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










Base de dados
Intervalo de ano de publicação
1.
Sci Adv ; 9(47): eadh9673, 2023 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-38000028

RESUMO

The mammalian intestine is one of the most rapidly self-renewing tissues, driven by stem cells residing at the crypt bottom. Paneth cells form a major element of the niche microenvironment providing various growth factors to orchestrate intestinal stem cell homeostasis, such as Wnt3. Different Wnt ligands can selectively activate ß-catenin-dependent (canonical) or -independent (noncanonical) signaling. Here, we report that the Dishevelled-associated activator of morphogenesis 1 (Daam1) and its paralogue Daam2 asymmetrically regulate canonical and noncanonical Wnt (Wnt/PCP) signaling. Daam1/2 interacts with the Wnt inhibitor RNF43, and Daam1/2 double knockout stimulates canonical Wnt signaling by preventing RNF43-dependent degradation of the Wnt receptor, Frizzled (Fzd). Single-cell RNA sequencing analysis revealed that Paneth cell differentiation is impaired by Daam1/2 depletion because of defective Wnt/PCP signaling. Together, we identified Daam1/2 as an unexpected hub molecule coordinating both canonical and noncanonical Wnt, which is fundamental for specifying an adequate number of Paneth cells.


Assuntos
Celulas de Paneth , Via de Sinalização Wnt , Animais , Intestinos , Diferenciação Celular , Células-Tronco/metabolismo , Mamíferos
2.
Life Sci Alliance ; 6(11)2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37591722

RESUMO

Cancer cells make extensive use of the folate cycle to sustain increased anabolic metabolism. Multiple chemotherapeutic drugs interfere with the folate cycle, including methotrexate and 5-fluorouracil that are commonly applied for the treatment of leukemia and colorectal cancer (CRC), respectively. Despite high success rates, therapy-induced resistance causes relapse at later disease stages. Depletion of folylpolyglutamate synthetase (FPGS), which normally promotes intracellular accumulation and activity of natural folates and methotrexate, is linked to methotrexate and 5-fluorouracil resistance and its association with relapse illustrates the need for improved intervention strategies. Here, we describe a novel antifolate (C1) that, like methotrexate, potently inhibits dihydrofolate reductase and downstream one-carbon metabolism. Contrary to methotrexate, C1 displays optimal efficacy in FPGS-deficient contexts, due to decreased competition with intracellular folates for interaction with dihydrofolate reductase. We show that FPGS-deficient patient-derived CRC organoids display enhanced sensitivity to C1, whereas FPGS-high CRC organoids are more sensitive to methotrexate. Our results argue that polyglutamylation-independent antifolates can be applied to exert selective pressure on FPGS-deficient cells during chemotherapy, using a vulnerability created by polyglutamylation deficiency.


Assuntos
Antagonistas do Ácido Fólico , Humanos , Antagonistas do Ácido Fólico/farmacologia , Metotrexato/farmacologia , Tetra-Hidrofolato Desidrogenase/genética , Ácido Fólico/farmacologia , Fluoruracila/farmacologia
3.
Am J Hum Genet ; 110(9): 1470-1481, 2023 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-37582359

RESUMO

Sclerosing skeletal dysplasias result from an imbalance between bone formation and resorption. We identified three homozygous, C-terminally truncating AXIN1 variants in seven individuals from four families affected by macrocephaly, cranial hyperostosis, and vertebral endplate sclerosis. Other frequent findings included hip dysplasia, heart malformations, variable developmental delay, and hematological anomalies. In line with AXIN1 being a central component of the ß-catenin destruction complex, analyses of primary and genome-edited cells harboring the truncating variants revealed enhanced basal canonical Wnt pathway activity. All three AXIN1-truncating variants resulted in reduced protein levels and impaired AXIN1 polymerization mediated by its C-terminal DIX domain but partially retained Wnt-inhibitory function upon overexpression. Addition of a tankyrase inhibitor attenuated Wnt overactivity in the AXIN1-mutant model systems. Our data suggest that AXIN1 coordinates the action of osteoblasts and osteoclasts and that tankyrase inhibitors can attenuate the effects of AXIN1 hypomorphic variants.


Assuntos
Luxação do Quadril , Osteosclerose , Tanquirases , Humanos , Tanquirases/genética , Tanquirases/metabolismo , Proteína Axina/genética , Proteína Axina/metabolismo , Via de Sinalização Wnt/genética , Osteosclerose/genética , beta Catenina/metabolismo
4.
J Control Release ; 356: 72-83, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36813038

RESUMO

The transmembrane receptor LGR5 potentiates Wnt/ß-catenin signaling by binding both secreted R-spondin (RSPOs) and the Wnt tumor suppressors RNF43/ZNRF3, directing clearance of RNF43/ZNRF3 from the cell surface. Besides being widely used as a stem cell marker in various tissues, LGR5 is overexpressed in many types of malignancies, including colorectal cancer. Its expression characterizes a subpopulation of cancer cells that play a crucial role in tumor initiation, progression and cancer relapse, known as cancer stem cells (CSCs). For this reason, ongoing efforts are aimed at eradicating LGR5-positive CSCs. Here, we engineered liposomes decorated with different RSPO proteins to specifically detect and target LGR5-positive cells. Using fluorescence-loaded liposomes, we show that conjugation of full-length RSPO1 to the liposomal surface mediates aspecific, LGR5-independent cellular uptake, largely mediated by heparan sulfate proteoglycan binding. By contrast, liposomes decorated only with the Furin (FuFu) domains of RSPO3 are taken up by cells in a highly specific, LGR5-dependent manner. Moreover, encapsulating doxorubicin in FuFuRSPO3 liposomes allowed us to selectively inhibit the growth of LGR5-high cells. Thus, FuFuRSPO3-coated liposomes allow for the selective detection and ablation of LGR5-high cells, providing a potential drug delivery system for LGR5-targeted anti-cancer strategies.


Assuntos
Lipossomos , Receptores Acoplados a Proteínas G , Receptores Acoplados a Proteínas G/metabolismo , Furina/metabolismo , Via de Sinalização Wnt , Sistemas de Liberação de Medicamentos , Células-Tronco Neoplásicas/metabolismo
6.
Curr Biol ; 32(14): R791-R805, 2022 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-35882203

RESUMO

The vertebrate intestine experiences a range of intrinsically generated and external forces during both development and adult homeostasis. It is increasingly understood how the coordination of these forces shapes the intestine through organ-scale folding and epithelial organization into crypt-villus compartments. Moreover, accumulating evidence shows that several cell types in the adult intestine can sense and respond to forces to regulate key cellular processes underlying adult intestinal functions and self-renewal. In this way, transduction of forces may direct both intestinal homeostasis as well as adaptation to external stimuli, such as food ingestion or injury. In this review, we will discuss recent insights from complementary model systems into the force-dependent mechanisms that establish and maintain the unique architecture of the intestine, as well as its homeostatic regulation and function throughout adult life.


Assuntos
Mucosa Intestinal , Adulto , Homeostase , Humanos , Mucosa Intestinal/metabolismo
7.
Mol Cell ; 81(16): 3241-3243, 2021 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-34416136

RESUMO

Ranes et al. (2021) report on an in vitro reconstituted ß-catenin destruction complex and elucidate the contributions of full-length and cancer-related mutated core components to ß-catenin turnover, thereby advancing our understanding of the inner workings of this tumor suppressor complex.

8.
Nat Rev Cancer ; 21(1): 5-21, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33097916

RESUMO

Mutation-induced activation of WNT-ß-catenin signalling is a frequent driver event in human cancer. Sustained WNT-ß-catenin pathway activation endows cancer cells with sustained self-renewing growth properties and is associated with therapy resistance. In healthy adult stem cells, WNT pathway activity is carefully controlled by core pathway tumour suppressors as well as negative feedback regulators. Gene inactivation experiments in mouse models unequivocally demonstrated the relevance of WNT tumour suppressor loss-of-function mutations for cancer growth. However, in human cancer, a far more complex picture has emerged in which missense or truncating mutations mediate stable expression of mutant proteins, with distinct functional and phenotypic ramifications. Herein, we review recent advances and challenges in our understanding of how different mutational subsets of WNT tumour suppressor genes link to distinct cancer types, clinical outcomes and treatment strategies.


Assuntos
Antineoplásicos/uso terapêutico , Terapia de Alvo Molecular , Mutação , Neoplasias/tratamento farmacológico , Proteínas Supressoras de Tumor/genética , Proteínas Wnt/genética , Via de Sinalização Wnt/efeitos dos fármacos , Animais , Humanos , Neoplasias/genética , Neoplasias/patologia
9.
Cell Death Differ ; 28(1): 95-107, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33208888

RESUMO

The intestinal epithelium harbors a remarkable adaptability to undergo injury-induced repair. A key part of the regenerative response is the transient reprogramming of epithelial cells into a fetal-like state, which drives uniform proliferation, tissue remodeling, and subsequent restoration of the homeostatic state. In this review, we discuss how Wnt and YAP signaling pathways control the intestinal repair response and the transitioning of cell states, in comparison with the process of intestinal development. Furthermore, we highlight how organoid-based applications have contributed to the characterization of the mechanistic principles and key players that guide these developmental and regenerative events.


Assuntos
Células Epiteliais/metabolismo , Intestinos/citologia , Organoides/metabolismo , Regeneração/fisiologia , Adulto , Animais , Homeostase , Humanos , Mucosa Intestinal/metabolismo , Intestinos/fisiologia , Organoides/citologia , Via de Sinalização Wnt , Proteínas de Sinalização YAP
11.
RNA ; 2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-33323527

RESUMO

Transition through cell cycle phases requires temporal and spatial regulation of gene expression to ensure accurate chromosome duplication and segregation. This regulation involves dynamic reprogramming of gene expression at multiple transcriptional and posttranscriptional levels. In transcriptionally silent oocytes, the CPEB-family of RNAbinding proteins coordinates temporal and spatial translation regulation of stored maternal mRNAs to drive meiotic progression. CPEB1 mediates mRNA localization to the meiotic spindle, which is required to ensure proper chromosome segregation. Temporal translational regulation also takes place in mitosis, where a large repertoire of transcripts are activated or repressed in specific cell cycle phases. However, whether control of localized translation at the spindle is required for mitosis is unclear, as mitotic and acentriolar-meiotic spindles are functionally and structurally different. Furthermore, the large differences in scale-ratio between cell volume and spindle size in oocytes compared to somatic mitotic cells may generate distinct requirements for gene expression compartmentalization in meiosis and mitosis. Here we show that mitotic spindles contain CPE-localized mRNAs and translating ribosomes. Moreover, CPEB1 and CPEB4 localize in the spindles and they may function sequentially in promoting mitotic stage transitions and correct chromosome segregation. Thus, CPEB1 and CPEB4 bind to specific spindle-associated transcripts controlling the expression and/or localization of their encoded factors that, respectively, drive metaphase and anaphase/cytokinesis.

12.
Cell Metab ; 32(5): 889-900.e7, 2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33147486

RESUMO

Differential WNT and Notch signaling regulates differentiation of Lgr5+ crypt-based columnar cells (CBCs) into intestinal cell lineages. Recently we showed that mitochondrial activity supports CBCs, while adjacent Paneth cells (PCs) show reduced mitochondrial activity. This implies that CBC differentiation into PCs involves a metabolic transition toward downregulation of mitochondrial dependency. Here we show that Forkhead box O (FoxO) transcription factors and Notch signaling interact in determining CBC fate. In agreement with the organoid data, Foxo1/3/4 deletion in mouse intestine induces secretory cell differentiation. Importantly, we show that FOXO and Notch signaling converge on regulation of mitochondrial fission, which in turn provokes stem cell differentiation into goblet cells and PCs. Finally, scRNA-seq-based reconstruction of CBC differentiation trajectories supports the role of FOXO, Notch, and mitochondria in secretory differentiation. Together, this points at a new signaling-metabolic axis in CBC differentiation and highlights the importance of mitochondria in determining stem cell fate.


Assuntos
Células Caliciformes , Intestinos/citologia , Mitocôndrias/metabolismo , Celulas de Paneth , Células-Tronco , Animais , Diferenciação Celular , Linhagem Celular , Fatores de Transcrição Forkhead/metabolismo , Células Caliciformes/citologia , Células Caliciformes/metabolismo , Camundongos , Dinâmica Mitocondrial , Celulas de Paneth/citologia , Celulas de Paneth/metabolismo , Receptores Notch/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo
13.
EMBO J ; 39(18): e103932, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32965059

RESUMO

Wnt/ß-catenin signaling is a primary pathway for stem cell maintenance during tissue renewal and a frequent target for mutations in cancer. Impaired Wnt receptor endocytosis due to loss of the ubiquitin ligase RNF43 gives rise to Wnt-hypersensitive tumors that are susceptible to anti-Wnt-based therapy. Contrary to this paradigm, we identify a class of RNF43 truncating cancer mutations that induce ß-catenin-mediated transcription, despite exhibiting retained Wnt receptor downregulation. These mutations interfere with a ubiquitin-independent suppressor role of the RNF43 cytosolic tail that involves Casein kinase 1 (CK1) binding and phosphorylation. Mechanistically, truncated RNF43 variants trap CK1 at the plasma membrane, thereby preventing ß-catenin turnover and propelling ligand-independent target gene transcription. Gene editing of human colon stem cells shows that RNF43 truncations cooperate with p53 loss to drive a niche-independent program for self-renewal and proliferation. Moreover, these RNF43 variants confer decreased sensitivity to anti-Wnt-based therapy. Our data demonstrate the relevance of studying patient-derived mutations for understanding disease mechanisms and improved applications of precision medicine.


Assuntos
Caseína Quinase I/metabolismo , Neoplasias/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Via de Sinalização Wnt , Caseína Quinase I/genética , Células HEK293 , Humanos , Neoplasias/genética , Neoplasias/patologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases/genética , beta Catenina/genética , beta Catenina/metabolismo
14.
EMBO J ; 39(3): e102771, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31867777

RESUMO

The intestinal stem cell (ISC) marker LGR5 is a receptor for R-spondin (RSPO) that functions to potentiate Wnt signalling in the proliferating crypt. It has been recently shown that Wnt plays a priming role for ISC self-renewal by inducing RSPO receptor LGR5 expression. Despite its pivotal role in homeostasis, regeneration and cancer, little is known about the post-translational regulation of LGR5. Here, we show that the HECT-domain E3 ligases NEDD4 and NEDD4L are expressed in the crypt stem cell regions and regulate ISC priming by degrading LGR receptors. Loss of Nedd4 and Nedd4l enhances ISC proliferation, increases sensitivity to RSPO stimulation and accelerates tumour development in Apcmin mice with increased numbers of high-grade adenomas. Mechanistically, we find that both NEDD4 and NEDD4L negatively regulate Wnt/ß-catenin signalling by targeting LGR5 receptor and DVL2 for proteasomal and lysosomal degradation. Our findings unveil the previously unreported post-translational control of LGR receptors via NEDD4/NEDD4L to regulate ISC priming. Inactivation of NEDD4 and NEDD4L increases Wnt activation and ISC numbers, which subsequently enhances tumour predisposition and progression.


Assuntos
Intestinos/citologia , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Receptores Acoplados a Proteínas G/química , Adenoma , Animais , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Células HCT116 , Células HEK293 , Humanos , Masculino , Camundongos , Organoides , Processamento de Proteína Pós-Traducional , Proteólise , Células-Tronco/citologia , Células-Tronco/metabolismo , Via de Sinalização Wnt
15.
Trends Cell Biol ; 30(1): 60-73, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31718893

RESUMO

Intestinal organoids grown from adult stem cells have emerged as prototype 3D organotypic models for studying tissue renewal and homeostasis. Owing to their strict dependence on Wnt signaling, intestinal organoids offer an unprecedented opportunity to examine Wnt pathway regulation in normal physiology and cancer. We review how alterations in growth factor dependency and organoid morphology can be exploited to identify Wnt signaling mechanisms, characterize mutated pathway components, and predict responses of patient-derived tumors to targeted therapy. We discuss current deficits in the understanding of genotype-phenotype relationships that are to be considered when interpreting mutation-induced changes in organoid morphology.


Assuntos
Imageamento Tridimensional , Intestinos/fisiologia , Organoides/metabolismo , Via de Sinalização Wnt , Animais , Estudos de Associação Genética , Humanos , Neoplasias/metabolismo
16.
Biol Open ; 8(8)2019 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-31362950

RESUMO

The culturing of mini-organs (organoids) in three-dimensions (3D) presents a simple and powerful tool to investigate the principles underlying human organ development and tissue self-organization in both healthy and diseased states. Applications of single molecule analysis are highly informative for a comprehensive understanding of the complexity underlying tissue and organ physiology. To fully exploit the potential of single molecule technologies, the adjustment of protocols and tools to 3D tissue culture is required. Single molecule RNA fluorescence in situ hybridization (smFISH) is a robust technique for visualizing and quantifying individual transcripts. In addition, smFISH can be employed to study splice variants, fusion transcripts as well as transcripts of multiple genes at the same time. Here, we develop a 3-day protocol and validation method to perform smFISH in 3D in whole human organoids. We provide a number of applications to exemplify the diverse possibilities for the simultaneous detection of distinct mRNA transcripts, evaluation of their spatial distribution and the identification of divergent cell lineages in 3D in organoids.

17.
iScience ; 13: 318-327, 2019 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-30878878

RESUMO

The establishment of neuronal polarity is driven by cytoskeletal remodeling that stabilizes and promotes the growth of a single axon from one of the multiple neurites. The importance of the local microtubule stabilization in this process has been revealed however, the external signals initiating the cytoskeletal rearrangements are not completely understood. In this study, we show that local activation of the canonical Wnt pathway regulates neuronal polarity and axonal outgrowth. We found that in the early stages of neuronal polarization, Wnt3a accumulates in one of the neurites of unpolarized cells and thereby could determine axon positioning. Subsequently, Wnt3a localizes to the growing axon, where it activates the canonical Wnt pathway and controls axon positioning and axonal length. We propose a model in which Wnt3a regulates the formation and growth of the axon by activating local intracellular signaling events leading to microtubule remodeling.

18.
Nat Commun ; 10(1): 365, 2019 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-30664649

RESUMO

Wnt-induced ß-catenin-mediated transcription is a driving force for stem cell self-renewal during adult tissue homeostasis. Enhanced Wnt receptor expression due to mutational inactivation of the ubiquitin ligases RNF43/ZNRF3 recently emerged as a leading cause for cancer development. Consequently, targeting canonical Wnt receptors such as LRP5/6 holds great promise for treatment of such cancer subsets. Here, we employ CIS display technology to identify single-domain antibody fragments (VHH) that bind the LRP6 P3E3P4E4 region with nanomolar affinity and strongly inhibit Wnt3/3a-induced ß-catenin-mediated transcription in cells, while leaving Wnt1 responses unaffected. Structural analysis reveal that individual VHHs variably employ divergent antigen-binding regions to bind a similar surface in the third ß-propeller of LRP5/6, sterically interfering with Wnt3/3a binding. Importantly, anti-LRP5/6 VHHs block the growth of Wnt-hypersensitive Rnf43/Znrf3-mutant intestinal organoids through stem cell exhaustion and collective terminal differentiation. Thus, VHH-mediated targeting of LRP5/6 provides a promising differentiation-inducing strategy for treatment of Wnt-hypersensitive tumors.


Assuntos
Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/química , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/química , Organoides/efeitos dos fármacos , Anticorpos de Domínio Único/química , Células-Tronco/efeitos dos fármacos , Proteína Wnt3A/genética , Animais , Sítios de Ligação , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Cristalografia por Raios X , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Intestino Delgado/citologia , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/metabolismo , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/antagonistas & inibidores , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/antagonistas & inibidores , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos , Modelos Moleculares , Organoides/citologia , Organoides/metabolismo , Ligação Proteica , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Anticorpos de Domínio Único/genética , Anticorpos de Domínio Único/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Transcrição Gênica , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteína Wnt3A/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
19.
Open Biol ; 8(9)2018 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-30209039

RESUMO

Rapidly renewing tissues such as the intestinal epithelium critically depend on the activity of small-sized stem cell populations that continuously generate new progeny to replace lost and damaged cells. The complex and tightly regulated process of intestinal homeostasis is governed by a variety of signalling pathways that balance cell proliferation and differentiation. Accumulating evidence suggests that stem cell control and daughter cell fate determination is largely dictated by the microenvironment. Here, we review recent developments in the understanding of intestinal stem cell dynamics, focusing on the roles, mechanisms and interconnectivity of prime signalling pathways that regulate stem cell behaviour in intestinal homeostasis. Furthermore, we discuss how mutational activation of these signalling pathways endows colorectal cancer cells with niche-independent growth advantages during carcinogenesis.


Assuntos
Mucosa Intestinal/citologia , Neoplasias Intestinais/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Animais , Diferenciação Celular , Plasticidade Celular , Proliferação de Células , Homeostase , Humanos , Mucosa Intestinal/metabolismo , Regeneração , Nicho de Células-Tronco , Células-Tronco/metabolismo
20.
Proc Natl Acad Sci U S A ; 115(17): E3996-E4005, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29632210

RESUMO

Wnt/ß-catenin signaling controls development and adult tissue homeostasis by regulating cell proliferation and cell fate decisions. Wnt binding to its receptors Frizzled (FZD) and low-density lipoprotein-related 6 (LRP6) at the cell surface initiates a signaling cascade that leads to the transcription of Wnt target genes. Upon Wnt binding, the receptors assemble into large complexes called signalosomes that provide a platform for interactions with downstream effector proteins. The molecular basis of signalosome formation and regulation remains elusive, largely due to the lack of tools to analyze its endogenous components. Here, we use internally tagged Wnt3a proteins to isolate and characterize activated, endogenous Wnt receptor complexes by mass spectrometry-based proteomics. We identify the single-span membrane protein TMEM59 as an interactor of FZD and LRP6 and a positive regulator of Wnt signaling. Mechanistically, TMEM59 promotes the formation of multimeric Wnt-FZD assemblies via intramembrane interactions. Subsequently, these Wnt-FZD-TMEM59 clusters merge with LRP6 to form mature Wnt signalosomes. We conclude that the assembly of multiprotein Wnt signalosomes proceeds along well-ordered steps that involve regulated intramembrane interactions.


Assuntos
Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Proteínas de Membrana/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Via de Sinalização Wnt/fisiologia , Proteína Wnt3A/metabolismo , Animais , Células HEK293 , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteínas de Membrana/genética , Camundongos , Complexos Multiproteicos/genética , Proteínas do Tecido Nervoso/genética , Proteína Wnt3A/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...