Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(39): e2309955120, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37725655

RESUMO

Cellular form and function are controlled by the assembly and stability of actin cytoskeletal structures-but disassembling/pruning these structures is equally essential for the plasticity and remodeling that underlie behavioral adaptations. Importantly, the mechanisms of actin assembly have been well-defined-including that it is driven by actin's polymerization into filaments (F-actin) and then often bundling by crosslinking proteins into stable higher-order structures. In contrast, it remains less clear how these stable bundled F-actin structures are rapidly disassembled. We now uncover mechanisms that rapidly and extensively disassemble bundled F-actin. Using biochemical, structural, and imaging assays with purified proteins, we show that F-actin bundled with one of the most prominent crosslinkers, fascin, is extensively disassembled by Mical, the F-actin disassembly enzyme. Furthermore, the product of this Mical effect, Mical-oxidized actin, is poorly bundled by fascin, thereby further amplifying Mical's disassembly effects on bundled F-actin. Moreover, another critical F-actin regulator, cofilin, also affects fascin-bundled filaments, but we find herein that it synergizes with Mical to dramatically amplify its disassembly of bundled F-actin compared to the sum of their individual effects. Genetic and high-resolution cellular assays reveal that Mical also counteracts crosslinking proteins/bundled F-actin in vivo to control cellular extension, axon guidance, and Semaphorin/Plexin cell-cell repulsion. Yet, our results also support the idea that fascin-bundling serves to dampen Mical's F-actin disassembly in vitro and in vivo-and that physiologically relevant cellular remodeling requires a fine-tuned interplay between the factors that build bundled F-actin networks and those that disassemble them.


Assuntos
Fatores de Despolimerização de Actina , Actinas , Citoesqueleto de Actina , Citoesqueleto , Orientação de Axônios
2.
Int J Mol Sci ; 22(4)2021 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-33671465

RESUMO

To change their behaviors, cells require actin proteins to assemble together into long polymers/filaments-and so a critical goal is to understand the factors that control this actin filament (F-actin) assembly and stability. We have identified a family of unusual actin regulators, the MICALs, which are flavoprotein monooxygenase/hydroxylase enzymes that associate with flavin adenine dinucleotide (FAD) and use the co-enzyme nicotinamide adenine dinucleotide phosphate (NADPH) in Redox reactions. F-actin is a specific substrate for these MICAL Redox enzymes, which oxidize specific amino acids within actin to destabilize actin filaments. Furthermore, this MICAL-catalyzed reaction is reversed by another family of Redox enzymes (SelR/MsrB enzymes)-thereby revealing a reversible Redox signaling process and biochemical mechanism regulating actin dynamics. Interestingly, in addition to the MICALs' Redox enzymatic portion through which MICALs covalently modify and affect actin, MICALs have multiple other domains. Less is known about the roles of these other MICAL domains. Here we provide approaches for obtaining high levels of recombinant protein for the Redox only portion of Mical and demonstrate its catalytic and F-actin disassembly activity. These results provide a ground state for future work aimed at defining the role of the other domains of Mical - including characterizing their effects on Mical's Redox enzymatic and F-actin disassembly activity.


Assuntos
Actinas/metabolismo , Drosophila melanogaster/enzimologia , Ensaios Enzimáticos , Oxigenases de Função Mista/química , Oxigenases de Função Mista/metabolismo , Animais , Biocatálise , Chaperoninas/metabolismo , Temperatura Baixa , Oxirredução , Domínios Proteicos , Proteínas Recombinantes/isolamento & purificação , Solubilidade
3.
Nature ; 463(7282): 823-7, 2010 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-20148037

RESUMO

How instructive cues present on the cell surface have their precise effects on the actin cytoskeleton is poorly understood. Semaphorins are one of the largest families of these instructive cues and are widely studied for their effects on cell movement, navigation, angiogenesis, immunology and cancer. Semaphorins/collapsins were characterized in part on the basis of their ability to drastically alter actin cytoskeletal dynamics in neuronal processes, but despite considerable progress in the identification of semaphorin receptors and their signalling pathways, the molecules linking them to the precise control of cytoskeletal elements remain unknown. Recently, highly unusual proteins of the Mical family of enzymes have been found to associate with the cytoplasmic portion of plexins, which are large cell-surface semaphorin receptors, and to mediate axon guidance, synaptogenesis, dendritic pruning and other cell morphological changes. Mical enzymes perform reduction-oxidation (redox) enzymatic reactions and also contain domains found in proteins that regulate cell morphology. However, nothing is known of the role of Mical or its redox activity in mediating morphological changes. Here we report that Mical directly links semaphorins and their plexin receptors to the precise control of actin filament (F-actin) dynamics. We found that Mical is both necessary and sufficient for semaphorin-plexin-mediated F-actin reorganization in vivo. Likewise, we purified Mical protein and found that it directly binds F-actin and disassembles both individual and bundled actin filaments. We also found that Mical utilizes its redox activity to alter F-actin dynamics in vivo and in vitro, indicating a previously unknown role for specific redox signalling events in actin cytoskeletal regulation. Mical therefore is a novel F-actin-disassembly factor that provides a molecular conduit through which actin reorganization-a hallmark of cell morphological changes including axon navigation-can be precisely achieved spatiotemporally in response to semaphorins.


Assuntos
Actinas/química , Actinas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Semaforinas/metabolismo , Animais , Moléculas de Adesão Celular/metabolismo , Forma Celular/fisiologia , Citoesqueleto/química , Citoesqueleto/metabolismo , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/enzimologia , Cones de Crescimento/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Oxirredução , Oxirredutases/deficiência , Oxirredutases/genética , Oxirredutases/metabolismo , Ligação Proteica
4.
Commun Integr Biol ; 11(1): e1405197, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29497471

RESUMO

To change their size, shape, and connectivity, cells require actin and tubulin proteins to assemble together into long polymers - and numerous extracellular stimuli have now been identified that alter the assembly and organization of these cytoskeletal structures. Yet, there remains a lack of defined signaling pathways from the cell surface to the cytoskeleton for many of these extracellular signals, and so we still know little of how they exert their precise structural effects. These extracellular cues may be soluble or substrate-bound and have historically been classified into two independently acting and antagonistic groups: growth-promoting/attractants (inducing turning toward the source of the factor/positive chemotropism) or growth-preventing/repellents (turning away from the source of the factor/negative chemotropism). Paradoxically, our recent results directly link the action of growth factors/chemoattractants and their signaling pathways to the promotion of the disassembly of the F-actin cytoskeleton (a defined readout of repellents/repulsive signaling). Herein, we add to this by simply driving a constitutively active form of Mical, which strongly disassembles F-actin/remodels cells in vivo independent of repulsive cues - and find that loss of Abl, which mediates growth factor signaling in these cells, decreases Mical's F-actin disassembly/cellular remodeling effects. Thus, our results are consistent with a hypothesis that cues defined as positive effectors of movement (growth factors/chemoattractants) can at least in some contexts enhance the F-actin disassembly and remodeling activity of repellents.

5.
Mol Cell Oncol ; 5(1): e1384881, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29404387

RESUMO

MICAL Redox enzymes have recently emerged as direct regulators of cell shape and motility - working through specific reversible post-translational oxidation of actin to disassemble and remodel the cytoskeleton. Links are also now emerging between MICALs and cancer, including our recent results that regulation of MICAL sensitizes cancer cells to the cancer drug Gleevec. Targeting this new actin regulatory enzyme system may thus provide new therapeutic options for cancer treatment.

6.
Sci Rep ; 8(1): 937, 2018 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-29343822

RESUMO

Cellular form and function - and thus normal development and physiology - are specified via proteins that control the organization and dynamic properties of the actin cytoskeleton. Using the Drosophila model, we have recently identified an unusual actin regulatory enzyme, Mical, which is directly activated by F-actin to selectively post-translationally oxidize and destabilize filaments - regulating numerous cellular behaviors. Mical proteins are also present in mammals, but their actin regulatory properties, including comparisons among different family members, remain poorly defined. We now find that each human MICAL family member, MICAL-1, MICAL-2, and MICAL-3, directly induces F-actin dismantling and controls F-actin-mediated cellular remodeling. Specifically, each human MICAL selectively associates with F-actin, which directly induces MICALs catalytic activity. We also find that each human MICAL uses an NADPH-dependent Redox activity to post-translationally oxidize actin's methionine (M) M44/M47 residues, directly dismantling filaments and limiting new polymerization. Genetic experiments also demonstrate that each human MICAL drives F-actin disassembly in vivo, reshaping cells and their membranous extensions. Our results go on to reveal that MsrB/SelR reductase enzymes counteract each MICAL's effect on F-actin in vitro and in vivo. Collectively, our results therefore define the MICALs as an important phylogenetically-conserved family of catalytically-acting F-actin disassembly factors.


Assuntos
Actinas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Drosophila/metabolismo , Oxirredutases/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Proteínas do Citoesqueleto/metabolismo , Humanos , Oxirredução
7.
Mol Cell Biol ; 38(17)2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29866653

RESUMO

Adenomatous polyposis coli (APC) is a key molecule to maintain cellular homeostasis in colonic epithelium by regulating cell-cell adhesion, cell polarity, and cell migration through activating the APC-stimulated guanine nucleotide-exchange factor (Asef). The APC-activated Asef stimulates the small GTPase, which leads to decreased cell-cell adherence and cell polarity, and enhanced cell migration. In colorectal cancers, while truncated APC constitutively activates Asef and promotes cancer initiation and progression, regulation of Asef by full-length APC is still unclear. Here, we report the autoinhibition mechanism of full-length APC. We found that the armadillo repeats in full-length APC interact with the APC residues 1362 to 1540 (APC-2,3 repeats), and this interaction competes off and inhibits Asef. Deletion of APC-2,3 repeats permits Asef interactions leading to downstream signaling events, including the induction of Golgi fragmentation through the activation of the Asef-ROCK-MLC2. Truncated APC also disrupts protein trafficking and cholesterol homeostasis by inhibition of SREBP2 activity in a Golgi fragmentation-dependent manner. Our study thus uncovers the autoinhibition mechanism of full-length APC and a novel gain of function of truncated APC in regulating Golgi structure, as well as cholesterol homeostasis, which provides a potential target for pharmaceutical intervention against colon cancers.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Proteína da Polipose Adenomatosa do Colo/metabolismo , Mutação com Ganho de Função , Genes APC , Complexo de Golgi/metabolismo , Proteína da Polipose Adenomatosa do Colo/química , Sequência de Aminoácidos , Proteínas do Domínio Armadillo/química , Proteínas do Domínio Armadillo/genética , Proteínas do Domínio Armadillo/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Movimento Celular , Colesterol/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Complexo de Golgi/patologia , Células HCT116 , Células HT29 , Homeostase , Humanos , Modelos Biológicos , Modelos Moleculares , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Deleção de Sequência , Transdução de Sinais
8.
Methods Mol Biol ; 1493: 119-128, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27787846

RESUMO

The MICALs are a family of phylogenetically conserved cytoplasmic proteins that modulate numerous cellular behaviors and play critical roles in semaphorin-plexin signaling. Our recent results have revealed that the MICALs are an unusual family of actin regulatory proteins that use actin filaments (F-actin) as a direct substrate-controlling F-actin dynamics via stereospecific oxidation of conserved methionine (Met44 and Met47) residues within actin. In particular, the MICALs have a highly conserved flavoprotein monooxygenase (redox) enzymatic domain in their N-terminus that directly oxidizes and destabilizes F-actin. Here, we describe methods to characterize MICAL-mediated F-actin disassembly using in vitro assays with purified proteins.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas com Domínio LIM/metabolismo , Semaforinas/metabolismo , Transdução de Sinais , Actinas/metabolismo , Eletroforese em Gel de Poliacrilamida , Humanos , Proteínas dos Microfilamentos , Oxigenases de Função Mista , Oxirredução , Espectrometria de Fluorescência
9.
Dev Cell ; 42(2): 117-129.e8, 2017 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-28689759

RESUMO

Extracellular cues that regulate cellular shape, motility, and navigation are generally classified as growth promoting (i.e., growth factors/chemoattractants and attractive guidance cues) or growth preventing (i.e., repellents and inhibitors). Yet, these designations are often based on complex assays and undefined signaling pathways and thus may misrepresent direct roles of specific cues. Here, we find that a recognized growth-promoting signaling pathway amplifies the F-actin disassembly and repulsive effects of a growth-preventing pathway. Focusing on Semaphorin/Plexin repulsion, we identified an interaction between the F-actin-disassembly enzyme Mical and the Abl tyrosine kinase. Biochemical assays revealed Abl phosphorylates Mical to directly amplify Mical Redox-mediated F-actin disassembly. Genetic assays revealed that Abl allows growth factors and Semaphorin/Plexin repellents to combinatorially increase Mical-mediated F-actin disassembly, cellular remodeling, and repulsive axon guidance. Similar roles for Mical in growth factor/Abl-related cancer cell behaviors further revealed contexts in which characterized positive effectors of growth/guidance stimulate such negative cellular effects as F-actin disassembly/repulsion.


Assuntos
Actinas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Transdução de Sinais , Animais , Orientação de Axônios/efeitos dos fármacos , Biocatálise/efeitos dos fármacos , Fenômenos Biomecânicos , Moléculas de Adesão Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Drosophila melanogaster/embriologia , Drosophila melanogaster/metabolismo , Humanos , Mesilato de Imatinib/farmacologia , Camundongos Nus , Oxigenases de Função Mista/química , Oxigenases de Função Mista/metabolismo , Modelos Biológicos , Neoplasias/patologia , Proteínas do Tecido Nervoso/metabolismo , Oxirredução , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Domínios Proteicos , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-abl/metabolismo , Semaforinas/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA