanogaster/microbiologia , Feminino , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Glutamato Desidrogenase/genética , Interações Hospedeiro-Patógeno , Concentração de Íons de Hidrogênio , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Nitrogênio , Fagossomos/imunologia , Fagossomos/metabolismo , Fagossomos/microbiologia , Virulência
14.
Genes Cells ; 25(10): 675-684, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32865275

RESUMO

Immunity is considered to be involved in the prevention of cancer. Although both humoral and cellular immune reactions may participate, underlying mechanisms have yet to be clarified. The present study was conducted to clarify this issue using a Drosophila model, in which neoplastic transformation was induced through the simultaneous inhibition of cell-cycle checkpoints and apoptosis. We first determined the location of hemocytes, blood cells of Drosophila playing a role of immune cells, in neoplasia-induced and normal larvae, but there was no significant difference between the two groups. When gene expression pattern in larval hemocytes was determined, the expression of immunity-related genes including those necessary for phagocytosis was reduced in the neoplasia model. We then asked the involvement of phagocytosis in the prevention of neoplasia examining animals where the expression of engulfment receptors instead of apoptosis was retarded. We found that the inhibition of engulfment receptor expression augmented the occurrence of neoplasia induced by a defect in cell-cycle checkpoints. This suggested a role for phagocytosis in the prevention of neoplastic transformation in Drosophila.


Assuntos
Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/metabolismo , Fagocitose/imunologia , Animais , Apoptose/imunologia , Linhagem Celular , Transformação Celular Neoplásica/genética , Drosophila melanogaster/genética , Drosophila melanogaster/imunologia , Drosophila melanogaster/metabolismo , Feminino , Hemócitos/citologia , Hemócitos/imunologia , Hemócitos/metabolismo , Larva/metabolismo , Masculino , Neoplasias/genética , Neoplasias/imunologia , Fagocitose/genética , Fagocitose/fisiologia
15.
Curr Biol ; 30(17): 3316-3329.e5, 2020 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-32649911

RESUMO

During hematopoiesis, progenitor cells receive and interpret a diverse array of regulatory signals from their environment. These signals control the maintenance of the progenitors and regulate the production of mature blood cells. Integrins are well known in vertebrates for their roles in hematopoiesis, particularly in assisting in the migration to, as well as the physical attachment of, progenitors to the niche. However, whether and how integrins are also involved in the signaling mechanisms that control hematopoiesis remains to be resolved. Here, we show that integrins play a key role during fly hematopoiesis in regulating cell signals that control the behavior of hematopoietic progenitors. Integrins can regulate hematopoiesis directly, via focal adhesion kinase (FAK) signaling, and indirectly, by directing extracellular matrix (ECM) assembly and/or maintenance. ECM organization and density controls blood progenitor behavior by modulating multiple signaling pathways, including bone morphogenetic protein (BMP) and Hedgehog (Hh). Furthermore, we show that integrins and the ECM are reduced following infection, which may assist in activating the immune response. Our results provide mechanistic insight into how integrins can shape the signaling environment around hematopoietic progenitors.


Assuntos
Células Sanguíneas/imunologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/imunologia , Matriz Extracelular/fisiologia , Hematopoese , Integrinas/metabolismo , Animais , Células Sanguíneas/metabolismo , Células Sanguíneas/parasitologia , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/parasitologia , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Integrinas/genética , Transdução de Sinais , Vespas/fisiologia
16.
J Cell Biol ; 219(7)2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32539109

RESUMO

Tissue damage triggers a rapid and robust inflammatory response in order to clear and repair a wound. Remarkably, many of the cell biology features that underlie the ability of leukocytes to home in to sites of injury and to fight infection-most of which are topics of intensive current research-were originally observed in various weird and wonderful translucent organisms over a century ago by Elie Metchnikoff, the "father of innate immunity," who is credited with discovering phagocytes in 1882. In this review, we use Metchnikoff's seminal lectures as a starting point to discuss the tremendous variety of cell biology features that underpin the function of these multitasking immune cells. Some of these are shared by other cell types (including aspects of motility, membrane trafficking, cell division, and death), but others are more unique features of innate immune cells, enabling them to fulfill their specialized functions, such as encapsulation of invading pathogens, cell-cell fusion in response to foreign bodies, and their self-sacrifice as occurs during NETosis.


Assuntos
Adaptação Fisiológica/imunologia , Imunidade Inata , Macrófagos/imunologia , Neutrófilos/imunologia , Cicatrização/imunologia , Ferimentos Penetrantes/imunologia , Alarminas/imunologia , Animais , Drosophila melanogaster/imunologia , Drosophila melanogaster/microbiologia , História do Século XIX , História do Século XX , História do Século XXI , Humanos , Inflamação/história , Macrófagos/microbiologia , Monócitos/imunologia , Monócitos/microbiologia , Neutrófilos/microbiologia , Moléculas com Motivos Associados a Patógenos/imunologia , Fagocitose , Ferimentos Penetrantes/microbiologia , Peixe-Zebra/imunologia , Peixe-Zebra/microbiologia
17.
Immunol Lett ; 226: 7-11, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32598968

RESUMO

As members of the mitogen-activated protein kinase (MAPK) family, the c-Jun N-terminal kinases (JNKs) regulate cell responses to a wide range of extrinsic and intrinsic insults, including irradiation, reactive oxygen species (ROS), DNA damage, heat, bacterial antigens, and inflammatory cytokines. Particularly, JNK signaling regulates and promotes many important physiological processes that influence metabolic and tissue homeostasis, cell death/survival, and cell damage repair and ultimately impacts the lifespan of an organism. This diverse functionality causes a variety of tissue-specific and context-specific cellular responses, mediated by various cross talks between JNK and other cellular signaling pathways. Thus, highlighting its significance as a determinant of stress responses, JNK loss-of-function mutations have been implicated in a multitude of pathologies, including neurodegenerative diseases, diabetes, and cancer. Because JNK functions are specified in a context-dependent manner and can greatly vary, the underlying causes for these different outcomes remain largely unresolved despite the gained knowledge of many regulatory roles of JNK signaling during the past two decades. In Drosophila melanogaster, JNK signaling is conserved and required for immune responses, as well as the development for morphogenetic processes (embryonic dorsal closure and thorax closure). Therefore, Drosophila innate immunity provides the ideal model to understand the complex mechanisms underlying JNK activation and regulation. In the following, we review studies in Drosophila that highlight several mechanisms by which JNK signaling influences immunity and homeostasis.


Assuntos
Drosophila melanogaster/imunologia , MAP Quinase Quinase 4/metabolismo , Animais , Morte Celular , Regulação da Expressão Gênica no Desenvolvimento , Homeostase , Humanos , Imunidade Inata , Espécies Reativas de Oxigênio , Transdução de Sinais
18.
Biochem Biophys Res Commun ; 526(4): 1106-1111, 2020 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-32312516

RESUMO

The immune system protects its host from not only invading parasites and parasitoids, but also altered self tissue, including dying cells. Necrotic cells are strongly immunogenic, but in Drosophila this has not been directly addressed, due partially to the fact that knowledge about necrosis in Drosophila currently lags behind that for other models. Upon the loss of cell matrix attachment, endocycling polyploid tissues of the Drosophila larva undergo autophagy instead of apoptosis; we employed this system as a model to examine cell death modalities and immunity. Here, we report that larval fat body cells depleted of integrin undergo not only autophagy, but also necrotic cell death, and that a blockade of reaper, grim, hid, or the downstream caspases enhances necrosis. These cells elicit melanotic mass formation, an autoimmune-like response. We also show that necrosis is the main cause of melanotic mass formation in these anchorage-depleted polyploid cells.


Assuntos
Drosophila melanogaster/imunologia , Melaninas/metabolismo , Necrose/patologia , Animais , Morte Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Dosagem de Genes , Integrinas/metabolismo , Larva/imunologia , Larva/metabolismo , Fenótipo , Poliploidia
19.
Nat Commun ; 11(1): 1580, 2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-32221286

RESUMO

ADAR RNA editing enzymes are high-affinity dsRNA-binding proteins that deaminate adenosines to inosines in pre-mRNA hairpins and also exert editing-independent effects. We generated a Drosophila AdarE374A mutant strain encoding a catalytically inactive Adar with CRISPR/Cas9. We demonstrate that Adar adenosine deamination activity is necessary for normal locomotion and prevents age-dependent neurodegeneration. The catalytically inactive protein, when expressed at a higher than physiological level, can rescue neurodegeneration in Adar mutants, suggesting also editing-independent effects. Furthermore, loss of Adar RNA editing activity leads to innate immune induction, indicating that Drosophila Adar, despite being the homolog of mammalian ADAR2, also has functions similar to mammalian ADAR1. The innate immune induction in fly Adar mutants is suppressed by silencing of Dicer-2, which has a RNA helicase domain similar to MDA5 that senses unedited dsRNAs in mammalian Adar1 mutants. Our work demonstrates that the single Adar enzyme in Drosophila unexpectedly has dual functions.


Assuntos
Adenosina Desaminase/genética , Encéfalo/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/imunologia , Imunidade Inata/genética , Edição de RNA/genética , Adenosina Desaminase/química , Monofosfato de Adenosina/metabolismo , Envelhecimento/patologia , Animais , Catálise , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica , Locomoção , Degeneração Neural/patologia , Mutação Puntual/genética , Domínios Proteicos , RNA Helicases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ribonuclease III/metabolismo
20.
J Cell Biol ; 219(3)2020 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-31940424

RESUMO

Phagocytes use their actomyosin cytoskeleton to migrate as well as to probe their environment by phagocytosis or macropinocytosis. Although migration and extracellular material uptake have been shown to be coupled in some immune cells, the mechanisms involved in such coupling are largely unknown. By combining time-lapse imaging with genetics, we here identify the lysosomal Ca2+ channel Trpml as an essential player in the coupling of cell locomotion and phagocytosis in hemocytes, the Drosophila macrophage-like immune cells. Trpml is needed for both hemocyte migration and phagocytic processing at distinct subcellular localizations: Trpml regulates hemocyte migration by controlling actomyosin contractility at the cell rear, whereas its role in phagocytic processing lies near the phagocytic cup in a myosin-independent fashion. We further highlight that Vamp7 also regulates phagocytic processing and locomotion but uses pathways distinct from those of Trpml. Our results suggest that multiple mechanisms may have emerged during evolution to couple phagocytic processing to cell migration and facilitate space exploration by immune cells.


Assuntos
Actomiosina/metabolismo , Movimento Celular , Citoesqueleto/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Hemócitos/metabolismo , Lisossomos/metabolismo , Macrófagos/metabolismo , Fagocitose , Canais de Potencial de Receptor Transitório/metabolismo , Actomiosina/genética , Animais , Animais Geneticamente Modificados , Cálcio/metabolismo , Sinalização do Cálcio , Citoesqueleto/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/imunologia , Hemócitos/imunologia , Lisossomos/genética , Macrófagos/imunologia , Miosina Tipo II/genética , Miosina Tipo II/metabolismo , Proteínas R-SNARE/genética , Proteínas R-SNARE/metabolismo , Fatores de Tempo , Canais de Potencial de Receptor Transitório/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA