Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Proc Natl Acad Sci U S A ; 112(30): E4085-93, 2015 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-26170296

RESUMO

Alcohol abuse is highly prevalent, but little is understood about the molecular causes. Here, we report that Ras suppressor 1 (Rsu1) affects ethanol consumption in flies and humans. Drosophila lacking Rsu1 show reduced sensitivity to ethanol-induced sedation. We show that Rsu1 is required in the adult nervous system for normal sensitivity and that it acts downstream of the integrin cell adhesion molecule and upstream of the Ras-related C3 botulinum toxin substrate 1 (Rac1) GTPase to regulate the actin cytoskeleton. In an ethanol preference assay, global loss of Rsu1 causes high naïve preference. In contrast, flies lacking Rsu1 only in the mushroom bodies of the brain show normal naïve preference but then fail to acquire ethanol preference like normal flies. Rsu1 is, thus, required in distinct neurons to modulate naïve and acquired ethanol preference. In humans, we find that polymorphisms in RSU1 are associated with brain activation in the ventral striatum during reward anticipation in adolescents and alcohol consumption in both adolescents and adults. Together, these data suggest a conserved role for integrin/Rsu1/Rac1/actin signaling in modulating reward-related phenotypes, including ethanol consumption, across phyla.


Assuntos
Consumo de Bebidas Alcoólicas/genética , Proteínas de Drosophila/fisiologia , Fatores de Transcrição/fisiologia , Actinas/metabolismo , Adolescente , Adulto , Animais , Encéfalo/metabolismo , Moléculas de Adesão Celular/metabolismo , Criança , Estudos de Coortes , Citoesqueleto/metabolismo , Proteínas de Drosophila/genética , Etanol/química , Feminino , GTP Fosfo-Hidrolases/metabolismo , Genes Dominantes , Humanos , Integrinas/metabolismo , Masculino , Mutação , Neurônios/metabolismo , Polimorfismo Genético , Inquéritos e Questionários , Fatores de Transcrição/genética
2.
PLoS Genet ; 9(3): e1003406, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23555310

RESUMO

A variety of human diseases arise from mutations that alter muscle contraction. Evolutionary conservation allows genetic studies in Drosophila melanogaster to be used to better understand these myopathies and suggest novel therapeutic strategies. Integrin-mediated adhesion is required to support muscle structure and function, and expression of Integrin adhesive complex (IAC) proteins is modulated to adapt to varying levels of mechanical stress within muscle. Mutations in flapwing (flw), a catalytic subunit of myosin phosphatase, result in non-muscle myosin hyperphosphorylation, as well as muscle hypercontraction, defects in size, motility, muscle attachment, and subsequent larval and pupal lethality. We find that moderately elevated expression of the IAC protein PINCH significantly rescues flw phenotypes. Rescue requires PINCH be bound to its partners, Integrin-linked kinase and Ras suppressor 1. Rescue is not achieved through dephosphorylation of non-muscle myosin, suggesting a mechanism in which elevated PINCH expression strengthens integrin adhesion. In support of this, elevated expression of PINCH rescues an independent muscle hypercontraction mutant in muscle myosin heavy chain, Mhc(Samba1). By testing a panel of IAC proteins, we show specificity for PINCH expression in the rescue of hypercontraction mutants. These data are consistent with a model in which PINCH is present in limiting quantities within IACs, with increasing PINCH expression reinforcing existing adhesions or allowing for the de novo assembly of new adhesion complexes. Moreover, in myopathies that exhibit hypercontraction, strategic PINCH expression may have therapeutic potential in preserving muscle structure and function.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Contração Muscular , Doenças Musculares , Fatores de Transcrição , Animais , Adesão Celular/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica , Humanos , Integrinas/genética , Integrinas/metabolismo , Contração Muscular/genética , Contração Muscular/fisiologia , Doenças Musculares/genética , Doenças Musculares/fisiopatologia , Mutação , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
3.
J Cell Sci ; 125(Pt 13): 3185-94, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22467865

RESUMO

PINCH, integrin-linked kinase (ILK) and Ras suppressor-1 (RSU-1) are molecular scaffolding proteins that form a physical complex downstream of integrins, and have overlapping roles in cellular adhesion. In Drosophila, PINCH and ILK colocalize in cells and have indistinguishable functions in maintaining wing adhesion and integrin to actin linkage in the muscle. We sought to determine whether the direct physical interaction between PINCH and ILK was essential for their functions using transgenic flies expressing a version of PINCH with a point mutation that disrupts ILK binding (PINCH(Q38A)). We demonstrate that the PINCH-ILK interaction is not required for viability, for integrin-mediated adhesion of the wing or muscle, or for maintaining appropriate localization or levels of either PINCH or ILK. These results suggest alternative modes for PINCH localization, stabilization and linkage to the actin cytoskeleton that are independent of a direct interaction with ILK. Furthermore, we identified a synthetic lethality in flies carrying both the PINCH(Q38A) mutation and a null mutation in the gene encoding RSU-1. This lethality does not result from PINCH mislocalization or destabilization, and illustrates a novel compensatory role for RSU-1 in maintaining viability in flies with compromised PINCH-ILK binding. Taken together, this work highlights the existence of redundant mechanisms in adhesion complex assembly that support integrin function in vivo.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição/metabolismo , Animais , Animais Geneticamente Modificados/embriologia , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/metabolismo , Western Blotting , Cruzamentos Genéticos , Drosophila/enzimologia , Drosophila/genética , Proteínas de Drosophila/genética , Embrião não Mamífero/metabolismo , Embrião não Mamífero/patologia , Feminino , Integrinas/genética , Integrinas/metabolismo , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Músculos/citologia , Músculos/metabolismo , Mutação Puntual , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Estabilidade Proteica , Fatores de Transcrição/genética , Asas de Animais/citologia , Asas de Animais/metabolismo
4.
Proteome Sci ; 11(1): 21, 2013 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-23663728

RESUMO

BACKGROUND: Here we describe a novel approach used to identify the constituents of protein complexes with high fidelity, using the integrin-associated scaffolding protein PINCH as a test case. PINCH is comprised of five LIM domains, zinc-finger protein interaction modules. In Drosophila melanogaster, PINCH has two known high-affinity binding partners-Integrin-linked kinase (ILK) that binds to LIM1 and Ras Suppressor 1 (RSU1) that binds to LIM5-but has been postulated to bind additional proteins as well. RESULTS: To purify PINCH complexes, in parallel we fused different affinity tags (Protein A and Flag) to different locations within the PINCH sequence (N- and C-terminus). We expressed these tagged versions of PINCH both in cell culture (overexpressed in Drosophila S2 cell culture in the presence of endogenous PINCH) and in vivo (at native levels in Drosophila lacking endogenous PINCH). After affinity purification, we analyzed PINCH complexes by a novel 2D-gel electrophoresis analysis, iGEO (interactions by 2D Gel Electrophoresis Overlap), with mass spectrometric identification of individual spots of interest. iGEO allowed the identification of protein partners that associate with PINCH under two independent purification strategies, providing confidence in the significance of the interaction. Proteins identified by iGEO were validated against a highly inclusive list of candidate PINCH interacting proteins identified in previous analyses by MuDPIT mass spectrometry. CONCLUSIONS: The iGEO strategy confirmed a core complex comprised of PINCH, RSU1, ILK, and ILK binding partner Parvin. Our iGEO method also identified five novel protein partners that specifically interacted with PINCH in Drosophila S2 cell culture. Because of the improved reproducibility of 2D-GE methodology and the increasing affordability of the required labeling reagents, iGEO is a method that is accessible to most moderately well-equipped biological laboratories. The biochemical co-purifications inherent in iGEO allow for rapid and unambiguous identification of the constituents of protein complexes, without the need for extensive follow-up experiments.

5.
Sci Rep ; 10(1): 19303, 2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-33168840

RESUMO

Platelet Derived Growth Factor Receptor (PDGFR) signaling is a central mitogenic pathway in development, as well as tissue repair and homeostasis. The rules governing the binding of PDGF ligand to the receptor to produce activation and downstream signaling have been well defined over the last several decades. In cultured cells after a period of serum deprivation, treatment with PDGF leads to the rapid formation of dramatic, actin-rich Circular Dorsal Ruffles (CDRs). Using CDRs as a robust visual readout of early PDGFR signaling, we have identified several contradictory elements in the widely accepted model of PDGF activity. Employing CRISPR/Cas9 gene editing to disrupt the Pdgfra gene in two different murine cell lines, we show that in addition to the widely accepted function for PDGFR-beta in CDR formation, PDGFR-alpha is also clearly capable of eliciting CDRs. Moreover, we demonstrate activity for heterodimeric PDGF-AB ligand in the vigorous activation of PDGFR-beta homodimers to produce CDRs. These findings are key to a more complete understanding of PDGF ligand-receptor interactions and their downstream signaling consequences. This knowledge will allow for more rigorous experimental design in future studies of PDGFR signaling and its contributions to development and disease.


Assuntos
Becaplermina/metabolismo , Fibroblastos/metabolismo , Melanoma/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Ligantes , Melanoma/genética , Camundongos , Ligação Proteica , Multimerização Proteica , Transdução de Sinais
6.
J Cell Biol ; 167(6): 1019-24, 2004 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-15596544

RESUMO

Cell adhesion and migration are dynamic processes requiring the coordinated action of multiple signaling pathways, but the mechanisms underlying signal integration have remained elusive. Drosophila embryonic dorsal closure (DC) requires both integrin function and c-Jun amino-terminal kinase (JNK) signaling for opposed epithelial sheets to migrate, meet, and suture. Here, we show that PINCH, a protein required for integrin-dependent cell adhesion and actin-membrane anchorage, is present at the leading edge of these migrating epithelia and is required for DC. By analysis of native protein complexes, we identify RSU-1, a regulator of Ras signaling in mammalian cells, as a novel PINCH binding partner that contributes to PINCH stability. Mutation of the gene encoding RSU-1 results in wing blistering in Drosophila, demonstrating its role in integrin-dependent cell adhesion. Genetic interaction analyses reveal that both PINCH and RSU-1 antagonize JNK signaling during DC. Our results suggest that PINCH and RSU-1 contribute to the integration of JNK and integrin functions during Drosophila development.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Proteínas de Drosophila/metabolismo , Epitélio/fisiologia , Integrinas/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Actinas/fisiologia , Animais , Animais Geneticamente Modificados , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Proteínas de Ligação a DNA/genética , Drosophila/embriologia , Drosophila/enzimologia , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Células Epiteliais/fisiologia , Epitélio/embriologia , Integrinas/metabolismo , Mutação , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética , Asas de Animais/fisiologia
7.
Curr Biol ; 25(17): 2215-27, 2015 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-26255846

RESUMO

Integrins are critical for barrier epithelial architecture. Integrin loss in vertebrate skin leads to blistering and wound healing defects. However, how integrins and associated proteins maintain the regular morphology of epithelia is not well understood. We found that targeted knockdown of the integrin focal adhesion (FA) complex components ß-integrin, PINCH, and integrin-linked kinase (ILK) caused formation of multinucleate epidermal cells within the Drosophila larval epidermis. This phenotype was specific to the integrin FA complex and not due to secondary effects on polarity or junctional structures. The multinucleate cells resembled the syncytia caused by physical wounding. Live imaging of wound-induced syncytium formation in the pupal epidermis suggested direct membrane breakdown leading to cell-cell fusion and consequent mixing of cytoplasmic contents. Activation of Jun N-terminal kinase (JNK) signaling, which occurs upon wounding, also correlated with syncytium formation induced by PINCH knockdown. Further, ectopic JNK activation directly caused epidermal syncytium formation. No mode of syncytium formation, including that induced by wounding, genetic loss of FA proteins, or local JNK hyperactivation, involved misregulation of mitosis or apoptosis. Finally, the mechanism of epidermal syncytium formation following JNK hyperactivation and wounding appeared to be direct disassembly of FA complexes. In conclusion, the loss-of-function phenotype of integrin FA components in the larval epidermis resembles a wound. Integrin FA loss in mouse and human skin also causes a wound-like appearance. Our results reveal a novel and unexpected role for proper integrin-based adhesion in suppressing larval epidermal cell-cell fusion--a role that may be conserved in other epithelia.


Assuntos
Proteínas de Drosophila/genética , Drosophila/genética , Cadeias beta de Integrinas/genética , Proteínas Serina-Treonina Quinases/genética , Fatores de Transcrição/genética , Animais , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Epiderme/crescimento & desenvolvimento , Epiderme/metabolismo , Técnicas de Silenciamento de Genes , Células Gigantes/metabolismo , Cadeias beta de Integrinas/metabolismo , Larva/crescimento & desenvolvimento , Larva/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Pupa/crescimento & desenvolvimento , Pupa/metabolismo , Fatores de Transcrição/metabolismo
8.
Cytoskeleton (Hoboken) ; 70(6): 304-16, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23606669

RESUMO

Stabilization of tissue architecture during development and growth is essential to maintain structural integrity. Because of its contractile nature, muscle is especially susceptible to physiological stresses, and has multiple mechanisms to maintain structural integrity. The Drosophila melanogaster Muscle LIM Protein (MLP), Mlp84B, participates in muscle maintenance, yet its precise mechanism of action is still controversial. Through a candidate approach, we identified α-actinin as a protein that functions with Mlp84B to ensure muscle integrity. α-actinin RNAi animals die primarily as pupae, and Mlp84B RNAi animals are adult viable. RNAi knockdown of Mlp84B and α-actinin together produces synergistic early larval lethality and destabilization of Z-line structures. We recapitulated these phenotypes using combinations of traditional loss-of-function alleles and single-gene RNAi. We observe that Mlp84B induces the formation of actin loops in muscle cell nuclei in the absence of nuclear α-actinin, suggesting Mlp84B has intrinsic actin cross-linking activity, which may complement α-actinin cross-linking activity at sites of actin filament anchorage. These results reveal a molecular mechanism for MLP stabilization of muscle and implicate reduced actin crosslinking as the primary destabilizing defect in MLP-associated cardiomyopathies. Our data support a model in which α-actinin and Mlp84B have important and overlapping functions at sites of actin filament anchorage to preserve muscle structure and function.


Assuntos
Actinina/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Proteínas com Domínio LIM/metabolismo , Proteínas Musculares/metabolismo , Actinina/genética , Animais , Western Blotting , Núcleo Celular , Proteínas de Drosophila/genética , Proteínas com Domínio LIM/genética , Larva , Proteínas dos Microfilamentos/metabolismo , Células Musculares/citologia , Células Musculares/metabolismo , Proteínas Musculares/genética
9.
Dev Dyn ; 236(8): 2207-15, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17584887

RESUMO

Receptor for Activated C Kinase 1 (RACK1) is a cytoplasmic molecular scaffolding protein. Many diverse protein-binding partners involved in key signaling pathways are reported to bind to RACK1, suggesting a role for RACK1 in signal integration. However, because loss-of-function phenotypes for RACK1 in an intact organism have not yet been reported, our current understanding of RACK1 is limited. Using Drosophila melanogaster, we show that RACK1 is expressed at all developmental stages and in many tissues, with specific enrichment in the ovary. By characterizing an allelic series of RACK1 mutants, we demonstrate that RACK1 is essential at multiple steps of Drosophila development, particularly in oogenesis, where somatic RACK1 is required for proper germ-line function.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Alelos , Animais , Proteínas de Drosophila/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Mutação , Oogênese , Ovário/química , Receptores de Quinase C Ativada , Receptores Citoplasmáticos e Nucleares/genética , Distribuição Tecidual
10.
Nat Rev Mol Cell Biol ; 5(11): 920-31, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15520811

RESUMO

First described 15 years ago as a cysteine-rich sequence that was common to a small group of homeodomain transcription factors, the LIM domain is now recognized as a tandem zinc-finger structure that functions as a modular protein-binding interface. LIM domains are present in many proteins that have diverse cellular roles as regulators of gene expression, cytoarchitecture, cell adhesion, cell motility and signal transduction. An emerging theme is that LIM proteins might function as biosensors that mediate communication between the cytosolic and the nuclear compartments.


Assuntos
Núcleo Celular/metabolismo , Citoesqueleto/química , Animais , Técnicas Biossensoriais , Cisteína/química , Citoesqueleto/metabolismo , Humanos , Modelos Biológicos , Modelos Moleculares , Neurônios/metabolismo , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA