Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38585873

RESUMO

Lysosomal storage diseases (LSDs) comprised ~50 monogenic diseases characterized by the accumulation of cellular material in lysosomes and associated defects in lysosomal function, but systematic molecular phenotyping is lacking. Here, we develop a nanoflow-based multi-omic single-shot technology (nMOST) workflow allowing simultaneously quantify HeLa cell proteomes and lipidomes from more than two dozen LSD mutants, revealing diverse molecular phenotypes. Defects in delivery of ferritin and its autophagic receptor NCOA4 to lysosomes (ferritinophagy) were pronounced in NPC2-/- cells, which correlated with increased lyso-phosphatidylcholine species and multi-lamellar membrane structures visualized by cryo-electron-tomography. Ferritinophagy defects correlated with loss of mitochondrial cristae, MICOS-complex components, and electron transport chain complexes rich in iron-sulfur cluster proteins. Strikingly, mitochondrial defects were alleviated when iron was provided through the transferrin system. This resource reveals how defects in lysosomal function can impact mitochondrial homeostasis in trans and highlights nMOST as a discovery tool for illuminating molecular phenotypes across LSDs.

2.
Nat Commun ; 13(1): 5924, 2022 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-36207292

RESUMO

Haploinsufficiency of GRN causes frontotemporal dementia (FTD). The GRN locus produces progranulin (PGRN), which is cleaved to lysosomal granulin polypeptides. The function of lysosomal granulins and why their absence causes neurodegeneration are unclear. Here we discover that PGRN-deficient human cells and murine brains, as well as human frontal lobes from GRN-mutation FTD patients have increased levels of gangliosides, glycosphingolipids that contain sialic acid. In these cells and tissues, levels of lysosomal enzymes that catabolize gangliosides were normal, but levels of bis(monoacylglycero)phosphates (BMP), lipids required for ganglioside catabolism, were reduced with PGRN deficiency. Our findings indicate that granulins are required to maintain BMP levels to support ganglioside catabolism, and that PGRN deficiency in lysosomes leads to gangliosidosis. Lysosomal ganglioside accumulation may contribute to neuroinflammation and neurodegeneration susceptibility observed in FTD due to PGRN deficiency and other neurodegenerative diseases.


Assuntos
Demência Frontotemporal , Gangliosidoses , Progranulinas/metabolismo , Animais , Demência Frontotemporal/genética , Demência Frontotemporal/metabolismo , Gangliosídeos/metabolismo , Gangliosidoses/metabolismo , Granulinas/metabolismo , Humanos , Lisossomos/metabolismo , Camundongos , Ácido N-Acetilneuramínico/metabolismo , Fosfatos/metabolismo , Progranulinas/genética
3.
Cell ; 166(2): 408-423, 2016 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-27419871

RESUMO

VAP (VAPA and VAPB) is an evolutionarily conserved endoplasmic reticulum (ER)-anchored protein that helps generate tethers between the ER and other membranes through which lipids are exchanged across adjacent bilayers. Here, we report that by regulating PI4P levels on endosomes, VAP affects WASH-dependent actin nucleation on these organelles and the function of the retromer, a protein coat responsible for endosome-to-Golgi traffic. VAP is recruited to retromer budding sites on endosomes via an interaction with the retromer SNX2 subunit. Cells lacking VAP accumulate high levels of PI4P, actin comets, and trans-Golgi proteins on endosomes. Such defects are mimicked by downregulation of OSBP, a VAP interactor and PI4P transporter that participates in VAP-dependent ER-endosomes tethers. These results reveal a role of PI4P in retromer-/WASH-dependent budding from endosomes. Collectively, our data show how the ER can control budding dynamics and association with the cytoskeleton of another membrane by direct contacts leading to bilayer lipid modifications.


Assuntos
Endossomos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Actinas/metabolismo , Retículo Endoplasmático/metabolismo , Técnicas de Inativação de Genes , Células HeLa , Humanos , Proteínas dos Microfilamentos/metabolismo , Receptores de Esteroides/metabolismo , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição , Proteínas de Transporte Vesicular/genética
4.
Development ; 142(8): 1502-15, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25852200

RESUMO

Evolutionarily conserved intercellular signaling pathways regulate embryonic development and adult tissue homeostasis in metazoans. The precise control of the state and amplitude of signaling pathways is achieved in part through the kinase- and phosphatase-mediated reversible phosphorylation of proteins. In this study, we performed a genome-wide in vivo RNAi screen for kinases and phosphatases that regulate the Wnt pathway under physiological conditions in the Drosophila wing disc. Our analyses have identified 54 high-confidence kinases and phosphatases capable of modulating the Wnt pathway, including 22 novel regulators. These candidates were also assayed for a role in the Notch pathway, and numerous phospho-regulators were identified. Additionally, each regulator of the Wnt pathway was evaluated in the wing disc for its ability to affect the mechanistically similar Hedgehog pathway. We identified 29 dual regulators that have the same effect on the Wnt and Hedgehog pathways. As proof of principle, we established that Cdc37 and Gilgamesh/CK1γ inhibit and promote signaling, respectively, by functioning at analogous levels of these pathways in both Drosophila and mammalian cells. The Wnt and Hedgehog pathways function in tandem in multiple developmental contexts, and the identification of several shared phospho-regulators serve as potential nodes of control under conditions of aberrant signaling and disease.


Assuntos
Proteínas de Drosophila/metabolismo , Proteínas Wnt/metabolismo , Animais , Caseína Quinase I/genética , Caseína Quinase I/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Drosophila , Proteínas de Drosophila/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Receptor Smoothened , Asas de Animais/metabolismo , Proteínas Wnt/genética , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
5.
Fly (Austin) ; 6(2): 126-31, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22634475

RESUMO

The Wnt/Wingless (Wg) pathway is an evolutionarily conserved signaling system that is used reiteratively, both spatially and temporally, to control the development of multicellular animals. The stability of cytoplasmic ß-catenin/Armadillo, the transcriptional effector of the pathway, is controlled by sequential N-terminal phosphorylation and ubiquitination that targets it for proteasome-mediated degradation. Orthologous members of the Homeodomain-interacting protein kinase family from Drosophila to vertebrates have been implicated in the regulation of Wnt/Wingless signaling. In Drosophila, as a consequence of Hipk activity, cells accumulate stabilized Armadillo that directs the expression of Wg-specific target genes. Hipk promotes the stabilization of Armadillo by inhibiting its ubiquitination (and hence subsequent degradation) by the SCF(Slimb) E3 ubiquitin ligase complex. Vertebrate Hipk2 impedes ß-catenin ubiquitination to promote its stability and the Wnt signal in a mechanism that is functionally conserved. Moreover, we describe here that Hipk proteins have a role independent of their effect on ß-catenin/Armadillo stability to enhance Wnt/Wingless signaling.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , Proteína Wnt1/metabolismo , Animais , Proteínas do Domínio Armadillo/metabolismo , Proteínas de Ciclo Celular/metabolismo , Drosophila/crescimento & desenvolvimento , Proteínas Hedgehog/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Contendo Repetições de beta-Transducina/metabolismo
6.
Artigo em Inglês | MEDLINE | ID: mdl-22535229

RESUMO

The Wingless (Wg) pathway represents one of the best-characterized intercellular signaling networks. Studies performed in Drosophila over the last 30 years have contributed to our understanding of the role of Wg signaling in the regulation of tissue growth, polarity, and patterning. These studies have revealed mechanisms conserved in the vertebrate Wnt pathways and illustrate the elegance of using the Drosophila model to understand evolutionarily conserved modes of gene regulation. In this article, we describe the function of Wg signaling in patterning the Drosophila embryonic epidermis and wing imaginal disc. As well, we present an overview of the establishment of the Wg morphogen gradient and discuss the differential modes of Wg-regulated gene expression.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , Proteína Wnt1/metabolismo , Animais , Modelos Teóricos
7.
Proc Natl Acad Sci U S A ; 108(24): 9887-92, 2011 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-21628596

RESUMO

Drosophila Homeodomain-interacting protein kinase (Hipk) has been shown to regulate in vivo, the stability of Armadillo, the transcriptional effector of Wingless signaling. The Wingless pathway culminates in the stabilization of Armadillo that, in the absence of signaling, is sequentially phosphorylated, polyubiquitinated and degraded. Loss-of-function clones for hipk result in reduced stabilized Armadillo, whereas overexpression of hipk elevates Armadillo levels to promote Wingless-responsive target gene expression. Here, we show that overexpression of hipk can suppress the effects of negative regulators of Armadillo to prevent its degradation in the wing imaginal disc. Hipk acts to stabilize Armadillo by impeding the function of the E3 ubiquitin ligase Skp1-Cul1-F-box (SCF)(Slimb), thereby inhibiting Armadillo ubiquitination and subsequent degradation. Vertebrate Hipk2 displays a similar ability to prevent ß-catenin ubiquitination in a functionally conserved mechanism. We find that Hipk's ability to inhibit SCF(Slimb)-mediated ubiquitination is not restricted to Armadillo and extends to other substrates of SCF(Slimb), including the Hedgehog signaling effector Ci. Thus, similar to casein kinase 1 and glycogen synthase kinase 3, Hipk dually regulates both Wingless and Hedgehog signaling by controlling the stability of their respective signaling effectors, but it is the first kinase to our knowledge identified that promotes the stability of both Armadillo and Ci.


Assuntos
Proteínas de Drosophila/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas Quinases/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Transdução de Sinais , Proteína Wnt1/metabolismo , Animais , Animais Geneticamente Modificados , Proteínas do Domínio Armadillo/genética , Proteínas do Domínio Armadillo/metabolismo , Western Blotting , Células COS , Linhagem Celular , Chlorocebus aethiops , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Feminino , Células HEK293 , Proteínas Hedgehog/genética , Humanos , Imunoprecipitação , Masculino , Fosforilação , Ligação Proteica , Proteínas Quinases/genética , Proteínas Ligases SKP Culina F-Box/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção , Ubiquitinação , Asas de Animais/metabolismo , Proteína Wnt1/genética
8.
Development ; 136(2): 241-51, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19088090

RESUMO

The Wnt/Wingless (Wg) pathway represents a conserved signaling cascade involved in diverse biological processes. Misregulation of Wnt/Wg signal transduction has profound effects on development. Homeodomain-interacting protein kinases (Hipks) represent a novel family of serine/threonine kinases. Members of this group (in particular Hipk2) are implicated as important factors in transcriptional regulation to control cell growth, apoptosis and development. Here, we provide genetic and phenotypic evidence that the sole Drosophila member of this family, Hipk, functions as a positive regulator in the Wg pathway. Expression of hipk in the wing rescues loss of the Wg signal, whereas loss of hipk can enhance decreased wg signaling phenotypes. Furthermore, loss of hipk leads to diminished Arm protein levels, whereas overexpression of hipk promotes the Wg signal by stabilizing Arm, resulting in activation of Wg responsive targets. In Wg transcriptional assays, Hipk enhanced Tcf/Arm-mediated gene expression in a kinase-dependent manner. In addition, Hipk can bind to Arm and Drosophila Tcf, and phosphorylate Arm. Using both in vitro and in vivo assays, Hipk was found to promote the stabilization of Arm. We observe similar molecular interactions between Lef1/beta-catenin and vertebrate Hipk2, suggesting a direct and conserved role for Hipk proteins in promoting Wnt signaling.


Assuntos
Proteínas do Domínio Armadillo/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt1/metabolismo , Animais , Animais Geneticamente Modificados , Padronização Corporal/genética , Padronização Corporal/fisiologia , Drosophila/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes de Insetos , Masculino , Modelos Biológicos , Complexos Multiproteicos , Fosforilação , Interferência de RNA , Proteínas Repressoras/metabolismo , Transdução de Sinais , Asas de Animais/crescimento & desenvolvimento , Asas de Animais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA