Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34620709

RESUMO

Germ cells possess the Piwi-interacting RNA pathway to repress transposable elements and maintain genome stability across generations. Transposable element mobilization in somatic cells does not affect future generations, but nonetheless can lead to pathological outcomes in host tissues. We show here that loss of function of the conserved zinc-finger transcription factor Hinfp causes dysregulation of many host genes and derepression of most transposable elements. There is also substantial DNA damage in somatic tissues of Drosophila after loss of Hinfp. Interference of transposable element mobilization by reverse-transcriptase inhibitors can suppress some of the DNA damage phenotypes. The key cell-autonomous target of Hinfp in this process is Histone1, which encodes linker histones essential for higher-order chromatin assembly. Transgenic expression of Hinfp or Histone1, but not Histone4 of core nucleosome, is sufficient to rescue the defects in repressing transposable elements and host genes. Loss of Hinfp enhances Ras-induced tissue growth and aging-related phenotypes. Therefore, Hinfp is a physiological regulator of Histone1-dependent silencing of most transposable elements, as well as many host genes, and serves as a venue for studying genome instability, cancer progression, neurodegeneration, and aging.


Assuntos
Elementos de DNA Transponíveis/genética , Drosophila melanogaster/genética , Instabilidade Genômica/genética , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Animais , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Histonas/genética , Histonas/metabolismo , RNA Interferente Pequeno/genética
2.
J Biol Chem ; 294(26): 10172-10181, 2019 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-31088910

RESUMO

The Toll signaling pathway in Drosophila melanogaster regulates several immune-related functions, including the expression of antimicrobial peptide (AMP) genes. The canonical Toll receptor (Toll-1) is activated by the cytokine Spätzle (Spz-1), but Drosophila encodes eight other Toll genes and five other Spz genes whose interactions with one another and associated functions are less well-understood. Here, we conducted in vitro assays in the Drosophila S2 cell line with the Toll/interleukin-1 receptor (TIR) homology domains of each Toll family member to determine whether they can activate a known target of Toll-1, the promoter of the antifungal peptide gene drosomycin. All TIR family members activated the drosomycin promoter, with Toll-1 and Toll-7 TIRs producing the highest activation. We found that the Toll-1 and Toll-7 ectodomains bind Spz-1, -2, and -5, and also vesicular stomatitis virus (VSV) virions, and that Spz-1, -2, -5, and VSV all activated the promoters of drosomycin and several other AMP genes in S2 cells expressing full-length Toll-1 or Toll-7. In vivo experiments indicated that Toll-1 and Toll-7 mutants could be systemically infected with two bacterial species (Enterococcus faecalis and Pseudomonas aeruginosa), the opportunistic fungal pathogen Candida albicans, and VSV with different survival times in adult females and males compared with WT fly survival. Our results suggest that all Toll family members can activate several AMP genes. Our results further indicate that Toll-1 and Toll-7 bind multiple Spz proteins and also VSV, but they differentially affect adult survival after systemic infection, potentially because of sex-specific differences in Toll-1 and Toll-7 expression.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Infecções Bacterianas/microbiologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica , Receptores Toll-Like/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Bactérias/isolamento & purificação , Infecções Bacterianas/genética , Infecções Bacterianas/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/microbiologia , Feminino , Masculino , Regiões Promotoras Genéticas , Transdução de Sinais , Receptores Toll-Like/genética
3.
J Cell Physiol ; 234(12): 21925-21936, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31042012

RESUMO

The intestinal epithelium has a high rate of cell turn over and is an excellent system to study stem cell-mediated tissue homeostasis. The Misshapen subfamily of the Ste20 kinases in mammals consists of misshapen like kinase 1 (MINK1), mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4), and TRAF2 and NCK interacting kinase (TNIK). Recent reports suggest that this subfamily has a novel function equal to the Hippo/MST subfamily as upstream kinases for Warts/Large tumor suppressor kinase (LATS) to suppress tissue growth. To study the in vivo functions of Mink1, Map4k4, and Tnik, we generated a compound knockout of these three genes in the mouse intestinal epithelium. The intestinal epithelia of the mutant animals were phenotypically normal up to approximately 12 months. The older animals then exhibited mildly increased proliferation throughout the lower GI tract. We also observed that the normally spatially organized Paneth cells in the crypt base became dispersed. The expression of one of the YAP pathway target genes Sox9 was increased while other target genes including CTGF did not show a significant change. Therefore, the Misshapen and Hippo subfamilies may have highly redundant functions to regulate growth in the intestinal epithelium, as illustrated in recent tissue culture models.


Assuntos
Envelhecimento , Proliferação de Células/fisiologia , Mucosa Intestinal/metabolismo , Células-Tronco/metabolismo , Animais , Camundongos Transgênicos , Fosforilação/fisiologia
4.
Development ; 141(13): 2621-32, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24961799

RESUMO

Intestinal stem cells (ISCs) in the adult Drosophila midgut can respond to tissue damage and support repair. We used genetic manipulation to increase the number of ISC-like cells in the adult midgut and performed gene expression profiling to identify potential ISC regulators. A detailed analysis of one of these potential regulators, the zinc-finger protein Charlatan, was carried out. MARCM clonal analysis and RNAi in precursor cells showed that loss of Chn function caused severe ISC division defects, including loss of EdU incorporation, phosphorylated histone 3 staining and expression of the mitotic protein Cdc2. Loss of Charlatan also led to a much reduced histone acetylation staining in precursor cells. Both the histone acetylation and ISC division defects could be rescued by the simultaneous decrease of the Histone Deacetylase 2. The overexpression of Charlatan blocked differentiation reversibly, but loss of Charlatan did not lead to automatic differentiation. The results together suggest that Charlatan does not simply act as an anti-differentiation factor but instead functions to maintain a chromatin structure that is compatible with stem cell properties, including proliferation.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila/genética , Intestinos/citologia , Células-Tronco/fisiologia , Fatores de Transcrição/fisiologia , Animais , Diferenciação Celular/genética , Drosophila/fisiologia , Perfilação da Expressão Gênica , Análise em Microsséries , Microscopia de Fluorescência , Interferência de RNA
5.
PLoS Genet ; 9(6): e1003556, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23818862

RESUMO

The sphingolipid ceramide elicits several stress responses, however, organisms survive despite increased ceramide but how they do so is poorly understood. We demonstrate here that the AKT/FOXO pathway regulates survival in increased ceramide environment by metabolic adaptation involving changes in glycolysis and lipolysis through novel downstream targets. We show that ceramide kinase mutants accumulate ceramide and this leads to reduction in energy levels due to compromised oxidative phosphorylation. Mutants show increased activation of Akt and a consequent decrease in FOXO levels. These changes lead to enhanced glycolysis by upregulating the activity of phosphoglyceromutase, enolase, pyruvate kinase, and lactate dehydrogenase to provide energy. A second major consequence of AKT/FOXO reprogramming in the mutants is the increased mobilization of lipid from the gut through novel lipase targets, CG8093 and CG6277 for energy contribution. Ubiquitous reduction of these targets by knockdown experiments results in semi or total lethality of the mutants, demonstrating the importance of activating them. The efficiency of these adaptive mechanisms decreases with age and leads to reduction in adult life span of the mutants. In particular, mutants develop cardiac dysfunction with age, likely reflecting the high energy requirement of a well-functioning heart. The lipases also regulate physiological triacylglycerol homeostasis and are important for energy metabolism since midgut specific reduction of them in wild type flies results in increased sensitivity to starvation and accumulation of triglycerides leading to cardiac defects. The central findings of increased AKT activation, decreased FOXO level and activation of phosphoglyceromutase and pyruvate kinase are also observed in mice heterozygous for ceramide transfer protein suggesting a conserved role of this pathway in mammals. These data reveal novel glycolytic and non-autonomous lipolytic pathways in response to increased ceramide for sustenance of high energy demanding organ functions like the heart.


Assuntos
Ceramidas/metabolismo , Fatores de Transcrição Forkhead/genética , Proteína Oncogênica v-akt/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Estresse Fisiológico/genética , Animais , Ceramidas/farmacologia , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Metabolismo Energético/genética , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glicólise/genética , Lipólise/genética , Camundongos , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais/efeitos dos fármacos
6.
J Cell Physiol ; 229(4): 512-520, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24343480

RESUMO

The Toll and Toll-like receptor signaling pathways are evolutionarily conserved pathways that regulate innate immunity in insects and mammals. While efforts have been made to clarify the signal transduction events that occur during infection, much less is known about the components that maintain immune quiescence. Here we show that retromer, an intracellular protein complex known for regulating vesicle trafficking, functions in modulating the Toll pathway in Drosophila melanogaster. In mutant animals lacking retromer function, the Toll pathway but not JAK-STAT or IMD pathway is activated, triggering both cellular and humoral responses. Genetic epistasis and clonal analysis suggest that retromer regulates a component that acts upstream of Toll. Our data further show that in the mutant the Toll ligand Spätzle has a processing pattern similar to that of after infection. Together, the results suggest a novel function of retromer in regulating Toll pathway and innate immunity at a step that modulates ligand processing or activity.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica/imunologia , Complexos Multiproteicos/metabolismo , Receptores Toll-Like/metabolismo , Animais , Proteínas de Drosophila/genética , Imunidade Inata , Janus Quinases/genética , Janus Quinases/metabolismo , Larva , Mutação , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Receptores Toll-Like/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
7.
Proc Natl Acad Sci U S A ; 108(27): 11127-32, 2011 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-21690388

RESUMO

The level of TGF-ß/bone morphogenetic protein (BMP) signaling through Smad is tightly regulated to ensure proper embryonic patterning and homeostasis. Here we show that Smad activation by TGF-ß/BMP is blocked by a highly conserved phosphorylation event in the α-helix 1 region of Smad [T312 in Drosophila Smad1 (MAD)]. α-helix 1 phosphorylation reduces Smad interaction with TGF-ß/BMP receptor kinase and affects all receptor-activated Smads except Smad3. Tissue culture and transgenic studies in Drosophila further demonstrate that the biological activity of MAD is repressed by T312 phosphorylation in vivo. Through RNAi screening of the kinome, we have identified Misshapen (Msn) and the mammalian orthologs TNIK, MINK1, and MAP4K4 as the kinases responsible for α-helix 1 phosphorylation. Targeted expression of an active form of Msn in the wing imaginal disk disrupted activation of endogenous MAD by Dpp and expression of the Dpp/MAD target gene. Msn kinases belong to the Ste20 kinase family that has been shown to act as MAP kinase kinase kinase kinase (MAP4K). Our findings thus reveal a function of Msn independent of its impact on MAP kinase cascades. This Smad inhibition mechanism by Msn likely has important implications for development and disease.


Assuntos
Proteínas de Drosophila/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Smad/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Sítios de Ligação , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Linhagem Celular , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Genes de Insetos , Humanos , Dados de Sequência Molecular , Fosforilação , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Estrutura Secundária de Proteína , Interferência de RNA , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Proteínas Smad/química , Proteínas Smad/genética , Proteínas Smad/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo
8.
bioRxiv ; 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38746415

RESUMO

Studies on Hippo pathway regulation of tumorigenesis largely center on YAP and TAZ, the transcriptional co-regulators of TEAD. Here, we present an oncogenic mechanism involving VGLL and TEAD fusions that is Hippo pathway-related but YAP/TAZ-independent. We characterize two recurrent fusions, VGLL2-NCOA2 and TEAD1-NCOA2, recently identified in spindle cell rhabdomyosarcoma. We demonstrate that, in contrast to VGLL2 and TEAD1, the fusion proteins are strong activators of TEAD-dependent transcription, and their function does not require YAP/TAZ. Furthermore, we identify that VGLL2 and TEAD1 fusions engage specific epigenetic regulation by recruiting histone acetyltransferase p300 to control TEAD-mediated transcriptional and epigenetic landscapes. We showed that small molecule p300 inhibition can suppress fusion proteins-induced oncogenic transformation both in vitro and in vivo. Overall, our study reveals a molecular basis for VGLL involvement in cancer and provides a framework for targeting tumors carrying VGLL, TEAD, or NCOA translocations.

9.
Proc Natl Acad Sci U S A ; 107(33): 14715-20, 2010 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-20679214

RESUMO

The innate immune response in Drosophila involves the inducible expression of antimicrobial peptide genes mediated by the Toll and IMD signaling pathways. Dorsal and DIF act downstream of Toll, whereas Relish acts downstream of IMD to regulate target gene expression. Dorsal, DIF, and Relish are NF-kappaB-related transcription factors and function as obligate dimers, but it is not clear how the various dimer combinations contribute to the innate immune response. We systematically examined the dimerization tendency of these proteins through the use of transgenic assays. The results show that all combinations of homo- and heterodimers are formed, but with varying degrees of efficiency. The formation of the DIF-Relish heterodimer is particularly interesting because it may mediate signaling for the seemingly independent Toll and IMD pathways. By incorporating a flexible peptide linker, we specifically tested the functions of the DIF;Relish (a ; sign represents the peptide linker) linked heterodimer. Our results demonstrate that the linked heterodimer can activate target genes of both the Toll and IMD pathways. The DIF and Relish complex is detectable in whole animal extracts, suggesting that this heterodimer may function in vivo to increase the spectrum and level of antimicrobial peptide production in response to different infections.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Fatores de Transcrição/metabolismo , Animais , Animais Geneticamente Modificados , Peptídeos Catiônicos Antimicrobianos/genética , Western Blotting , Clonagem Molecular , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/microbiologia , Escherichia coli/fisiologia , Feminino , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Masculino , NF-kappa B/química , NF-kappa B/genética , NF-kappa B/metabolismo , Multimerização Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Staphylococcus aureus/fisiologia , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/genética
10.
Proc Natl Acad Sci U S A ; 107(49): 21064-9, 2010 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-21078993

RESUMO

Intestinal stem cells (ISCs) in the Drosophila adult midgut are essential for maintaining tissue homeostasis and replenishing lost cells in response to tissue damage. Here we demonstrate that the Hippo (Hpo) signaling pathway, an evolutionarily conserved pathway implicated in organ size control and tumorigenesis, plays an essential role in regulating ISC proliferation. Loss of Hpo signaling in either midgut precursor cells or epithelial cells stimulates ISC proliferation. We provide evidence that loss of Hpo signaling in epithelial cells increases the production of cytokines of the Upd family and multiple EGFR ligands that activate JAK-STAT and EGFR signaling pathways in ISCs to stimulate their proliferation, thus revealing a unique non-cell-autonomous role of Hpo signaling in blocking ISC proliferation. Finally, we show that the Hpo pathway mediator Yorkie (Yki) is also required in precursor cells for injury-induced ISC proliferation in response to tissue-damaging reagent DSS.


Assuntos
Proliferação de Células , Proteínas de Drosophila/fisiologia , Intestinos/citologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia , Animais , Citocinas/biossíntese , Proteínas de Drosophila/metabolismo , Células Epiteliais , Receptores ErbB/metabolismo , Janus Quinases/metabolismo , Proteínas Nucleares , Receptores de Peptídeos de Invertebrados/metabolismo , Transativadores , Proteínas de Sinalização YAP
11.
Proc Natl Acad Sci U S A ; 107(46): 19939-44, 2010 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-21041636

RESUMO

In the Drosophila ovary, bone morphogenetic protein (BMP) signaling activated by the niche promotes germline stem cell (GSC) self-renewal and proliferation, whereas E-cadherin-mediated cell adhesion anchors GSCs in the niche for their continuous self-renewal. Here we show that Lissencephaly-1 (Lis1) regulates BMP signaling and E-cadherin-mediated adhesion between GSCs and their niche and thereby controls GSC self-renewal. Lis1 mutant GSCs are lost faster than control GSCs because of differentiation but not because of cell death, indicating that Lis1 controls GSC self-renewal. The Lis1 mutant GSCs exhibit reduced BMP signaling activity, and Lis1 interacts genetically with the BMP pathway components in the regulation of GSC maintenance. Mechanistically, Lis1 binds directly to and stabilizes the SMAD protein Mothers against decapentaplegic (Mad), facilitates its phosphorylation, and thereby regulates BMP signaling. Finally, the Lis1 mutant GSCs accumulate less E-cadherin in the stem cell-niche junction than do their wild-type counterparts. Germline-specific expression of an activated BMP receptor thickveins (Tkv) or E-cadherin can partially rescue the loss phenotype of Lis1 mutant GSCs. Therefore, this study has revealed a role of Lis1 in the control of Drosophila ovarian GSC self-renewal, at least partly by regulating niche signal transduction and niche adhesion. It has been known that Lis1 controls neural precursor/stem cell proliferation in the developing mammalian brain; this study further suggests that Lis1, which is widely expressed in adult mammalian tissues, could regulate adult tissue stem cells through modulating niche signaling and adhesion.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Células Germinativas/citologia , Proteínas Associadas aos Microtúbulos/metabolismo , Transdução de Sinais , Nicho de Células-Tronco/citologia , Células-Tronco/citologia , Animais , Caderinas/metabolismo , Adesão Celular , Proliferação de Células , Proteínas de Ligação a DNA/metabolismo , Drosophila melanogaster/metabolismo , Células Germinativas/metabolismo , Fosforilação , Ligação Proteica , Estabilidade Proteica , Nicho de Células-Tronco/metabolismo , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo
12.
Sci Adv ; 9(21): eadc9660, 2023 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-37224252

RESUMO

Adult stem cells are essential for tissue maintenance and repair. Although genetic pathways for controlling adult stem cells are extensively investigated in various tissues, much less is known about how mechanosensing could regulate adult stem cells and tissue growth. Here, we demonstrate that shear stress sensing regulates intestine stem cell proliferation and epithelial cell number in adult Drosophila. Ca2+ imaging in ex vivo midguts shows that shear stress, but not other mechanical forces, specifically activates enteroendocrine cells among all epithelial cell types. This activation is mediated by transient receptor potential A1 (TrpA1), a Ca2+-permeable channel expressed in enteroendocrine cells. Furthermore, specific disruption of shear stress, but not chemical, sensitivity of TrpA1 markedly reduces proliferation of intestinal stem cells and midgut cell number. Therefore, we propose that shear stress may act as a natural mechanical stimulation to activate TrpA1 in enteroendocrine cells, which, in turn, regulates intestine stem cell behavior.


Assuntos
Células-Tronco Adultas , Proteínas de Drosophila , Drosophila , Canais Iônicos , Animais , Proliferação de Células , Intestinos/citologia , Estresse Mecânico , Canais Iônicos/metabolismo , Proteínas de Drosophila/metabolismo
13.
J Cell Biol ; 178(6): 981-94, 2007 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-17785517

RESUMO

Nuclear translocation of Smad proteins is a critical step in signal transduction of transforming growth factor beta (TGF-beta) and bone morphogenetic proteins (BMPs). Using nuclear accumulation of the Drosophila Smad Mothers against Decapentaplegic (Mad) as the readout, we carried out a whole-genome RNAi screening in Drosophila cells. The screen identified moleskin (msk) as important for the nuclear import of phosphorylated Mad. Genetic evidence in the developing eye imaginal discs also demonstrates the critical functions of msk in regulating phospho-Mad. Moreover, knockdown of importin 7 and 8 (Imp7 and 8), the mammalian orthologues of Msk, markedly impaired nuclear accumulation of Smad1 in response to BMP2 and of Smad2/3 in response to TGF-beta. Biochemical studies further suggest that Smads are novel nuclear import substrates of Imp7 and 8. We have thus identified new evolutionarily conserved proteins that are important in the signal transduction of TGF-beta and BMP into the nucleus.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Drosophila/fisiologia , Drosophila/metabolismo , Carioferinas/fisiologia , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Proteína Morfogenética Óssea 2 , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Genoma de Inseto , Humanos , Carioferinas/metabolismo , Interferência de RNA , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo
14.
Cell Rep ; 39(7): 110817, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35584678

RESUMO

Drosophila Toll-1 and all mammalian Toll-like receptors regulate innate immunity. However, the functions of the remaining eight Toll-related proteins in Drosophila are not fully understood. Here, we show that Drosophila Toll-9 is necessary and sufficient for a special form of compensatory proliferation after apoptotic cell loss (undead apoptosis-induced proliferation [AiP]). Mechanistically, for AiP, Toll-9 interacts with Toll-1 to activate the intracellular Toll-1 pathway for nuclear translocation of the NF-κB-like transcription factor Dorsal, which induces expression of the pro-apoptotic genes reaper and hid. This activity contributes to the feedback amplification loop that operates in undead cells. Given that Toll-9 also functions in loser cells during cell competition, we define a general role of Toll-9 in cellular stress situations leading to the expression of pro-apoptotic genes that trigger apoptosis and apoptosis-induced processes such as AiP. This work identifies conceptual similarities between cell competition and AiP.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Apoptose/genética , Proliferação de Células , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Retroalimentação , Mamíferos/metabolismo
15.
Dev Growth Differ ; 52(9): 771-83, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21158756

RESUMO

The Drosophila genome encodes a total of nine Toll and related proteins. The immune and developmental functions of Toll and 18Wheeler (18W) have been analyzed extensively, while the in vivo functions of the other Toll-related proteins require further investigation. We performed transgenic experiments and found that overexpression of Toll-related genes caused different extents of lethality and developmental defects. Moreover, 18w, Toll-6, Toll-7 and Toll-8 often caused related phenotypic changes, consistent with the idea that these four genes have more conserved molecular structure and thus may regulate similar processes in vivo. Deletion alleles of Toll-6, Toll-7 and Toll-8 were generated by targeted homologous recombination or P element excision. These mutant alleles were viable, fertile, and had no detectable defect in the inducible expression of antimicrobial peptide genes except for the Toll-8 mutant had some defects in leg development. The expression of 18w, Toll-7 and Toll-8 mRNA showed wide and overlapping patterns in imaginal discs and the 18w, Toll-8 double and Toll-7, Toll-8 double mutants showed substantially increased lethality. Overall our results suggest that some of the Toll-related proteins, such as 18W, Toll-7 and Toll-8, may have redundant functions in regulating developmental processes.


Assuntos
Drosophila melanogaster/genética , Genes de Insetos/genética , Receptores Toll-Like/genética , Alelos , Animais , Drosophila melanogaster/crescimento & desenvolvimento , Mutação , Fenótipo
16.
Mol Cell Biol ; 27(12): 4578-88, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17438142

RESUMO

The inducible expression of antimicrobial peptide genes in Drosophila melanogaster is regulated by the conserved Toll and peptidoglycan recognition protein LC/immune deficiency (PGRP-LC/IMD) signaling pathways. It has been proposed that the two pathways have independent functions and mediate the specificity of innate immune responses towards different microorganisms. Scattered evidence also suggests that some antimicrobial target genes can be activated by both Toll and IMD, albeit to different extents. This dual activation can be mediated by independent stimulation or by cross-regulation of the two pathways. We show in this report that the Toll and IMD pathways can interact synergistically, demonstrating that cross-regulation occurs. The presence of Spätzle (the Toll ligand) and gram-negative peptidoglycan (the PGRP-LC ligand) together caused synergistic activation of representative target genes of the two pathways, including Drosomycin, Diptericin, and AttacinA. Constitutive activation of Toll and PGRP-LC/IMD could mimic the synergistic stimulation. RNA interference assays and promoter analyses demonstrate that cooperation of different NF-kappaB-related transcription factors mediates the synergy. These results illustrate how specific ligand binding by separate upstream pattern recognition receptors can be translated into a broad-spectrum host response, a hallmark of innate immunity.


Assuntos
Proteínas de Transporte/imunologia , Drosophila melanogaster/imunologia , Imunidade Inata , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia , Animais , Animais Geneticamente Modificados , Proteínas de Transporte/metabolismo , Células Cultivadas , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Masculino , Receptores Toll-Like/metabolismo
17.
Stem Cell Reports ; 14(5): 828-844, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32330445

RESUMO

Tight junctions in mammals and septate junctions in insects are essential for epithelial integrity. We show here that, in the Drosophila intestine, smooth septate junction proteins provide barrier and signaling functions. During an RNAi screen for genes that regulate adult midgut tissue growth, we found that loss of two smooth septate junction components, Snakeskin and Mesh, caused a hyperproliferation phenotype. By examining epitope-tagged endogenous Snakeskin and Mesh, we demonstrate that the two proteins are present in the cytoplasm of differentiating enteroblasts and in cytoplasm and septate junctions of mature enterocytes. In both enteroblasts and enterocytes, loss of Snakeskin and Mesh causes Yorkie-dependent expression of the JAK-STAT pathway ligand Upd3, which in turn promotes proliferation of intestinal stem cells. Snakeskin and Mesh form a complex with each other, with other septate junction proteins and with Yorkie. Therefore, the Snakeskin-Mesh complex has both barrier and signaling function to maintain stem cell-mediated tissue homeostasis.


Assuntos
Proteínas de Drosophila/metabolismo , Enterócitos/metabolismo , Junções Comunicantes/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Nucleares/metabolismo , Transativadores/metabolismo , Animais , Diferenciação Celular , Autorrenovação Celular , Citoplasma/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster , Enterócitos/citologia , Homeostase , Janus Quinases/metabolismo , Proteínas de Membrana/genética , Proteínas Nucleares/genética , Fatores de Transcrição STAT/metabolismo , Transativadores/genética , Proteínas de Sinalização YAP
18.
Cell Stem Cell ; 26(5): 675-692.e8, 2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32259481

RESUMO

Intestinal homeostasis is tightly regulated by complex yet poorly understood signaling networks. Here, we demonstrate that Lats1/2, the core Hippo kinases, are essential to maintain Wnt pathway activity and intestinal stem cells. Lats1/2 deletion leads to loss of intestinal stem cells but drives Wnt-uncoupled crypt expansion. To explore the function of downstream transcriptional enhanced associate domain (TEAD) transcription factors, we identified a selective small-molecule reversible inhibitor of TEAD auto-palmitoylation that directly occupies its lipid-binding site and inhibits TEAD-mediated transcription in vivo. Combining this chemical tool with genetic and proteomics approaches, we show that intestinal Wnt inhibition by Lats deletion is Yes-associated protein (YAP)/transcriptional activator with PDZ-binding domain (TAZ) dependent but TEAD independent. Mechanistically, nuclear YAP/TAZ interact with Groucho/Transducin-Like Enhancer of Split (TLE) to block Wnt/T-cell factor (TCF)-mediated transcription, and dual inhibition of TEAD and Lats suppresses Wnt-uncoupled Myc upregulation and epithelial over-proliferation in Adenomatous polyposis coli (APC)-mutated intestine. Our studies highlight a pharmacological approach to inhibit TEAD palmitoylation and have important implications for targeting Wnt and Hippo signaling in human malignancies.


Assuntos
Neoplasias , Fatores de Transcrição , Humanos , Intestinos , Fosfoproteínas/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo
19.
J Cell Physiol ; 220(3): 664-71, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19452446

RESUMO

Stem cell-mediated tissue repair is a promising approach for many diseases. Mammalian intestine is an actively regenerating tissue such that epithelial cells are constantly shedding and underlying precursor cells are constantly replenishing the loss of cells. An imbalance of these processes will lead to intestinal diseases including inflammation and cancer. Mammalian intestinal stem cells (ISCs) are located in bases of crypts but at least two groups of cells have been cited as stem cells. Moreover, precursor cells in the transit amplifying zone can also proliferate. The involvement of multiple cell types makes it more difficult to examine tissue damage response in mammalian intestine. In adult Drosophila midgut, the ISCs are the only cells that can go through mitosis. By feeding pathogenic bacteria and stress inducing chemicals to adult flies, we demonstrate that Drosophila ISCs in the midgut can respond by increasing their division. The resulting enteroblasts, precursor cells for enterocytes and enteroendocrine cells, also differentiate faster to become cells resembling enterocyte lineage. These results are consistent with the idea that Drosophila midgut stem cells can respond to tissue damage induced by pathogens and initiate tissue repair. This system should allow molecular and genetic analyses of stem cell-mediated tissue repair.


Assuntos
Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Enterócitos/patologia , Intestinos/patologia , Regeneração , Células-Tronco/patologia , Animais , Animais Geneticamente Modificados , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Drosophila , Enterócitos/efeitos dos fármacos , Enterócitos/microbiologia , Proteínas de Fluorescência Verde/genética , Peróxido de Hidrogênio/toxicidade , Intestinos/efeitos dos fármacos , Intestinos/microbiologia , Oxidantes/toxicidade , Estresse Oxidativo , Paraquat/toxicidade , Fenótipo , Pseudomonas/patogenicidade , Regeneração/efeitos dos fármacos , Serratia marcescens/patogenicidade , Células-Tronco/efeitos dos fármacos , Células-Tronco/microbiologia , Fatores de Tempo
20.
Cell Rep ; 29(10): 3200-3211.e4, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31801083

RESUMO

Uveal melanoma (UM), the most common ocular malignancy, is characterized by GNAQ/11 mutations. Hippo/YAP and Ras/mitogen-activated protein kinase (MAPK) emerge as two important signaling pathways downstream of G protein alpha subunits of the Q class (GαQ/11)-mediated transformation, although whether and how they contribute to UM genesis in vivo remain unclear. Here, we adapt an adeno-associated virus (AAV)-based ocular injection method to directly deliver Cre recombinase into the mouse uveal tract and demonstrate that Lats1/2 kinases suppress UM formation specifically in uveal melanocytes. We find that genetic activation of YAP, but not Kras, is sufficient to initiate UM. We show that YAP/TAZ activation induced by Lats1/2 deletion cooperates with Kras to promote UM progression via downstream transcriptional reinforcement. Furthermore, dual inhibition of YAP/TAZ and Ras/MAPK synergizes to suppress oncogenic growth of human UM cells. Our data highlight the functional significance of Lats-YAP/TAZ in UM initiation and progression in vivo and suggest combination inhibition of YAP/TAZ and Ras/MAPK as a new therapeutic strategy for UM.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas de Ciclo Celular/genética , Melanoma/genética , Melanoma/patologia , Transativadores/genética , Fatores de Transcrição/genética , Neoplasias Uveais/genética , Neoplasias Uveais/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Progressão da Doença , Feminino , Células HEK293 , Humanos , Melanócitos/patologia , Camundongos , Proteínas Quinases Ativadas por Mitógeno/genética , Mutação/genética , Transdução de Sinais/genética , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Proteínas de Sinalização YAP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA