Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Mol Biol Cell ; 35(6): ar80, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38598293

ABSTRACT

The actin cytoskeleton is essential for many functions of eukaryotic cells, but the factors that nucleate actin assembly are not well understood at the organismal level or in the context of disease. To explore the function of the actin nucleation factor WHAMM in mice, we examined how Whamm inactivation impacts kidney physiology and cellular proteostasis. We show that male WHAMM knockout mice excrete elevated levels of albumin, glucose, phosphate, and amino acids, and display structural abnormalities of the kidney proximal tubule, suggesting that WHAMM activity is important for nutrient reabsorption. In kidney tissue, the loss of WHAMM results in the accumulation of the lipidated autophagosomal membrane protein LC3, indicating an alteration in autophagy. In mouse fibroblasts and human proximal tubule cells, WHAMM and its binding partner the Arp2/3 complex control autophagic membrane closure and cargo receptor recruitment. These results reveal a role for WHAMM-mediated actin assembly in maintaining kidney function and promoting proper autophagosome membrane remodeling.


Subject(s)
Actins , Autophagosomes , Autophagy , Kidney , Mice, Knockout , Animals , Mice , Actins/metabolism , Autophagy/physiology , Humans , Autophagosomes/metabolism , Kidney/metabolism , Male , Kidney Tubules, Proximal/metabolism , Actin Cytoskeleton/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Polymerization , Fibroblasts/metabolism
2.
bioRxiv ; 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38559247

ABSTRACT

Autophagy is an intracellular degradation process that maintains homeostasis, responds to stress, and plays key roles in the prevention of aging and disease. Autophagosome biogenesis, vesicle rocketing, and autolysosome tubulation are controlled by multiple actin nucleation factors, but the impact of actin assembly on completion of the autophagic pathway is not well understood. Here we studied autophagosome and lysosome remodeling in fibroblasts harboring an inducible knockout (iKO) of the Arp2/3 complex, an essential actin nucleator. Arp2/3 complex ablation resulted in increased basal levels of autophagy receptors and lipidated membrane proteins from the LC3 and GABARAP families. Under both steady-state and starvation conditions, Arp2/3 iKO cells accumulated abnormally high numbers of autolysosomes, suggesting a defect in autophagic flux. The inability of Arp2/3 complex-deficient cells to complete autolysosome degradation and turnover is explained by the presence of damaged, leaky lysosomes. In cells treated with an acute lysosomal membrane-damaging agent, the Arp2/3-activating protein WHAMM is recruited to lysosomes, where Arp2/3 complex-dependent actin assembly is crucial for restoring intact lysosomal structure. These results establish the Arp2/3 complex as a central player late in the canonical autophagy pathway and reveal a new role for the actin nucleation machinery in maintaining lysosomal integrity.

3.
bioRxiv ; 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38328079

ABSTRACT

The actin cytoskeleton is essential for many functions of eukaryotic cells, but the factors that nucleate actin assembly are not well understood at the organismal level or in the context of disease. To explore the function of the actin nucleation factor WHAMM in mice, we examined how Whamm inactivation impacts kidney physiology and cellular proteostasis. We show that male WHAMM knockout mice excrete elevated levels of albumin, glucose, phosphate, and amino acids, and display abnormalities of the kidney proximal tubule, suggesting that WHAMM activity is important for nutrient reabsorption. In kidney tissue, the loss of WHAMM results in the accumulation of the lipidated autophagosomal membrane protein LC3, indicating an alteration in autophagy. In mouse fibroblasts and human proximal tubule cells, WHAMM and its binding partner the Arp2/3 complex control autophagic membrane closure and cargo receptor recruitment. These results reveal a role for WHAMM-mediated actin assembly in maintaining kidney function and promoting proper autophagosome membrane remodeling.

4.
Eur J Cell Biol ; 102(2): 151301, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36907023

ABSTRACT

The actin cytoskeleton impacts practically every function of a eukaryotic cell. Historically, the best-characterized cytoskeletal activities are in cell morphogenesis, motility, and division. The structural and dynamic properties of the actin cytoskeleton are also crucial for establishing, maintaining, and changing the organization of membrane-bound organelles and other intracellular structures. Such activities are important in nearly all animal cells and tissues, although distinct anatomical regions and physiological systems rely on different regulatory factors. Recent work indicates that the Arp2/3 complex, a broadly expressed actin nucleator, drives actin assembly during several intracellular stress response pathways. These newly described Arp2/3-mediated cytoskeletal rearrangements are coordinated by members of the Wiskott-Aldrich Syndrome Protein (WASP) family of actin nucleation-promoting factors. Thus, the Arp2/3 complex and WASP-family proteins are emerging as crucial players in cytoplasmic and nuclear activities including autophagy, apoptosis, chromatin dynamics, and DNA repair. Characterizations of the functions of the actin assembly machinery in such stress response mechanisms are advancing our understanding of both normal and pathogenic processes, and hold great promise for providing insights into organismal development and interventions for disease.


Subject(s)
Actins , Wiskott-Aldrich Syndrome Protein Family , Animals , Wiskott-Aldrich Syndrome Protein Family/metabolism , Actins/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Actin Cytoskeleton/metabolism , Cytoskeleton/metabolism , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/metabolism , Actin-Related Protein 3/metabolism
5.
Mol Biol Cell ; 34(5): ar41, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36920061

ABSTRACT

The actin cytoskeleton is a ubiquitous participant in cellular functions that maintain viability, but how it controls programmed cell death is not well understood. Here we show that in response to DNA damage, human cells form a juxtanuclear F-actin-rich territory that coordinates the organized progression of apoptosome assembly to caspase activation. This cytoskeletal compartment is created by the actin nucleation factors JMY, WHAMM, and the Arp2/3 complex, and it excludes proteins that inhibit JMY and WHAMM activity. Within the territory, mitochondria undergo outer membrane permeabilization and JMY localization overlaps with punctate structures containing the core apoptosome components cytochrome c and Apaf-1. The F-actin-rich area also encompasses initiator caspase-9 and clusters of a cleaved form of executioner caspase-3 but restricts accessibility of the caspase inhibitor XIAP. The clustering and potency of caspase-3 activation are positively regulated by the amount of actin polymerized by JMY and WHAMM. These results indicate that JMY-mediated actin reorganization functions in apoptotic signaling by coupling the biogenesis of apoptosomes to the localized processing of caspases.


Subject(s)
Actins , Apoptosomes , Humans , Actins/metabolism , Caspase 3 , Apoptosomes/metabolism , Apoptosis/physiology , Caspases/metabolism , Actin Cytoskeleton/metabolism , DNA Damage , Membrane Proteins/metabolism , Microtubule-Associated Proteins
6.
PLoS Genet ; 19(1): e1010045, 2023 01.
Article in English | MEDLINE | ID: mdl-36706133

ABSTRACT

The Arp2/3 complex is an actin nucleator with well-characterized activities in cell morphogenesis and movement, but its roles in nuclear processes are relatively understudied. We investigated how the Arp2/3 complex affects genomic integrity and cell cycle progression using mouse fibroblasts containing an inducible knockout (iKO) of the ArpC2 subunit. We show that permanent Arp2/3 complex ablation results in DNA damage, the formation of cytosolic micronuclei, and cellular senescence. Micronuclei arise in ArpC2 iKO cells due to chromatin segregation defects during mitosis and premature mitotic exits. Such phenotypes are explained by the presence of damaged DNA fragments that fail to attach to the mitotic spindle, abnormalities in actin assembly during metaphase, and asymmetric microtubule architecture during anaphase. In the nuclei of Arp2/3-depleted cells, the tumor suppressor p53 is activated and the cell cycle inhibitor Cdkn1a/p21 mediates a G1 arrest. In the cytosol, micronuclei are recognized by the DNA sensor cGAS, which is important for stimulating a STING- and IRF3-associated interferon response. These studies establish functional requirements for the mammalian Arp2/3 complex in mitotic spindle organization and genome stability. They also expand our understanding of the mechanisms leading to senescence and suggest that cytoskeletal dysfunction is an underlying factor in biological aging.


Subject(s)
Actin-Related Protein 2-3 Complex , Actins , Animals , Mice , Actin-Related Protein 2-3 Complex/genetics , Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Cellular Senescence/genetics , DNA/metabolism , Genomic Instability/genetics , Mitosis/genetics
7.
PLoS Genet ; 17(4): e1009512, 2021 04.
Article in English | MEDLINE | ID: mdl-33872315

ABSTRACT

The actin cytoskeleton is a well-known player in most vital cellular processes, but comparably little is understood about how the actin assembly machinery impacts programmed cell death pathways. In the current study, we explored roles for the human Wiskott-Aldrich Syndrome Protein (WASP) family of actin nucleation factors in DNA damage-induced apoptosis. Inactivation of each WASP-family gene revealed that two of them, JMY and WHAMM, are necessary for rapid apoptotic responses. JMY and WHAMM participate in a p53-dependent cell death pathway by enhancing mitochondrial permeabilization, initiator caspase cleavage, and executioner caspase activation. JMY-mediated apoptosis requires actin nucleation via the Arp2/3 complex, and actin filaments are assembled in cytoplasmic territories containing clusters of cytochrome c and active caspase-3. The loss of JMY additionally results in significant changes in gene expression, including upregulation of the WHAMM-interacting G-protein RhoD. Depletion or deletion of RHOD increases cell death, suggesting that RhoD normally contributes to cell survival. These results give rise to a model in which JMY and WHAMM promote intrinsic cell death responses that can be opposed by RhoD.


Subject(s)
Membrane Proteins/genetics , Microtubule-Associated Proteins/genetics , Nuclear Proteins/genetics , Trans-Activators/genetics , Tumor Suppressor Protein p53/genetics , Wiskott-Aldrich Syndrome/genetics , rho GTP-Binding Proteins/genetics , Actin Cytoskeleton/genetics , Actin-Related Protein 2/genetics , Actin-Related Protein 2-3 Complex/genetics , Actin-Related Protein 3/genetics , Apoptosis/genetics , Cytochromes c/genetics , DNA Damage/genetics , Humans , Mitochondria/genetics , Mitochondria/metabolism , RNA, Small Interfering/genetics , Wiskott-Aldrich Syndrome Protein/genetics
8.
Methods Mol Biol ; 2291: 145-162, 2021.
Article in English | MEDLINE | ID: mdl-33704752

ABSTRACT

The bacteriophage Lambda (λ) "Red" recombination system has enabled the development of efficient methods for engineering bacterial chromosomes. This system has been particularly important to the field of bacterial pathogenesis, where it has advanced the study of virulence factors from Shiga toxin-producing and enteropathogenic Escherichia coli (STEC and EPEC). Transient plasmid-driven expression of Lambda Red allows homologous recombination between PCR-derived linear DNA substrates and target loci in the STEC/EPEC chromosomes. Red-associated techniques can be used to create individual gene knockouts, generate deletions of large pathogenicity islands, and make markerless allelic exchanges. This chapter describes specific strategies and procedures for performing Lambda Red-mediated genome engineering in STEC.


Subject(s)
Bacteriophage lambda/metabolism , Enteropathogenic Escherichia coli/metabolism , Recombination, Genetic , Shiga-Toxigenic Escherichia coli/metabolism , Viral Proteins/metabolism , Bacteriophage lambda/genetics , Enteropathogenic Escherichia coli/genetics , Escherichia coli Infections/genetics , Escherichia coli Infections/metabolism , Shiga-Toxigenic Escherichia coli/genetics , Viral Proteins/genetics
9.
J Cell Biol ; 219(4)2020 04 06.
Article in English | MEDLINE | ID: mdl-32328644

ABSTRACT

Filopodia are actin-rich protrusions important for sensing and responding to the extracellular environment, but the repertoire of factors required for filopodia formation is only partially understood. Jarsch et al. (2020. J. Cell. Biol. https://doi.org/10.1083/jcb.201909178) combine an in vitro system of filopodia biogenesis with a phage display screen to show that SNX9 drives filopodial assembly.


Subject(s)
Actins , Pseudopodia
10.
PLoS Genet ; 16(3): e1008694, 2020 03.
Article in English | MEDLINE | ID: mdl-32196488

ABSTRACT

Cell motility is governed by cooperation between the Arp2/3 complex and nucleation-promoting factors from the Wiskott-Aldrich Syndrome Protein (WASP) family, which together assemble actin filament networks to drive membrane protrusion. Here we identify WHIMP (WAVE Homology In Membrane Protrusions) as a new member of the WASP family. The Whimp gene is encoded on the X chromosome of a subset of mammals, including mice. Murine WHIMP promotes Arp2/3-dependent actin assembly, but is less potent than other nucleation factors. Nevertheless, WHIMP-mediated Arp2/3 activation enhances both plasma membrane ruffling and wound healing migration, whereas WHIMP depletion impairs protrusion and slows motility. WHIMP expression also increases Src-family kinase activity, and WHIMP-induced ruffles contain the additional nucleation-promoting factors WAVE1, WAVE2, and N-WASP, but not JMY or WASH. Perturbing the function of Src-family kinases, WAVE proteins, or Arp2/3 complex inhibits WHIMP-driven ruffling. These results suggest that WHIMP-associated actin assembly plays a direct role in membrane protrusion, but also results in feedback control of tyrosine kinase signaling to modulate the activation of multiple WASP-family members.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Movement/physiology , Cell Surface Extensions/physiology , Wiskott-Aldrich Syndrome Protein/metabolism , src-Family Kinases/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Animals , Cell Line , Endocytosis/physiology , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Protein Domains , Signal Transduction , Wiskott-Aldrich Syndrome Protein Family/metabolism
11.
PLoS Pathog ; 14(12): e1007485, 2018 12.
Article in English | MEDLINE | ID: mdl-30550556

ABSTRACT

Enteropathogenic and enterohemorrhagic E. coli (EPEC and EHEC) are closely related extracellular pathogens that reorganize host cell actin into "pedestals" beneath the tightly adherent bacteria. This pedestal-forming activity is both a critical step in pathogenesis, and it makes EPEC and EHEC useful models for studying the actin rearrangements that underlie membrane protrusions. To generate pedestals, EPEC relies on the tyrosine phosphorylated bacterial effector protein Tir to bind host adaptor proteins that recruit N-WASP, a nucleation-promoting factor that activates the Arp2/3 complex to drive actin polymerization. In contrast, EHEC depends on the effector EspFU to multimerize N-WASP and promote Arp2/3 activation. Although these core pathways of pedestal assembly are well-characterized, the contributions of additional actin nucleation factors are unknown. We investigated potential cooperation between the Arp2/3 complex and other classes of nucleators using chemical inhibitors, siRNAs, and knockout cell lines. We found that inhibition of formins impairs actin pedestal assembly, motility, and cellular colonization for bacteria using the EPEC, but not the EHEC, pathway of actin polymerization. We also identified mDia1 as the formin contributing to EPEC pedestal assembly, as its expression level positively correlates with the efficiency of pedestal formation, and it localizes to the base of pedestals both during their initiation and once they have reached steady state. Collectively, our data suggest that mDia1 enhances EPEC pedestal biogenesis and maintenance by generating seed filaments to be used by the N-WASP-Arp2/3-dependent actin nucleation machinery and by sustaining Src-mediated phosphorylation of Tir.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Enteropathogenic Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Host-Pathogen Interactions/physiology , Actin Cytoskeleton , Caco-2 Cells , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Proteins/metabolism , Formins , HeLa Cells , Humans , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism
13.
PLoS Pathog ; 13(8): e1006501, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28771584

ABSTRACT

Enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC) are closely-related pathogens that attach tightly to intestinal epithelial cells, efface microvilli, and promote cytoskeletal rearrangements into protrusions called actin pedestals. To trigger pedestal formation, EPEC employs the tyrosine phosphorylated transmembrane receptor Tir, while EHEC relies on the multivalent scaffolding protein EspFU. The ability to generate these structures correlates with bacterial colonization in several animal models, but the precise function of pedestals in infection remains unclear. To address this uncertainty, we characterized the colonization properties of EPEC and EHEC during infection of polarized epithelial cells. We found that EPEC and EHEC both formed distinct bacterial communities, or "macrocolonies," that encompassed multiple host cells. Tir and EspFU, as well as the host Arp2/3 complex, were all critical for the expansion of macrocolonies over time. Unexpectedly, EspFU accelerated the formation of larger macrocolonies compared to EPEC Tir, as EspFU-mediated actin assembly drove faster bacterial motility to cell junctions, where bacteria formed a secondary pedestal on a neighboring cell and divided, allowing one of the daughters to disengage and infect the second cell. Collectively, these data reveal that EspFU enhances epithelial colonization by increasing actin-based motility and promoting an efficient method of cell-to-cell transmission.


Subject(s)
Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Actins/metabolism , Caco-2 Cells , Carrier Proteins/metabolism , Chemotaxis/physiology , Cytoskeleton/metabolism , Enterohemorrhagic Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Proteins/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Microscopy, Electron, Transmission , Microscopy, Fluorescence
14.
Mol Biol Cell ; 28(19): 2492-2507, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28720660

ABSTRACT

Actin nucleation factors function to organize, shape, and move membrane-bound organelles, yet they remain poorly defined in relation to disease. Galloway-Mowat syndrome (GMS) is an inherited disorder characterized by microcephaly and nephrosis resulting from mutations in the WDR73 gene. This core clinical phenotype appears frequently in the Amish, where virtually all affected individuals harbor homozygous founder mutations in WDR73 as well as the closely linked WHAMM gene, which encodes a nucleation factor. Here we show that patient cells with both mutations exhibit cytoskeletal irregularities and severe defects in autophagy. Reintroduction of wild-type WHAMM restored autophagosomal biogenesis to patient cells, while inactivation of WHAMM in healthy cell lines inhibited lipidation of the autophagosomal protein LC3 and clearance of ubiquitinated protein aggregates. Normal WHAMM function involved binding to the phospholipid PI(3)P and promoting actin nucleation at nascent autophagosomes. These results reveal a cytoskeletal pathway controlling autophagosomal remodeling and illustrate several molecular processes that are perturbed in Amish GMS patients.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Amish/genetics , Frameshift Mutation , Membrane Proteins/genetics , Microtubule-Associated Proteins/genetics , Phosphatidylinositol Phosphates/metabolism , Actin-Related Protein 2-3 Complex/genetics , Actins/metabolism , Autophagosomes/metabolism , Autophagosomes/physiology , Cells, Cultured , Cytoskeleton/metabolism , Founder Effect , Hernia, Hiatal/genetics , Homozygote , Humans , Membrane Proteins/metabolism , Microcephaly/genetics , Microtubule-Associated Proteins/metabolism , Models, Molecular , Nephrosis/genetics , Phosphatidylinositol Phosphates/genetics , Proteins/genetics , Proteins/metabolism
15.
Mol Biol Cell ; 27(6): 967-78, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26823012

ABSTRACT

Small G-proteins are key regulatory molecules that activate the actin nucleation machinery to drive cytoskeletal rearrangements during plasma membrane remodeling. However, the ability of small G-proteins to interact with nucleation factors on internal membranes to control trafficking processes has not been well characterized. Here we investigated roles for members of the Rho, Arf, and Rab G-protein families in regulating WASP homologue associated with actin, membranes, and microtubules (WHAMM), an activator of Arp2/3 complex-mediated actin nucleation. We found that Rab1 stimulated the formation and elongation of WHAMM-associated membrane tubules in cells. Active Rab1 recruited WHAMM to dynamic tubulovesicular structures in fibroblasts, and an active prenylated version of Rab1 bound directly to an N-terminal domain of WHAMM in vitro. In contrast to other G-protein-nucleation factor interactions, Rab1 binding inhibited WHAMM-mediated actin assembly. This ability of Rab1 to regulate WHAMM and the Arp2/3 complex represents a distinct strategy for membrane remodeling in which a Rab G-protein recruits the actin nucleation machinery but dampens its activity.


Subject(s)
Actin Cytoskeleton/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Cell Membrane/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Binding Sites , Chlorocebus aethiops , Dogs , Fibroblasts/metabolism , Humans , Mice , Microtubules/metabolism , Protein Binding , Protein Processing, Post-Translational , rho GTP-Binding Proteins
16.
Mol Biol Cell ; 27(2): 247-59, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26609071

ABSTRACT

The Nck adaptor protein recruits cytosolic effectors such as N-WASP that induce localized actin polymerization. Experimental aggregation of Nck SH3 domains at the membrane induces actin comet tails--dynamic, elongated filamentous actin structures similar to those that drive the movement of microbial pathogens such as vaccinia virus. Here we show that experimental manipulation of the balance between unbranched/branched nucleation altered the morphology and dynamics of Nck-induced actin comets. Inhibition of linear, formin-based nucleation with the small-molecule inhibitor SMIFH2 or overexpression of the formin FH1 domain resulted in formation of predominantly circular-shaped actin structures with low mobility (actin blobs). These results indicate that formin-based linear actin polymerization is critical for the formation and maintenance of Nck-dependent actin comet tails. Consistent with this, aggregation of an exclusively branched nucleation-promoting factor (the VCA domain of N-WASP), with density and turnover similar to those of N-WASP in Nck comets, did not reconstitute dynamic, elongated actin comets. Furthermore, enhancement of branched Arp2/3-mediated nucleation by N-WASP overexpression caused loss of the typical actin comet tail shape induced by Nck aggregation. Thus the ratio of linear to dendritic nucleation activity may serve to distinguish the properties of actin structures induced by various viral and bacterial pathogens.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Oncogene Proteins/metabolism , Animals , Dendrites/metabolism , Fetal Proteins/metabolism , Formins , HeLa Cells , Humans , Mice , Microfilament Proteins/metabolism , NIH 3T3 Cells , Nuclear Proteins/metabolism , Structure-Activity Relationship , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , src Homology Domains
17.
J Cell Biol ; 199(1): 111-24, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-23027905

ABSTRACT

The microtubule (MT) and actin cytoskeletons drive many essential cellular processes, yet fairly little is known about how their functions are coordinated. One factor that mediates important cross talk between these two systems is WHAMM, a Golgi-associated protein that utilizes MT binding and actin nucleation activities to promote membrane tubulation during intracellular transport. Using cryoelectron microscopy and other biophysical and biochemical approaches, we unveil the underlying mechanisms for how these activities are coordinated. We find that WHAMM bound to the outer surface of MT protofilaments via a novel interaction between its central coiled-coil region and tubulin heterodimers. Upon the assembly of WHAMM onto MTs, its N-terminal membrane-binding domain was exposed at the MT periphery, where it can recruit vesicles and remodel them into tubular structures. In contrast, MT binding masked the C-terminal portion of WHAMM and prevented it from promoting actin nucleation. These results give rise to a model whereby distinct MT-bound and actin-nucleating populations of WHAMM collaborate during membrane tubulation.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Membrane/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Animals , Golgi Apparatus/metabolism , Humans , Membrane Proteins/genetics , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spodoptera
18.
J Biol Chem ; 287(24): 20613-24, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22544751

ABSTRACT

Many bacterial pathogens reorganize the host actin cytoskeleton during the course of infection, including enterohemorrhagic Escherichia coli (EHEC), which utilizes the effector protein EspF(U) to assemble actin filaments within plasma membrane protrusions called pedestals. EspF(U) activates N-WASP, a host actin nucleation-promoting factor that is normally auto-inhibited and found in a complex with the actin-binding protein WIP. Under native conditions, this N-WASP/WIP complex is activated by the small GTPase Cdc42 in concert with several different SH3 (Src-homology-3) domain-containing proteins. In the current study, we tested whether SH3 domains from the F-BAR (FCH-Bin-Amphiphysin-Rvs) subfamily of membrane-deforming proteins are involved in actin pedestal formation. We found that three F-BAR proteins: CIP4, FBP17, and TOCA1 (transducer of Cdc42-dependent actin assembly), play different roles during actin pedestal biogenesis. Whereas CIP4 and FBP17 inhibited actin pedestal assembly, TOCA1 stimulated this process. TOCA1 was recruited to pedestals by its SH3 domain, which bound directly to proline-rich sequences within EspF(U). Moreover, EspF(U) and TOCA1 activated the N-WASP/WIP complex in an additive fashion in vitro, suggesting that TOCA1 can augment actin assembly within pedestals. These results reveal that EspF(U) acts as a scaffold to recruit multiple actin assembly factors whose functions are normally regulated by Cdc42.


Subject(s)
Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Enterohemorrhagic Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , cdc42 GTP-Binding Protein/metabolism , Animals , COS Cells , Carrier Proteins/genetics , Chlorocebus aethiops , Cytoskeletal Proteins/genetics , Enterohemorrhagic Escherichia coli/genetics , Escherichia coli Infections/genetics , Escherichia coli Proteins/genetics , Fatty Acid-Binding Proteins , Humans , Intracellular Signaling Peptides and Proteins/genetics , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Minor Histocompatibility Antigens , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics , cdc42 GTP-Binding Protein/genetics , src Homology Domains
19.
Autophagy ; 7(1): 17-26, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20980813

ABSTRACT

Autophagy mediates the degradation of cytoplasmic components in eukaryotic cells and plays a key role in immunity. The mechanism of autophagosome formation is not clear. Here we examined two potential membrane sources for antibacterial autophagy: the ER and mitochondria. DFCP1, a marker of specialized ER domains known as 'omegasomes,' associated with Salmonella-containing autophagosomes via its PtdIns(3)P and ER-binding domains, while a mitochondrial marker (cytochrome b5-GFP) did not. Rab1 also localized to autophagosomes, and its activity was required for autophagosome formation, clearance of protein aggregates and peroxisomes, and autophagy of Salmonella. Overexpression of Rab1 enhanced antibacterial autophagy. The role of Rab1 in antibacterial autophagy was independent of its role in ER-to-Golgi transport. Our data suggest that antibacterial autophagy occurs at omegasomes and reveal that the Rab1 GTPase plays a crucial role in mammalian autophagy.


Subject(s)
Autophagy , Endoplasmic Reticulum/enzymology , Intracellular Membranes/enzymology , Phosphatidylinositol Phosphates/metabolism , Salmonella typhimurium/immunology , rab1 GTP-Binding Proteins/metabolism , Animals , Carrier Proteins/metabolism , Endoplasmic Reticulum/drug effects , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , HeLa Cells , Humans , Intracellular Membranes/drug effects , Mice , Peroxisomes/drug effects , Peroxisomes/metabolism , Phagosomes/microbiology , Protein Structure, Quaternary , Protein Transport/drug effects , Sirolimus/pharmacology , Ubiquitinated Proteins/chemistry
20.
Trends Cell Biol ; 20(11): 650-61, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20888769

ABSTRACT

Arp2/3 complex mediates the nucleation of actin filaments in multiple subcellular processes, and is activated by nucleation-promoting factors (NPFs) from the Wiskott-Aldrich Syndrome family. In exciting new developments, this family has grown by three members: WASH, WHAMM and JMY, which extend the repertoire of dynamic membrane structures that are remodeled following Arp2/3 activation in vivo. These novel NPFs share an actin- and Arp2/3-interacting WCA module, and combine Arp2/3 activation with additional biochemical functions, including capping protein inhibition, microtubule engagement or Arp2/3-independent actin nucleation, none of which had been previously associated with canonical WCA-harboring proteins. Uncovering the physiological relevance of these unique activities will require concerted efforts from multiple disciplines, and is sure to impact our understanding of how the cytoskeleton controls so many dynamic subcellular events.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Microfilament Proteins/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Animals , Endosomes/metabolism , Humans , Microtubules/metabolism , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...