Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 2071, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38453915

RESUMEN

IL-11 and IL-6 activate signalling via assembly of the cell surface receptor gp130; however, it is unclear how signals are transmitted across the membrane to instruct cellular responses. Here we solve the cryoEM structure of the IL-11 receptor recognition complex to discover how differences in gp130-binding interfaces may drive signalling outcomes. We explore how mutations in the IL6ST gene encoding for gp130, which cause severe immune deficiencies in humans, impair signalling without blocking cytokine binding. We use cryoEM to solve structures of both IL-11 and IL-6 complexes with a mutant form of gp130 associated with human disease. Together with molecular dynamics simulations, we show that the disease-associated variant led to an increase in flexibility including motion within the cytokine-binding core and increased distance between extracellular domains. However, these distances are minimized as the transmembrane helix exits the membrane, suggesting a stringency in geometry for signalling and dimmer switch mode of action.


Asunto(s)
Interleucina-11 , Interleucina-6 , Humanos , Interleucina-11/genética , Interleucina-6/metabolismo , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/metabolismo , Transducción de Señal , Receptores de Interleucina-6/genética
2.
Toxins (Basel) ; 15(7)2023 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-37505699

RESUMEN

CD59 is a GPI-anchored cell surface receptor that serves as a gatekeeper to controlling pore formation. It is the only membrane-bound inhibitor of the complement membrane attack complex (MAC), an immune pore that can damage human cells. While CD59 blocks MAC pores, the receptor is co-opted by bacterial pore-forming proteins to target human cells. Recent structures of CD59 in complexes with binding partners showed dramatic differences in the orientation of its ectodomain relative to the membrane. Here, we show how GPI-anchored CD59 can satisfy this diversity in binding modes. We present a PyLipID analysis of coarse-grain molecular dynamics simulations of a CD59-inhibited MAC to reveal residues of complement proteins (C6:Y285, C6:R407 C6:K412, C7:F224, C8ß:F202, C8ß:K326) that likely interact with lipids. Using modules of the MDAnalysis package to investigate atomistic simulations of GPI-anchored CD59, we discover properties of CD59 that encode the flexibility necessary to bind both complement proteins and bacterial virulence factors.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento , Proteínas del Sistema Complemento , Humanos , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Antígenos CD59/química , Antígenos CD59/metabolismo , Bacterias/metabolismo
3.
Wellcome Open Res ; 8: 76, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37234743

RESUMEN

Background: Hyaluronic acid (HA) is a major polysaccharide component of the extracellular matrix. HA has essential functions in tissue architecture and the regulation of cell behaviour. HA turnover needs to be finely balanced. Increased HA degradation is associated with cancer, inflammation, and other pathological situations. Transmembrane protein 2 (TMEM2) is a cell surface protein that has been reported to degrade HA into ~5 kDa fragments and play an essential role in systemic HA turnover. Methods: We produced the soluble TMEM2 ectodomain (residues 106-1383; sTMEM2) in human embryonic kidney cells (HEK293) and determined its structure using X-ray crystallography. We tested sTMEM2 hyaluronidase activity using fluorescently labelled HA and size fractionation of reaction products. We tested HA binding in solution and using a glycan microarray. Results: Our crystal structure of sTMEM2 confirms a remarkably accurate prediction by AlphaFold. sTMEM2 contains a parallel ß-helix typical of other polysaccharide-degrading enzymes, but an active site cannot be assigned with confidence. A lectin-like domain is inserted into the ß-helix and predicted to be functional in carbohydrate binding. A second lectin-like domain at the C-terminus is unlikely to bind carbohydrates. We did not observe HA binding in two assay formats, suggesting a modest affinity at best. Unexpectedly, we were unable to observe any HA degradation by sTMEM2. Our negative results set an upper limit for k cat of approximately 10 -5 min -1. Conclusions: Although sTMEM2 contains domain types consistent with its suggested role in TMEM2 degradation, its hyaluronidase activity was undetectable. HA degradation by TMEM2 may require additional proteins and/or localisation at the cell surface.

4.
Nat Commun ; 14(1): 890, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36797260

RESUMEN

CD59 is an abundant immuno-regulatory receptor that protects human cells from damage during complement activation. Here we show how the receptor binds complement proteins C8 and C9 at the membrane to prevent insertion and polymerization of membrane attack complex (MAC) pores. We present cryo-electron microscopy structures of two inhibited MAC precursors known as C5b8 and C5b9. We discover that in both complexes, CD59 binds the pore-forming ß-hairpins of C8 to form an intermolecular ß-sheet that prevents membrane perforation. While bound to C8, CD59 deflects the cascading C9 ß-hairpins, rerouting their trajectory into the membrane. Preventing insertion of C9 restricts structural transitions of subsequent monomers and indirectly halts MAC polymerization. We combine our structural data with cellular assays and molecular dynamics simulations to explain how the membrane environment impacts the dual roles of CD59 in controlling pore formation of MAC, and as a target of bacterial virulence factors which hijack CD59 to lyse human cells.


Asunto(s)
Complemento C9 , Complejo de Ataque a Membrana del Sistema Complemento , Humanos , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Complemento C9/metabolismo , Microscopía por Crioelectrón , Antígenos CD59/metabolismo , Complemento C8/metabolismo , Activación de Complemento
5.
PLoS Biol ; 21(2): e3001962, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36753519

RESUMEN

Macroautophagy/autophagy is an intracellular degradation process central to cellular homeostasis and defense against pathogens in eukaryotic cells. Regulation of autophagy relies on hierarchical binding of autophagy cargo receptors and adaptors to ATG8/LC3 protein family members. Interactions with ATG8/LC3 are typically facilitated by a conserved, short linear sequence, referred to as the ATG8/LC3 interacting motif/region (AIM/LIR), present in autophagy adaptors and receptors as well as pathogen virulence factors targeting host autophagy machinery. Since the canonical AIM/LIR sequence can be found in many proteins, identifying functional AIM/LIR motifs has proven challenging. Here, we show that protein modelling using Alphafold-Multimer (AF2-multimer) identifies both canonical and atypical AIM/LIR motifs with a high level of accuracy. AF2-multimer can be modified to detect additional functional AIM/LIR motifs by using protein sequences with mutations in primary AIM/LIR residues. By combining protein modelling data from AF2-multimer with phylogenetic analysis of protein sequences and protein-protein interaction assays, we demonstrate that AF2-multimer predicts the physiologically relevant AIM motif in the ATG8-interacting protein 2 (ATI-2) as well as the previously uncharacterized noncanonical AIM motif in ATG3 from potato (Solanum tuberosum). AF2-multimer also identified the AIM/LIR motifs in pathogen-encoded virulence factors that target ATG8 members in their plant and human hosts, revealing that cross-kingdom ATG8-LIR/AIM associations can also be predicted by AF2-multimer. We conclude that the AF2-guided discovery of autophagy adaptors/receptors will substantially accelerate our understanding of the molecular basis of autophagy in all biological kingdoms.


Asunto(s)
Furilfuramida , Proteínas Asociadas a Microtúbulos , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Filogenia , Secuencias de Aminoácidos , Familia de las Proteínas 8 Relacionadas con la Autofagia/química , Autofagia/fisiología , Proteínas Portadoras/metabolismo , Unión Proteica
6.
EMBO Rep ; 23(10): e55450, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-35920255

RESUMEN

Interleukin 27 (IL-27) is a heterodimeric cytokine that elicits potent immunosuppressive responses. Comprised of EBI3 and p28 subunits, IL-27 binds GP130 and IL-27Rα receptor chains to activate the JAK/STAT signaling cascade. However, how these receptors recognize IL-27 and form a complex capable of phosphorylating JAK proteins remains unclear. Here, we used cryo electron microscopy (cryoEM) and AlphaFold modeling to solve the structure of the IL-27 receptor recognition complex. Our data show how IL-27 serves as a bridge connecting IL-27Rα (domains 1-2) with GP130 (domains 1-3) to initiate signaling. While both receptors contact the p28 component of the heterodimeric cytokine, EBI3 stabilizes the complex by binding a positively charged surface of IL-27Rα and Domain 1 of GP130. We find that assembly of the IL-27 receptor recognition complex is distinct from both IL-12 and IL-6 cytokine families and provides a mechanistic blueprint for tuning IL-27 pleiotropic actions.


Asunto(s)
Receptor gp130 de Citocinas , Interleucina-27 , Receptores de Interleucina , Receptor gp130 de Citocinas/química , Humanos , Interleucina-12 , Interleucina-27/química , Interleucina-6 , Interleucinas , Receptores de Interleucina/química
7.
Curr Opin Struct Biol ; 75: 102401, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35700576

RESUMEN

Deployed by both pathogenic bacteria and host immune systems, pore-forming proteins rupture target membranes and can serve as conduits for effector proteins. Understanding how these proteins work relies on capturing assembly intermediates. Advances in cryoEM allowing in silico purification of heterogeneous assemblies has led to new insights into two main classes of pore-forming proteins: membrane attack complex perforin (MACPF) proteins and binary toxins. The structure of an immune activation complex, sMAC, shows how pores form by sequential templating and insertion of ß-hairpins. CryoEM structures of bacterial binary toxins present a series of transitions along the pore formation pathway and reveal a general mechanism of effector protein translocation. Future developments in time-resolved cryoEM could capture and place short-lived states along the trajectory of pore-formation.


Asunto(s)
Toxinas Bacterianas , Complejo de Ataque a Membrana del Sistema Complemento , Toxinas Bacterianas/química , Membrana Celular/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/química , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Microscopía por Crioelectrón , Perforina/química , Perforina/metabolismo
8.
Nat Commun ; 12(1): 6086, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34667172

RESUMEN

Unregulated complement activation causes inflammatory and immunological pathologies with consequences for human disease. To prevent bystander damage during an immune response, extracellular chaperones (clusterin and vitronectin) capture and clear soluble precursors to the membrane attack complex (sMAC). However, how these chaperones block further polymerization of MAC and prevent the complex from binding target membranes remains unclear. Here, we address that question by combining cryo electron microscopy (cryoEM) and cross-linking mass spectrometry (XL-MS) to solve the structure of sMAC. Together our data reveal how clusterin recognizes and inhibits polymerizing complement proteins by binding a negatively charged surface of sMAC. Furthermore, we show that the pore-forming C9 protein is trapped in an intermediate conformation whereby only one of its two transmembrane ß-hairpins has unfurled. This structure provides molecular details for immune pore formation and helps explain a complement control mechanism that has potential implications for how cell clearance pathways mediate immune homeostasis.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/química , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Complemento C8/química , Complemento C8/metabolismo , Complemento C9/química , Complemento C9/inmunología , Microscopía por Crioelectrón , Humanos , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios Proteicos
9.
Subcell Biochem ; 96: 273-295, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33252733

RESUMEN

The complement system is essential for immune defence against infection and modulation of proinflammatory responses. Activation of the terminal pathway of complement triggers formation of the membrane attack complex (MAC), a multi-protein pore that punctures membranes. Recent advances in structural biology, specifically cryo-electron microscopy (cryoEM), have provided atomic resolution snapshots along the pore formation pathway. These structures have revealed dramatic conformational rearrangements that enable assembly and membrane rupture. Here we review the structural basis for MAC formation and show how soluble proteins transition into a giant ß-barrel pore. We also discuss regulatory complexes of the terminal pathway and their impact on structure-guided drug discovery of complement therapeutics.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/química , Complejo de Ataque a Membrana del Sistema Complemento/ultraestructura , Diseño de Fármacos , Microscopía por Crioelectrón , Humanos
10.
Nat Commun ; 11(1): 5818, 2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-33199689

RESUMEN

Cholesterol-dependent cytolysins (CDCs) are pore-forming proteins that serve as major virulence factors for pathogenic bacteria. They target eukaryotic cells using different mechanisms, but all require the presence of cholesterol to pierce lipid bilayers. How CDCs use cholesterol to selectively lyse cells is essential for understanding virulence strategies of several pathogenic bacteria, and for repurposing CDCs to kill new cellular targets. Here we address that question by trapping an early state of pore formation for the CDC intermedilysin, bound to the human immune receptor CD59 in a nanodisc model membrane. Our cryo electron microscopy map reveals structural transitions required for oligomerization, which include the lateral movement of a key amphipathic helix. We demonstrate that the charge of this helix is crucial for tuning lytic activity of CDCs. Furthermore, we discover modifications that overcome the requirement of cholesterol for membrane rupture, which may facilitate engineering the target-cell specificity of pore-forming proteins.


Asunto(s)
Membrana Celular/metabolismo , Citotoxinas/química , Citotoxinas/metabolismo , Antígenos CD59/metabolismo , Membrana Celular/ultraestructura , Microscopía por Crioelectrón , Citotoxinas/genética , Humanos , Modelos Biológicos , Modelos Moleculares , Mutación/genética , Estructura Secundaria de Proteína , Relación Estructura-Actividad
11.
Front Immunol ; 11: 585108, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33240274

RESUMEN

The soluble membrane attack complex (sMAC, a.k.a., sC5b-9 or TCC) is generated on activation of complement and contains the complement proteins C5b, C6, C7, C8, C9 together with the regulatory proteins clusterin and/or vitronectin. sMAC is a member of the MACPF/cholesterol-dependent-cytolysin superfamily of pore-forming molecules that insert into lipid bilayers and disrupt cellular integrity and function. sMAC is a unique complement activation macromolecule as it is comprised of several different subunits. To date no complement-mediated function has been identified for sMAC. sMAC is present in blood and other body fluids under homeostatic conditions and there is abundant evidence documenting changes in sMAC levels during infection, autoimmune disease and trauma. Despite decades of scientific interest in sMAC, the mechanisms regulating its formation in healthy individuals and its biological functions in both health and disease remain poorly understood. Here, we review the structural differences between sMAC and its membrane counterpart, MAC, and examine sMAC immunobiology with respect to its presence in body fluids in health and disease. Finally, we discuss the diagnostic potential of sMAC for diagnostic and prognostic applications and potential utility as a companion diagnostic.


Asunto(s)
Activación de Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Animales , Humanos
12.
Proc Natl Acad Sci U S A ; 116(42): 20984-20990, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31570616

RESUMEN

Plants, algae, and cyanobacteria fix carbon dioxide to organic carbon with the Calvin-Benson (CB) cycle. Phosphoribulokinase (PRK) and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) are essential CB-cycle enzymes that control substrate availability for the carboxylation enzyme Rubisco. PRK consumes ATP to produce the Rubisco substrate ribulose bisphosphate (RuBP). GAPDH catalyzes the reduction step of the CB cycle with NADPH to produce the sugar glyceraldehyde 3-phosphate (GAP), which is used for regeneration of RuBP and is the main exit point of the cycle. GAPDH and PRK are coregulated by the redox state of a conditionally disordered protein CP12, which forms a ternary complex with both enzymes. However, the structural basis of CB-cycle regulation by CP12 is unknown. Here, we show how CP12 modulates the activity of both GAPDH and PRK. Using thermophilic cyanobacterial homologs, we solve crystal structures of GAPDH with different cofactors and CP12 bound, and the ternary GAPDH-CP12-PRK complex by electron cryo-microscopy, we reveal that formation of the N-terminal disulfide preorders CP12 prior to binding the PRK active site, which is resolved in complex with CP12. We find that CP12 binding to GAPDH influences substrate accessibility of all GAPDH active sites in the binary and ternary inhibited complexes. Our structural and biochemical data explain how CP12 integrates responses from both redox state and nicotinamide dinucleotide availability to regulate carbon fixation.


Asunto(s)
Proteínas Bacterianas/química , Cianobacterias/enzimología , Gliceraldehído-3-Fosfato Deshidrogenasas/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Fotosíntesis/efectos de la radiación , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cianobacterias/química , Cianobacterias/genética , Cianobacterias/metabolismo , Gliceraldehído 3-Fosfato/metabolismo , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Luz , NADP/química , NADP/metabolismo , Oxidación-Reducción/efectos de la radiación , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Unión Proteica , Ribulosa-Bifosfato Carboxilasa/genética , Ribulosa-Bifosfato Carboxilasa/metabolismo , Thermosynechococcus
13.
Nat Commun ; 10(1): 2066, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-31061395

RESUMEN

The membrane attack complex (MAC) is a hetero-oligomeric protein assembly that kills pathogens by perforating their cell envelopes. The MAC is formed by sequential assembly of soluble complement proteins C5b, C6, C7, C8 and C9, but little is known about the rate-limiting steps in this process. Here, we use rapid atomic force microscopy (AFM) imaging to show that MAC proteins oligomerize within the membrane, unlike structurally homologous bacterial pore-forming toxins. C5b-7 interacts with the lipid bilayer prior to recruiting C8. We discover that incorporation of the first C9 is the kinetic bottleneck of MAC formation, after which rapid C9 oligomerization completes the pore. This defines the kinetic basis for MAC assembly and provides insight into how human cells are protected from bystander damage by the cell surface receptor CD59, which is offered a maximum temporal window to halt the assembly at the point of C9 insertion.


Asunto(s)
Antígenos CD59/metabolismo , Membrana Celular/ultraestructura , Complemento C9/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Multimerización de Proteína , Membrana Celular/metabolismo , Complemento C5/metabolismo , Complemento C8/metabolismo , Humanos , Cinética , Microscopía de Fuerza Atómica/métodos , Imagen Individual de Molécula/métodos
14.
Nat Commun ; 9(1): 5316, 2018 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-30552328

RESUMEN

The membrane attack complex (MAC) is one of the immune system's first responders. Complement proteins assemble on target membranes to form pores that lyse pathogens and impact tissue homeostasis of self-cells. How MAC disrupts the membrane barrier remains unclear. Here we use electron cryo-microscopy and flicker spectroscopy to show that MAC interacts with lipid bilayers in two distinct ways. Whereas C6 and C7 associate with the outer leaflet and reduce the energy for membrane bending, C8 and C9 traverse the bilayer increasing membrane rigidity. CryoEM reconstructions reveal plasticity of the MAC pore and demonstrate how C5b6 acts as a platform, directing assembly of a giant ß-barrel whose structure is supported by a glycan scaffold. Our work provides a structural basis for understanding how ß-pore forming proteins breach the membrane and reveals a mechanism for how MAC kills pathogens and regulates cell functions.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/química , Complejo de Ataque a Membrana del Sistema Complemento/ultraestructura , Microscopía por Crioelectrón/métodos , Membrana Dobles de Lípidos/química , Complemento C6/química , Complemento C6/metabolismo , Complemento C6/ultraestructura , Complemento C7/química , Complemento C7/metabolismo , Complemento C7/ultraestructura , Complemento C8/química , Complemento C8/metabolismo , Complemento C8/ultraestructura , Complemento C9/química , Complemento C9/metabolismo , Complemento C9/ultraestructura , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Membrana Dobles de Lípidos/metabolismo , Liposomas , Modelos Moleculares , Polisacáridos/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Análisis Espectral/métodos
15.
Curr Opin Struct Biol ; 52: 41-49, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30125772

RESUMEN

Deployed by both hosts and pathogens, ß-pore-forming proteins (ß-PFPs) rupture membranes and lyse target cells. Soluble protein monomers oligomerize on the lipid bilayer where they undergo dramatic structural rearrangements, resulting in a transmembrane ß-barrel pore. Advances in electron cryo-microscopy (cryoEM) sample preparation, image detection, and computational algorithms have led to a number of recent structures that reveal a molecular mechanism of pore formation in atomic detail.


Asunto(s)
Microscopía por Crioelectrón , Modelos Moleculares , Proteínas Citotóxicas Formadoras de Poros/química , Antígenos Bacterianos/química , Toxinas Bacterianas/química , Microscopía por Crioelectrón/métodos , Humanos , Conformación Proteica
16.
Semin Cell Dev Biol ; 72: 124-132, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28647534

RESUMEN

The membrane attack complex (MAC) is the pore-forming toxin of the complement system, a relatively early evolutionary acquisition that confers upon complement the capacity to directly kill pathogens. The MAC is more than just a bactericidal missile, having the capacity when formed on self-cells to initiate a host of cell activation events that can have profound consequences for tissue homeostasis in the face of infection or injury. Although the capacity of complement to directly kill pathogens has been recognised for over a century, and the pore-forming killing mechanism for at least 50 years, there remains considerable uncertainty regarding precisely how MAC mediates its killing and cell activation activities. A recent burst of new information on MAC structure provides context and opportunity to re-assess the ways in which MAC kills bacteria and modulates cell functions. In this brief review we will describe key aspects of MAC evolution, function and structure and seek to use the new structural information to better explain how the MAC works.


Asunto(s)
Bacterias/inmunología , Infecciones Bacterianas/inmunología , Membrana Celular/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Animales , Bacterias/clasificación , Infecciones Bacterianas/microbiología , Membrana Celular/metabolismo , Membrana Celular/microbiología , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/ultraestructura , Humanos , Microscopía Electrónica , Modelos Moleculares , Conformación Proteica
17.
Philos Trans R Soc Lond B Biol Sci ; 372(1726)2017 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-28630159

RESUMEN

The membrane attack complex (MAC) is an important innate immune effector of the complement terminal pathway that forms cytotoxic pores on the surface of microbes. Despite many years of research, MAC structure and mechanism of action have remained elusive, relying heavily on modelling and inference from biochemical experiments. Recent advances in structural biology, specifically cryo-electron microscopy, have provided new insights into the molecular mechanism of MAC assembly. Its unique 'split-washer' shape, coupled with an irregular giant ß-barrel architecture, enable an atypical mechanism of hole punching and represent a novel system for which to study pore formation. This review will introduce the complement terminal pathway that leads to formation of the MAC. Moreover, it will discuss how structures of the pore and component proteins underpin a mechanism for MAC function, modulation and inhibition.This article is part of the themed issue 'Membrane pores: from structure and assembly, to medicine and technology'.


Asunto(s)
Membrana Celular/química , Complejo de Ataque a Membrana del Sistema Complemento/química , Microscopía por Crioelectrón , Modelos Moleculares
18.
Sci Rep ; 6: 38446, 2016 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-27910935

RESUMEN

The plasma membrane provides an essential barrier, shielding a cell from the pressures of its external environment. Pore-forming proteins, deployed by both hosts and pathogens alike, breach this barrier to lyse target cells. Intermedilysin is a cholesterol-dependent cytolysin that requires the human immune receptor CD59, in addition to cholesterol, to form giant ß-barrel pores in host membranes. Here we integrate biochemical assays with electron microscopy and atomic force microscopy to distinguish the roles of these two receptors in mediating structural transitions of pore formation. CD59 is required for the specific coordination of intermedilysin (ILY) monomers and for triggering collapse of an oligomeric prepore. Movement of Domain 2 with respect to Domain 3 of ILY is essential for forming a late prepore intermediate that releases CD59, while the role of cholesterol may be limited to insertion of the transmembrane segments. Together these data define a structural timeline for ILY pore formation and suggest a mechanism that is relevant to understanding other pore-forming toxins that also require CD59.


Asunto(s)
Bacteriocinas/metabolismo , Antígenos CD59/metabolismo , Colesterol/metabolismo , Interacciones Huésped-Patógeno , Bacteriocinas/química , Bacteriocinas/genética , Sitios de Unión , Antígenos CD59/química , Antígenos CD59/genética , Membrana Celular/química , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Colesterol/química , Humanos , Microscopía de Fuerza Atómica , Porinas/química , Porinas/genética , Porinas/metabolismo , Dominios Proteicos/genética
19.
Immunol Rev ; 274(1): 141-151, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27782334

RESUMEN

Complement is a key component of innate immunity in health and a powerful driver of inflammation and tissue injury in disease. The biological and pathological effects of complement activation are mediated by activation products. These come in two flavors: (i) proteolytic fragments of complement proteins (C3, C4, C5) generated during activation that bind specific receptors on target cells to mediate effects; (ii) the multimolecular membrane attack complex generated from the five terminal complement proteins that directly binds to and penetrates target cell membranes. Several recent publications have described structural insights that have changed perceptions of the nature of this membrane attack complex. This review will describe these recent advances in understanding of the structure of the membrane attack complex and its by-product the fluid-phase terminal complement complex and relate these new structural insights to functional consequences and cell responses to complement membrane attack.


Asunto(s)
Membrana Celular/metabolismo , Activación de Complemento , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Proteínas del Sistema Complemento/inmunología , Animales , Humanos , Inmunidad Innata , Conformación Proteica , Relación Estructura-Actividad
20.
Sci Rep ; 6: 24656, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27098162

RESUMEN

Polypeptide aggregation into amyloid is linked with several debilitating human diseases. Despite the inherent risk of aggregation-induced cytotoxicity, bacteria control the export of amyloid-prone subunits and assemble adhesive amyloid fibres during biofilm formation. An Escherichia protein, CsgC potently inhibits amyloid formation of curli amyloid proteins. Here we unlock its mechanism of action, and show that CsgC strongly inhibits primary nucleation via electrostatically-guided molecular encounters, which expands the conformational distribution of disordered curli subunits. This delays the formation of higher order intermediates and maintains amyloidogenic subunits in a secretion-competent form. New structural insight also reveal that CsgC is part of diverse family of bacterial amyloid inhibitors. Curli assembly is therefore not only arrested in the periplasm, but the preservation of conformational flexibility also enables efficient secretion to the cell surface. Understanding how bacteria safely handle amyloidogenic polypeptides contribute towards efforts to control aggregation in disease-causing amyloids and amyloid-based biotechnological applications.


Asunto(s)
Amiloide/química , Proteínas de Escherichia coli/química , Chaperonas Moleculares/química , Electricidad Estática , Transporte Activo de Núcleo Celular , Amiloide/clasificación , Amiloide/genética , Amiloide/metabolismo , Proteínas de Escherichia coli/metabolismo , Cinética , Chaperonas Moleculares/metabolismo , Concentración Osmolar , Unión Proteica , Conformación Proteica , Pliegue de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...