Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
EMBO J ; 38(4)2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30643019

RESUMEN

The immune system kills bacteria by the formation of lytic membrane attack complexes (MACs), triggered when complement enzymes cleave C5. At present, it is not understood how the MAC perturbs the composite cell envelope of Gram-negative bacteria. Here, we show that the role of C5 convertase enzymes in MAC assembly extends beyond the cleavage of C5 into the MAC precursor C5b. Although purified MAC complexes generated from preassembled C5b6 perforate artificial lipid membranes and mammalian cells, these components lack bactericidal activity. In order to permeabilize both the bacterial outer and inner membrane and thus kill a bacterium, MACs need to be assembled locally by the C5 convertase enzymes. Our data indicate that C5b6 rapidly loses the capacity to form bactericidal pores; therefore, bacterial killing requires both in situ conversion of C5 and immediate insertion of C5b67 into the membrane. Using flow cytometry and atomic force microscopy, we show that local assembly of C5b6 at the bacterial surface is required for the efficient insertion of MAC pores into bacterial membranes. These studies provide basic molecular insights into MAC assembly and bacterial killing by the immune system.


Asunto(s)
Actividad Bactericida de la Sangre , Membrana Celular/metabolismo , Convertasas de Complemento C3-C5/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Bacterias Gramnegativas/crecimiento & desarrollo , Hemólisis , Permeabilidad de la Membrana Celular , Activación de Complemento , Bacterias Gramnegativas/metabolismo , Humanos
2.
PLoS Pathog ; 16(6): e1008606, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32569291

RESUMEN

An important effector function of the human complement system is to directly kill Gram-negative bacteria via Membrane Attack Complex (MAC) pores. MAC pores are assembled when surface-bound convertase enzymes convert C5 into C5b, which together with C6, C7, C8 and multiple copies of C9 forms a transmembrane pore that damages the bacterial cell envelope. Recently, we found that bacterial killing by MAC pores requires local conversion of C5 by surface-bound convertases. In this study we aimed to understand why local assembly of MAC pores is essential for bacterial killing. Here, we show that rapid interaction of C7 with C5b6 is required to form bactericidal MAC pores on Escherichia coli. Binding experiments with fluorescently labelled C6 show that C7 prevents release of C5b6 from the bacterial surface. Moreover, trypsin shaving experiments and atomic force microscopy revealed that this rapid interaction between C7 and C5b6 is crucial to efficiently anchor C5b-7 to the bacterial cell envelope and form complete MAC pores. Using complement-resistant clinical E. coli strains, we show that bacterial pathogens can prevent complement-dependent killing by interfering with the anchoring of C5b-7. While C5 convertase assembly was unaffected, these resistant strains blocked efficient anchoring of C5b-7 and thus prevented stable insertion of MAC pores into the bacterial cell envelope. Altogether, these findings provide basic molecular insights into how bactericidal MAC pores are assembled and how bacteria evade MAC-dependent killing.


Asunto(s)
Actividad Bactericida de la Sangre , Membrana Celular/metabolismo , Pared Celular/metabolismo , Complemento C5/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Escherichia coli/metabolismo , Proteínas del Sistema Complemento/metabolismo , Células HEK293 , Humanos
3.
Anal Chem ; 92(5): 4082-4093, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-31995983

RESUMEN

Exosomes are endocytic lipid-membrane bound bodies with the potential to be used as biomarkers in cancer and neurodegenerative disease. The limitations and scarcity of current exosome characterization approaches have led to a growing demand for translational techniques, capable of determining their molecular composition and physical properties in physiological fluids. Here, we investigate label-free immunosensing, using a quartz crystal microbalance with dissipation monitoring (QCM-D), to detect exosomes by exploiting their surface protein profile. Exosomes expressing the transmembrane protein CD63 were isolated by size-exclusion chromatography from cell culture media. QCM-D sensors functionalized with anti-CD63 antibodies formed a direct immunoassay toward CD63-positive exosomes in 75% v/v serum, exhibiting a limit-of-detection of 2.9 × 108 and 1.4 × 108 exosome sized particles (ESPs)/mL for frequency and dissipation response, respectively, i.e., clinically relevant concentrations. Our proof-of-concept findings support the adoption of dual-mode acoustic analysis of exosomes, leveraging both frequency and dissipation monitoring for use in bioanalytical characterization.


Asunto(s)
Exosomas/química , Tecnicas de Microbalanza del Cristal de Cuarzo/métodos , Anticuerpos/inmunología , Exosomas/metabolismo , Humanos , Inmunoensayo , Límite de Detección , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Nanopartículas/química , Nanopartículas/metabolismo , Tetraspanina 30/metabolismo
4.
Langmuir ; 31(12): 3678-86, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25742392

RESUMEN

Ceramides are a group of sphingolipids that act as highly important signaling molecules in a variety of cellular processes including differentiation and apoptosis. The predominant in vivo synthetic pathway for ceramide formation is via sphingomyelinase catalyzed hydrolysis of sphingomyelin. The biochemistry of this essential pathway has been studied in detail; however, there is currently a lack of information on the structural behavior of sphingomyelin- and ceramide-rich model membrane systems, which is essential for developing a bottom-up understanding of ceramide signaling and platform formation. We have studied the lyotropic phase behavior of sphingomyelin-ceramide mixtures in excess water as a function of temperature (30-70 °C) and pressure (1-200 MPa) by small- and wide-angle X-ray scattering. At low ceramide concentrations the mixtures form the ripple gel phase (P(ß)') below the gel transition temperature for sphingomyelin, and this observation has been confirmed by atomic force microscopy. Formation of the ripple gel phase can also be induced at higher temperatures via the application of hydrostatic pressure. At high ceramide concentration an inverse hexagonal phase (HII) is formed coexisting with a cubic phase.


Asunto(s)
Ceramidas/química , Transición de Fase , Presión , Esfingomielinas/química , Temperatura , Animales
5.
Soft Matter ; 11(16): 3279-86, 2015 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-25790335

RESUMEN

Lipid bicontinuous cubic phases have attracted enormous interest as bio-compatible scaffolds for use in a wide range of applications including membrane protein crystallisation, drug delivery and biosensing. One of the major bottlenecks that has hindered exploitation of these structures is an inability to create targeted highly swollen bicontinuous cubic structures with large and tunable pore sizes. In contrast, cubic structures found in vivo have periodicities approaching the micron scale. We have been able to engineer and control highly swollen bicontinuous cubic phases of spacegroup Im3m containing only lipids by (a) increasing the bilayer stiffness by adding cholesterol and (b) inducing electrostatic repulsion across the water channels by addition of anionic lipids to monoolein. By controlling the composition of the ternary mixtures we have been able to achieve lattice parameters up to 470 Å, which is 5 times that observed in pure monoolein and nearly twice the size of any lipidic cubic phase reported previously. These lattice parameters significantly exceed the predicted maximum swelling for bicontinuous cubic lipid structures, which suggest that thermal fluctuations should destroy such phases for lattice parameters larger than 300 Å.


Asunto(s)
Lípidos/química , Colesterol/química , Glicéridos/química , Fosfatidilgliceroles/química , Electricidad Estática , Agua/química
6.
Methods Mol Biol ; 2208: 225-235, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32856266

RESUMEN

Recent advances in biomolecular design require accurate measurements performed in native or near-native environments in real time. Atomic force microscopy (AFM) is a powerful tool to observe the dynamics of biologically relevant processes at aqueous interfaces with high spatial resolution. Here, we describe imaging protocols to characterize the effects of peptide materials on phospholipid membranes in solution by AFM. These protocols can be used to determine the mechanism and kinetics of membrane-associated activities at the nanoscale.


Asunto(s)
Membranas/química , Microscopía de Fuerza Atómica/métodos , Péptidos/química , Fosfolípidos/química , Cinética
7.
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
8.
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
9.
Nat Commun ; 10(1): 4288, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31537793

RESUMEN

Macrophage-expressed gene 1 (MPEG1/Perforin-2) is a perforin-like protein that functions within the phagolysosome to damage engulfed microbes. MPEG1 is thought to form pores in target membranes, however, its mode of action remains unknown. We use cryo-Electron Microscopy (cryo-EM) to determine the 2.4 Å structure of a hexadecameric assembly of MPEG1 that displays the expected features of a soluble prepore complex. We further discover that MPEG1 prepore-like assemblies can be induced to perforate membranes through acidification, such as would occur within maturing phagolysosomes. We next solve the 3.6 Å cryo-EM structure of MPEG1 in complex with liposomes. These data reveal that a multi-vesicular body of 12 kDa (MVB12)-associated ß-prism (MABP) domain binds membranes such that the pore-forming machinery of MPEG1 is oriented away from the bound membrane. This unexpected mechanism of membrane interaction suggests that MPEG1 remains bound to the phagolysosome membrane while simultaneously forming pores in engulfed bacterial targets.


Asunto(s)
Membrana Celular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Bacterias/inmunología , Microscopía por Crioelectrón , Humanos , Liposomas/metabolismo , Lisosomas/fisiología , Macrófagos/inmunología , Microscopía de Fuerza Atómica , Dominios Proteicos , Estructura Secundaria de Proteína
10.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA