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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
EMBO J ; 39(23): e105753, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33124082

RESUMO

The discovery of alternative signaling pathways that regulate cell death has revealed multiple strategies for promoting cell death with diverse consequences at the tissue and organism level. Despite the divergence in the molecular components involved, membrane permeabilization is a common theme in the execution of regulated cell death. In apoptosis, the permeabilization of the outer mitochondrial membrane by BAX and BAK releases apoptotic factors that initiate the caspase cascade and is considered the point of no return in cell death commitment. Pyroptosis and necroptosis also require the perforation of the plasma membrane at the execution step, which involves Gasdermins in pyroptosis, and MLKL in the case of necroptosis. Although BAX/BAK, Gasdermins and MLKL share certain molecular features like oligomerization, they form pores in different cellular membranes via distinct mechanisms. Here, we compare and contrast how BAX/BAK, Gasdermins, and MLKL alter membrane permeability from a structural and biophysical perspective and discuss the general principles of membrane permeabilization in the execution of regulated cell death.


Assuntos
Morte Celular/imunologia , Morte Celular/fisiologia , Morte Celular Regulada/imunologia , Morte Celular Regulada/fisiologia , Animais , Apoptose/fisiologia , Autofagia/fisiologia , Caspases/metabolismo , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular , Humanos , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Necroptose/fisiologia , Proteínas Quinases/metabolismo , Piroptose/fisiologia , Transdução de Sinais/fisiologia
2.
Int J Mol Sci ; 24(12)2023 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-37373257

RESUMO

Necroptosis is a recently discovered form of regulated cell death characterized by the disruption of plasma membrane integrity and the release of intracellular content. Mixed lineage kinase domain-like (MLKL) protein is the main player of this cell death pathway as it mediates the final step of plasma membrane permeabilization. Despite the significant progress in our knowledge of the necroptotic pathway and MLKL biology, the precise mechanism of how MLKL functions remain unclear. To understand in what way MLKL executes necroptosis, it is crucial to decipher how the molecular machinery of regulated cell death is activated in response to different stimuli or stressors. It is also indispensable to unveiling the structural elements of MLKL and the cellular players that are required for its regulation. In this review, we discuss the key steps that lead to MLKL activation, possible models that explain how it becomes the death executor in necroptosis, and its emerging alternative functions. We also summarize the current knowledge about the role of MLKL in human disease and provide an overview of existing strategies aimed at developing new inhibitors that target MLKL for necroptosis intervention.


Assuntos
Apoptose , Proteínas Quinases , Humanos , Apoptose/fisiologia , Proteínas Quinases/metabolismo , Necroptose , Morte Celular , Membrana Celular/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo
3.
Int J Mol Sci ; 21(7)2020 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-32244433

RESUMO

Pyroptosis, necroptosis, and ferroptosis are well-characterized forms of regulated necrosis that have been associated with human diseases. During regulated necrosis, plasma membrane damage facilitates the movement of ions and molecules across the bilayer, which finally leads to cell lysis and release of intracellular content. Therefore, these types of cell death have an inflammatory phenotype. Each type of regulated necrosis is mediated by a defined machinery comprising protein and lipid molecules. Here, we discuss how the interaction and reshaping of these cellular components are essential and distinctive processes during pyroptosis, necroptosis, and ferroptosis. We point out that although the plasma membrane is the common target in regulated necrosis, different mechanisms of permeabilization have emerged depending on the cell death form. Pore formation by gasdermins (GSDMs) is a hallmark of pyroptosis, while mixed lineage kinase domain-like (MLKL) protein facilitates membrane permeabilization in necroptosis, and phospholipid peroxidation leads to membrane damage in ferroptosis. This diverse repertoire of mechanisms leading to membrane permeabilization contributes to define the specific inflammatory and immunological outcome of each type of regulated necrosis. Current efforts are focused on new therapies that target critical protein and lipid molecules on these pathways to fight human pathologies associated with inflammation.


Assuntos
Membranas/metabolismo , Necrose/metabolismo , Proteínas/metabolismo , Animais , Apoptose/fisiologia , Morte Celular , Membrana Celular/metabolismo , Ferroptose/fisiologia , Humanos , Inflamação , Lipídeos , Necroptose/fisiologia , Permeabilidade , Piroptose
4.
Biophys J ; 117(9): 1563-1576, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31587828

RESUMO

Sticholysins are pore-forming toxins of biomedical interest and represent a prototype of proteins acting through the formation of protein-lipid or toroidal pores. Peptides spanning the N-terminus of sticholysins can mimic their permeabilizing activity and, together with the full-length toxins, have been used as a tool to understand the mechanism of pore formation in membranes. However, the lytic mechanism of these peptides and the lipid shape modulating their activity are not completely clear. In this article, we combine molecular dynamics simulations and experimental biophysical tools to dissect different aspects of the pore-forming mechanism of StII1-30, a peptide derived from the N-terminus of sticholysin II (StII). With this combined approach, membrane curvature induction and flip-flop movement of the lipids were identified as two important membrane remodeling steps mediated by StII1-30. Pore formation by this peptide was enhanced by the presence of the negatively curved lipid phosphatidylethanolamine in membranes. This lipid emerged not only as a facilitator of membrane interactions but also as a structural element of the StII1-30 pore that is recruited to the ring upon its assembly. Collectively, these, to our knowledge, new findings support a toroidal model for the architecture of the pore formed by StII1-30 and provide new molecular insight into the role of phosphatidylethanolamine as a membrane component that can easily integrate into the ring of toroidal pores, thus probably aiding in their stabilization. This study contributes to a better understanding of the molecular mechanism underlying the permeabilizing activity of StII1-30 and peptides or proteins acting via a toroidal pore mechanism and offers an informative framework for the optimization of the biomedical application of this and similar molecules.


Assuntos
Membrana Celular/metabolismo , Venenos de Cnidários/metabolismo , Modelos Moleculares , Sequência de Aminoácidos , Animais , Venenos de Cnidários/química , Bicamadas Lipídicas/química , Simulação de Dinâmica Molecular , Permeabilidade , Fosfatidiletanolaminas/química , Soluções , Suínos
5.
Biopolymers ; 2018 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-29359791

RESUMO

Sticholysin II (StII) is a pore-forming actinoporin from the sea anemone Stichodactyla helianthus. A mechanistic model of its action has been proposed: proteins bind to cell membrane, insert their N-termini into the lipid core and assemble into homo-tetramer pores responsible for host-cell death. Because very likely the first 10 residues of StII N-terminus are critical for membrane penetration, to dissect the molecular details of that functionality, we studied two synthetic peptides: StII1-30 and StII16-35 . They show diverse haemolytic and candidacidal activity that correlate with distinct orientations in SDS micelles. NMR shows that StII1-30 partly inserts into the micelle, while StII16-35 lays on the micelle surface. These results justify the diverse concentration dependence of their candidacidal activity supposing a different mechanism of action and providing new hints on StII lytic activity at molecular level. Biotechnological application of these peptides, focused on the development of therapeutic immunocomplexes, may be envisaged.

6.
Biochim Biophys Acta ; 1858(3): 457-466, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26375417

RESUMO

Pore forming proteins (PFPs) share the ability of creating pores that allow the passage of ions, proteins or other constituents through a wide variety of target membranes, ranging from bacteria to humans. They often cause cell death, as pore formation disrupts the membrane permeability barrier required for maintaining cell homeostasis. The organization into supramolecular complexes or oligomers that pierce the membrane is a common feature of PFPs. However, the molecular pathway of self-assembly and pore opening remains unclear. Here, we review the most recent discoveries in the mechanism of membrane oligomerization and pore formation of a subset of PFPs, the α-PFPs, whose pore-forming domains are formed by helical segments. Only now we are starting to grasp the molecular details of their function, mainly thanks to the introduction of single molecule microscopy and nanoscopy techniques. This article is part of a Special Issue entitled: Pore-forming toxins edited by Mauro Dalla Serra and Franco Gambale.


Assuntos
Membrana Celular/química , Porinas/química , Multimerização Proteica , Animais , Membrana Celular/metabolismo , Humanos , Porinas/metabolismo , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
7.
Biochim Biophys Acta Biomembr ; 1859(5): 982-992, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28173991

RESUMO

Pore-forming toxins (PFTs) form holes in membranes causing one of the most catastrophic damages to a target cell. Target organisms have evolved a regulated response against PFTs damage including cell membrane repair. This ability of cells strongly depends on the toxin concentration and the properties of the pores. It has been hypothesized that there is an inverse correlation between the size of the pores and the time required to repair the membrane, which has been for long a non-intuitive concept and far to be completely understood. Moreover, there is a lack of information about how cells react to the injury triggered by eukaryotic PFTs. Here, we investigated some molecular events related with eukaryotic cells response against the membrane damage caused by sticholysin II (StII), a eukaryotic PFT produced by a sea anemone. We evaluated the change in the cytoplasmic potassium, identified the main MAPK pathways activated after pore-formation by StII, and compared its effect with those from two well-studied bacterial PFTs: aerolysin and listeriolysin O (LLO). Strikingly, we found that membrane recovery upon StII damage takes place in a time scale similar to LLO in spite of the fact that they form pores by far different in size. Furthermore, our data support a common role of the potassium ion, as well as MAPKs in the mechanism that cells use to cope with these toxins injury.


Assuntos
Venenos de Cnidários/toxicidade , Células Eucarióticas/efeitos dos fármacos , Proteínas Citotóxicas Formadoras de Poros/toxicidade , Potássio/metabolismo , Anêmonas-do-Mar/patogenicidade , Animais , Células Cultivadas , Cricetinae , Células Eucarióticas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
8.
J Biol Chem ; 290(8): 4856-4865, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25525270

RESUMO

α-Pore-forming toxins (α-PFTs) are ubiquitous defense tools that kill cells by opening pores in the target cell membrane. Despite their relevance in host/pathogen interactions, very little is known about the pore stoichiometry and assembly pathway leading to membrane permeabilization. Equinatoxin II (EqtII) is a model α-PFT from sea anemone that oligomerizes and forms pores in sphingomyelin-containing membranes. Here, we determined the spatiotemporal organization of EqtII in living cells by single molecule imaging. Surprisingly, we found that on the cell surface EqtII did not organize into a unique oligomeric form. Instead, it existed as a mixture of oligomeric species mostly including monomers, dimers, tetramers, and hexamers. Mathematical modeling based on our data supported a new model in which toxin clustering happened in seconds and proceeded via condensation of EqtII dimer units formed upon monomer association. Furthermore, altering the pathway of EqtII assembly strongly affected its toxic activity, which highlights the relevance of the assembly mechanism on toxicity.


Assuntos
Permeabilidade da Membrana Celular , Venenos de Cnidários/química , Membrana Eritrocítica/química , Proteínas Citotóxicas Formadoras de Poros/química , Multimerização Proteica , Hemólise , Humanos
9.
Biochim Biophys Acta ; 1838(7): 1752-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24680653

RESUMO

Sticholysin I (St I) is a pore-forming toxin (PFT) produced by the Caribbean Sea anemone Stichodactyla helianthus belonging to the actinoporin protein family, a unique class of eukaryotic PFT exclusively found in sea anemones. As for actinoporins, it has been proposed that the presence of sphingomyelin (SM) and the coexistence of lipid phases increase binding to the target membrane. However, little is known about the role of membrane structure and dynamics (phase state, fluidity, presence of lipid domains) on actinoporins' activity or which regions of the membrane are the most favorable platforms for protein insertion. To gain insight into the role of SM on the interaction of St I to lipid membranes we studied their binding to monolayers of phosphatidylcholine (PC) and SM in different proportions. Additionally, the effect of acyl chain length and unsaturation, two features related to membrane fluidity, was evaluated on St I binding to monolayers. This study revealed that St I binds and penetrates preferentially and with a faster kinetic to liquid-expanded films with high lateral mobility and moderately enriched in SM. A high content of SM induces a lower lateral diffusion and/or liquid-condensed phases, which hinder St I binding and penetration to the lipid monolayer. Furthermore, the presence of lipid domain borders does not appear as an important factor for St I binding to the lipid monolayer.


Assuntos
Fluidez de Membrana/fisiologia , Lipídeos de Membrana/metabolismo , Esfingomielinas/metabolismo , Animais , Venenos de Cnidários/metabolismo , Cinética , Membranas/metabolismo , Compostos Orgânicos/metabolismo , Fosfatidilcolinas/metabolismo , Anêmonas-do-Mar
10.
J Membr Biol ; 248(3): 545-61, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26087906

RESUMO

Pore-forming proteins (PFPs) punch holes in their target cell membrane to alter their permeability. Permeabilization of lipid membranes by PFPs has received special attention to study the basic molecular mechanisms of protein insertion into membranes and the development of biotechnological tools. PFPs act through a general multi-step mechanism that involves (i) membrane partitioning, (ii) insertion into the hydrophobic core of the bilayer, (iii) oligomerization, and (iv) pore formation. Interestingly, PFPs and membranes show a dynamic interplay. As PFPs are usually produced as soluble proteins, they require a large conformational change for membrane insertion. Moreover, membrane structure is modified upon PFPs insertion. In this context, the toroidal pore model has been proposed to describe a pore architecture in which not only protein molecules but also lipids are directly involved in the structure. Here, we discuss how PFPs and lipids cooperate and remodel each other to achieve pore formation, and explore new evidences of protein-lipid pore structures.


Assuntos
Membrana Celular/química , Proteínas Citotóxicas Formadoras de Poros/química , Animais , Permeabilidade da Membrana Celular , Humanos , Interações Hidrofóbicas e Hidrofílicas , Lipídeos de Membrana/química , Modelos Moleculares , Porosidade , Estrutura Secundária de Proteína
11.
Langmuir ; 31(36): 9911-23, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26273899

RESUMO

Sticholysin I (St I) is a pore-forming toxin (PFT) produced by the Caribbean Sea anemone Stichodactyla helianthus belonging to the actinoporin protein family, a unique class of eukaryotic PFT. As for actinoporins, it has been proposed that the presence of cholesterol (Chol) and the coexistence of lipid phases increase binding to the target membrane and pore-forming ability. However, little is known about the role of membrane structure and dynamics (phase state, fluidity, and the presence of lipid domains) on the activity of actinoporins or which regions of the membrane are the most favorable for protein insertion, oligomerization, and eventually pore formation. To gain insight into the role of membrane properties on the functional activity of St I, we studied its binding to monolayers and vesicles of phosphatidylcholine (PC), sphingomyelin (SM), and sterols inducing (ergosterol -Erg and cholesterol -Chol) or not (cholestenone - Cln) membrane phase segregation in liquid ordered (Lo) and liquid disordered (Ld) domains. This study revealed that St I binds and permeabilizes with higher efficiency sterol-containing membranes independently of their ability to form domains. We discuss the results in terms of the relevance of different membrane properties for the actinoporins mechanism of action, namely, molecular heterogeneity, specially potentiated in membranes with sterols inducers of phase separation (Chol or Erg) or Cln, a sterol noninducer of phase separation but with a high propensity to induce nonlamellar phase. The role of the Ld phase is pointed out as the most suitable platform for pore formation. In this regard, such regions in Chol-containing membranes seem to be the most favored due to its increased fluidity; this property promotes toxin insertion, diffusion, and oligomerization leading to pore formation.


Assuntos
Esteróis/química , Compostos Orgânicos/química , Lipossomas Unilamelares
12.
Biochim Biophys Acta ; 1828(11): 2757-62, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23954588

RESUMO

Sticholysins (Sts) I and II (StI/II) are pore-forming toxins (PFTs) produced by the Caribbean Sea anemone Stichodactyla helianthus belonging to the actinoporin family, a unique class of eukaryotic PFTs exclusively found in sea anemones. The role of lipid phase co-existence in the mechanism of the action of membranolytic proteins and peptides is not clearly understood. As for actinoporins, it has been proposed that phase separation promotes pore forming activity. However little is known about the effect of sticholysins on the phase separation of lipids in membranes. To gain insight into the mechanism of action of sticholysins, we evaluated the effect of these proteins on lipid segregation using differential scanning calorimetry (DSC) and atomic force microscopy (AFM). New evidence was obtained reflecting that these proteins reduce line tension in the membrane by promoting lipid mixing. In terms of the relevance for the mechanism of action of actinoporins, we hypothesize that expanding lipid disordered phases into lipid ordered phases decreases the lipid packing at the borders of the lipid raft, turning it into a more suitable environment for N-terminal insertion and pore formation.


Assuntos
Venenos de Cnidários/farmacologia , Lipídeos/química , Microdomínios da Membrana/metabolismo , Anêmonas-do-Mar/metabolismo , Animais , Varredura Diferencial de Calorimetria , Microscopia de Força Atômica , Compostos Orgânicos/farmacologia , Anêmonas-do-Mar/química
13.
Biopolymers ; 100(4): 337-46, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23868208

RESUMO

Sticholysin II (St II) is the most potent cytolysin produced by the sea anemone Stichodactyla helianthus, exerting hemolytic activity via pore formation in membranes. The toxin's N-terminus contains an amphipathic α-helix that is very likely involved in pore formation. We have previously demonstrated that the synthetic peptide StII(1-30) encompassing the 1-30 segment of St II forms pores of similar radius to that of the protein (around 1 nm), being a good model of toxin functionality. Here we have studied the functional and conformational properties of fluorescent analogs of StII(1-30) in lipid membranes. The analogs were obtained by replacing Leu residues at positions 2, 12, 17, and 24 with the intrinsically fluorescent amino acid Trp (StII(1-30L2W), StII(1-30L12W), StII(1-30L17W), or StII(1-30L24W), respectively). The exchange by Trp did not significantly modify the activity and conformation of the parent peptide. The blue-shift and intensity enhancement of fluorescence in the presence of membrane indicated that Trp at position 2 is more deeply buried in the hydrophobic region of the bilayer. These experiments, as well as assays with water-soluble or spin-labeled lipid-soluble fluorescence quenchers suggest an orientation of StII(1-30) with its N-terminus oriented towards the hydrophobic core of the bilayer while the rest of the peptide is more exposed to the aqueous environment, as hypothesized for sticholysins.


Assuntos
Compostos Orgânicos , Anêmonas-do-Mar , Sequência de Aminoácidos , Animais , Lipídeos de Membrana , Dados de Sequência Molecular , Peptídeos/química , Estrutura Secundária de Proteína
14.
Cell Calcium ; 114: 102778, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37356350

RESUMO

Regulation of proliferation and cell death is fundamental for organismal development and for restoring tissue homeostasis after biological stress. During the last years, several forms of regulated cell death have been discovered that share the loss of plasma membrane integrity as a common hallmark and that are collectively known as regulated necrosis (RN) pathways. During RN, plasma membrane damage is sensed by the cell by increases in the levels of intracellular calcium. Interestingly, cytosolic calcium influx can either lead to cell death or survival, given the versatile role of this ion in regulating multiple signaling processes. Among them, membrane repair enables the cells to tolerate the injury and, even in some conditions, survive. Here, we review calcium signaling in the context of RN pathways, with a focus on ferroptosis, a type of RN in which plasma membrane damage is elicited by the accumulation of oxidized lipids. In contrast, other forms of RN such as necroptosis and pyroptosis require dedicated pore-forming proteins for plasma membrane damage and cell death. We first focus on the current knowledge regarding the contribution of calcium to ferroptosis, and then illustrate the similarities and differences in calcium signaling with necroptosis and pyroptosis. Calcium signaling emerges as a key event in the cellular responses to membrane damage and in the regulation of cell death.


Assuntos
Cálcio , Ferroptose , Humanos , Necrose/metabolismo , Apoptose , Morte Celular
15.
Toxins (Basel) ; 15(1)2023 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-36668899

RESUMO

Actinoporins have emerged as archetypal α-pore-forming toxins (PFTs) that promote the formation of pores in membranes upon oligomerization and insertion of an α-helix pore-forming domain in the bilayer. These proteins have been used as active components of immunotoxins, therefore, understanding their lytic mechanism is crucial for developing this and other applications. However, the mechanism of how the biophysical properties of the membrane modulate the properties of pores generated by actinoporins remains unclear. Here we studied the effect of membrane fluidity on the permeabilizing activity of sticholysin I (St I), a toxin that belongs to the actinoporins family of α-PFTs. To modulate membrane fluidity we used vesicles made of an equimolar mixture of phosphatidylcholine (PC) and egg sphingomyelin (eggSM), in which PC contained fatty acids of different acyl chain lengths and degrees of unsaturation. Our detailed single-vesicle analysis revealed that when membrane fluidity is high, most of the vesicles are partially permeabilized in a graded manner. In contrast, more rigid membranes can be either completely permeabilized or not, indicating an all-or-none mechanism. Altogether, our results reveal that St I pores can be heterogeneous in size and stability, and that these properties depend on the fluid state of the lipid bilayer. We propose that membrane fluidity at different regions of cellular membranes is a key factor to modulate the activity of the actinoporins, which has implications for the design of different therapeutic strategies based on their lytic action.


Assuntos
Venenos de Cnidários , Anêmonas-do-Mar , Animais , Fluidez de Membrana , Compostos Orgânicos/química , Bicamadas Lipídicas , Membrana Celular/metabolismo , Fosfatidilcolinas , Venenos de Cnidários/química , Anêmonas-do-Mar/química
16.
J Phys Chem Lett ; 13(3): 822-829, 2022 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-35044771

RESUMO

Analysis of single-molecule brightness allows subunit counting of high-order oligomeric biomolecular complexes. Although the theory behind the method has been extensively assessed, systematic analysis of the experimental conditions required to accurately quantify the stoichiometry of biological complexes remains challenging. In this work, we develop a high-throughput, automated computational pipeline for single-molecule brightness analysis that requires minimal human input. We use this strategy to systematically quantify the accuracy of counting under a wide range of experimental conditions in simulated ground-truth data and then validate its use on experimentally obtained data. Our approach defines a set of conditions under which subunit counting by brightness analysis is designed to work optimally and helps in establishing the experimental limits in quantifying the number of subunits in a complex of interest. Finally, we combine these features into a powerful, yet simple, software that can be easily used for the analysis of the stoichiometry of such complexes.


Assuntos
Imagem Individual de Molécula
17.
Curr Opin Struct Biol ; 69: 108-116, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33945958

RESUMO

Pore-forming proteins (PFPs) are of special interest because of the association of their activity with the disruption of the membrane impermeability barrier and cell death. They generally convert from a monomeric, soluble form into transmembrane oligomers that induce the opening of membrane pores. The study of pore formation in membranes with molecular detail remains a challenging endeavor because of its highly dynamic and complex nature, usually involving diverse oligomeric structures with different functionalities. Here we discuss current methods applied for the structural and functional characterization of PFPs at the individual vesicle and cell level. We highlight how the development of high-resolution and single-molecule imaging techniques allows the analysis of the structural organization of protein oligomers and pore entities in lipid membranes.


Assuntos
Porinas , Membrana Celular , Substâncias Macromoleculares
18.
Chem Phys Lipids ; 234: 105026, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33309552

RESUMO

Pore-forming proteins (PFPs) and small antimicrobial peptides (AMPs) represent a large family of molecules with the common ability to punch holes in cell membranes to alter their permeability. They play a fundamental role as infectious bacteria's defensive tools against host's immune system and as executors of endogenous machineries of regulated cell death in eukaryotic cells. Despite being highly divergent in primary sequence and 3D structure, specific folds of pore-forming domains have been conserved. In fact, pore formation is considered an ancient mechanism that takes place through a general multistep process involving: membrane partitioning and insertion, oligomerization and pore formation. However, different PFPs and AMPs assemble and form pores following different mechanisms that could end up either in the formation of protein-lined or protein-lipid pores. In this review, we analyze the current findings in the mechanism of action of different PFPs and AMPs that support a wide role of membrane pore formation in nature. We also provide the newest insights into the development of state-of-art techniques that have facilitated the characterization of membrane pores. To understand the physiological role of these peptides/proteins or develop clinical applications, it is essential to uncover the molecular mechanism of how they perforate membranes.


Assuntos
Perforina/metabolismo , Morte Celular , Membrana Celular/metabolismo , Humanos , Proteínas Citotóxicas Formadoras de Poros/metabolismo
19.
Cell Death Differ ; 28(5): 1644-1657, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33335287

RESUMO

Ferroptosis is an iron-dependent form of regulated necrosis associated with lipid peroxidation. Despite its key role in the inflammatory outcome of ferroptosis, little is known about the molecular events leading to the disruption of the plasma membrane during this type of cell death. Here we show that a sustained increase in cytosolic Ca2+ is a hallmark of ferroptosis that precedes complete bursting of the cell. We report that plasma membrane damage leading to ferroptosis is associated with membrane nanopores of a few nanometers in radius and that ferroptosis, but not lipid peroxidation, can be delayed by osmoprotectants. Importantly, Ca2+ fluxes during ferroptosis induce the activation of the ESCRT-III-dependent membrane repair machinery, which counterbalances the kinetics of cell death and modulates the immunological signature of ferroptosis. Our findings with ferroptosis provide a unifying concept that sustained increase of cytosolic Ca2+ prior to plasma membrane rupture is a common feature of regulated types of necrosis and position ESCRT-III activation as a general protective mechanism in these lytic cell death pathways.


Assuntos
Cálcio/metabolismo , Morte Celular/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Ferroptose/genética , Humanos , Cinética , Transfecção
20.
Toxins (Basel) ; 13(9)2021 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-34564674

RESUMO

Equinatoxin II (EqtII) and Fragaceatoxin C (FraC) are pore-forming toxins (PFTs) from the actinoporin family that have enhanced membrane affinity in the presence of sphingomyelin (SM) and phase coexistence in the membrane. However, little is known about the effect of these proteins on the nanoscopic properties of membrane domains. Here, we used combined confocal microscopy and force mapping by atomic force microscopy to study the effect of EqtII and FraC on the organization of phase-separated phosphatidylcholine/SM/cholesterol membranes. To this aim, we developed a fast, high-throughput processing tool to correlate structural and nano-mechanical information from force mapping. We found that both proteins changed the lipid domain shape. Strikingly, they induced a reduction in the domain area and circularity, suggesting a decrease in the line tension due to a lipid phase height mismatch, which correlated with proteins binding to the domain interfaces. Moreover, force mapping suggested that the proteins affected the mechanical properties at the edge, but not in the bulk, of the domains. This effect could not be revealed by ensemble force spectroscopy measurements supporting the suitability of force mapping to study local membrane topographical and mechanical alterations by membranotropic proteins.


Assuntos
Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Venenos de Cnidários/metabolismo , Venenos de Cnidários/toxicidade , Microdomínios da Membrana/metabolismo , Anêmonas-do-Mar/química , Anêmonas-do-Mar/metabolismo , Esfingomielinas/metabolismo , Animais , Microdomínios da Membrana/efeitos dos fármacos , Microscopia de Força Atômica , Microscopia Confocal
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA