Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Photochem Photobiol ; 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38752609

RESUMEN

Large Stokes shift red fluorescent proteins (LSS-RFPs) are genetically encoded and exhibit a significant difference of a few hundreds of nanometers between their excitation and emission peak maxima (i.e., the Stokes shift). These LSS-RFPs (absorbing blue light and emitting red light) feature a unique photocycle responsible for their significant Stokes shift. The photocycle associated with this LSS characteristic in certain RFPs is quite perplexing, hinting at the complex nature of excited-state photophysics. This article provides a brief review on the fundamental mechanisms governing the photocycle of various LSS-RFPs, followed by a discussion on experimental results on mKeima emphasizing its relaxation pathways which garnered attention due to its >200 nm Stokes shift. Corroborating steady-state spectroscopy with computational studies, four different forms of chromophore of mKeima contributing to the cis-trans conformers of the neutral and anionic forms were identified in a recent study. Furthering these findings, in this account a detailed discussion on the photocycle of mKeima, which encompasses sequential excited-state isomerization, proton transfer, and subsequent structural reorganization involving three isomers, leading to an intriguing temperature and pH-dependent dual fluorescence, is explored using broadband femtosecond transient absorption spectroscopy.

2.
Mol Microbiol ; 120(6): 845-873, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37818865

RESUMEN

Thermostable direct haemolysin (TDH) is the key virulence factor secreted by the human gastroenteric bacterial pathogen Vibrio parahaemolyticus. TDH is a membrane-damaging pore-forming toxin. It evokes potent cytotoxicity, the mechanism of which still remains under-explored. Here, we have elucidated the mechanistic details of cell death response elicited by TDH. Employing Caco-2 intestinal epithelial cells and THP-1 monocytic cells, we show that TDH induces some of the hallmark features of apoptosis-like programmed cell death. TDH triggers caspase-3 and 7 activations in the THP-1 cells, while caspase-7 activation is observed in the Caco-2 cells. Interestingly, TDH appears to induce caspase-independent cell death. Higher XIAP level and lower Smac/Diablo level upon TDH intoxication provide plausible explanation for the functional inability of caspases in the THP-1 cells, in particular. Further exploration reveals that mitochondria play a central role in the TDH-induced cell death. TDH triggers mitochondrial damage, resulting in the release of AIF and endonuclease G, responsible for the execution of caspase-independent cell death. Among the other critical mediators of cell death, ROS is found to play an important role in the THP-1 cells, while PARP-1 appears to play a critical role in the Caco-2 cells. Altogether, our work provides critical new insights into the mechanism of cell death induction by TDH, showing a common central theme of non-classical programmed cell death. Our study also unravels the interplay of crucial molecules in the underlying signalling processes. Our findings add valuable insights into the role of TDH in the context of the host-pathogen interaction processes.


Asunto(s)
Vibrio parahaemolyticus , Humanos , Células CACO-2 , Apoptosis , Caspasas
3.
J Phys Chem B ; 127(14): 3197-3207, 2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-37014205

RESUMEN

Elucidating the origin of large Stokes shift (LSS) in certain fluorescent proteins absorbing in blue/blue-green and emitting in red/far-red has been quite illusive. Using a combination of spectroscopic measurements, corroborated by theoretical calculations, the presence of four distinct forms of the chromophore of the red fluorescent protein mKeima is confirmed, two of which are found to be emissive: a feeble bluish-green fluorescence (∼520 nm), which is enhanced appreciably in a low pH or deuterated medium but significantly at cryogenic temperatures, and a strong emission in red (∼615 nm). Using femtosecond transient absorption spectroscopy, the trans-protonated form is found to isomerize within hundreds of femtoseconds to the cis-protonated form, which further yields the cis-deprotonated form within picoseconds followed by structural reorganization of the local environment of the chromophore. Thus, the mechanism of LSS is substantiated to proceed via stepwise excited-state isomerization followed by proton transfer involving three isomers, leaving the fourth one (trans-deprotonated) as a bystander. The exquisite pH sensitivity of the dual emission is further exploited in fluorescence microscopy.


Asunto(s)
Protones , Isomerismo , Proteínas Luminiscentes/química , Análisis Espectral , Temperatura , Concentración de Iones de Hidrógeno , Proteínas Fluorescentes Verdes/química
4.
Proteins ; 91(2): 137-146, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36000388

RESUMEN

Thermostable direct hemolysin (TDH) is a ~19 kDa, hemolytic pore-forming toxin from the gram-negative marine bacterium Vibrio parahaemolyticus, one of the causative agents of seafood-borne acute gastroenteritis and septicemia. Previous studies have established that TDH exists as a tetrameric assembly in physiological state; however, there is limited knowledge regarding the molecular arrangement of its disordered N-terminal region (NTR)-the absence of which has been shown to compromise TDH's hemolytic and cytotoxic abilities. In our current study, we have employed single-particle cryo-electron microscopy to resolve the solution-state structures of wild-type TDH and a TDH construct with deletion of the NTR (NTD), in order to investigate structural aspects of NTR on the overall tetrameric architecture. We observed that both TDH and NTD electron density maps, resolved at global resolutions of 4.5 and 4.2 Å, respectively, showed good correlation in their respective oligomeric architecture. Additionally, we were able to locate extra densities near the pore opening of TDH which might correspond to the disordered NTR. Surprisingly, under cryogenic conditions, we were also able to observe novel supramolecular assemblies of TDH tetramers, which we were able to resolve to 4.3 Å. We further investigated the tetrameric and inter-tetrameric interaction interfaces to elaborate upon the key residues involved in both TDH tetramers and TDH super assemblies. Our current structural study will aid in understanding the mechanistic aspects of this pore-forming toxin and the role of its disordered NTR in membrane interaction.


Asunto(s)
Toxinas Bacterianas , Vibrio parahaemolyticus , Vibrio parahaemolyticus/química , Microscopía por Crioelectrón , Proteínas Hemolisinas/química , Proteínas Hemolisinas/toxicidad , Toxinas Bacterianas/química
5.
J Biol Chem ; 298(10): 102441, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36055404

RESUMEN

Vibrio cholerae cytolysin (VCC) is a potent membrane-damaging ß-barrel pore-forming toxin. Upon binding to the target membranes, VCC monomers first assemble into oligomeric prepore intermediates and subsequently transform into transmembrane ß-barrel pores. VCC harbors a designated pore-forming motif, which, during oligomeric pore formation, inserts into the membrane and generates a transmembrane ß-barrel scaffold. It remains an enigma how the molecular architecture of the pore-forming motif regulates the VCC pore-formation mechanism. Here, we show that a specific pore-forming motif residue, E289, plays crucial regulatory roles in the pore-formation mechanism of VCC. We find that the mutation of E289A drastically compromises pore-forming activity, without affecting the structural integrity and membrane-binding potential of the toxin monomers. Although our single-particle cryo-EM analysis reveals WT-like oligomeric ß-barrel pore formation by E289A-VCC in the membrane, we demonstrate that the mutant shows severely delayed kinetics in terms of pore-forming ability that can be rescued with elevated temperature conditions. We find that the pore-formation efficacy of E289A-VCC appears to be more profoundly dependent on temperature than that of the WT toxin. Our results suggest that the E289A mutation traps membrane-bound toxin molecules in the prepore-like intermediate state that is hindered from converting into the functional ß-barrel pores by a large energy barrier, thus highlighting the importance of this residue for the pore-formation mechanism of VCC.


Asunto(s)
Proteínas Bacterianas , Citotoxinas , Proteínas Citotóxicas Formadoras de Poros , Vibrio cholerae , Factores de Virulencia , Membrana Celular/metabolismo , Citotoxinas/química , Citotoxinas/genética , Vibrio cholerae/patogenicidad , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Factores de Virulencia/química , Factores de Virulencia/genética , Proteínas Citotóxicas Formadoras de Poros/química , Proteínas Citotóxicas Formadoras de Poros/genética , Secuencias de Aminoácidos , Mutación , Ácido Glutámico/química , Ácido Glutámico/genética
6.
FASEB J ; 36(10): e22557, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36125006

RESUMEN

Vibrio cholerae cytolysin (VCC) is a ß-barrel pore-forming toxin (ß-PFT). It exhibits potent hemolytic activity against erythrocytes that appears to be a direct outcome of its pore-forming functionality. However, VCC-mediated cell-killing mechanism is more complicated in the case of nucleated mammalian cells. It induces apoptosis in the target nucleated cells, mechanistic details of which are still unclear. Furthermore, it has never been explored whether the ability of VCC to trigger programmed cell death is stringently dependent on its pore-forming activity. Here, we show that VCC can evoke hallmark features of the caspase-dependent apoptotic cell death even in the absence of the pore-forming ability. Our study demonstrates that VCC mutants with abortive pore-forming hemolytic activity can trigger apoptotic cell death responses and cytotoxicity, similar to those elicited by the wild-type toxin. VCC as well as its pore formation-deficient mutants display prominent propensity to translocate to the target cell mitochondria and cause mitochondrial membrane damage. Therefore, our results for the first time reveal that VCC, despite being an archetypical ß-PFT, can kill target nucleated cells independent of its pore-forming functionality. These findings are intriguing for a ß-PFT, whose destination is generally expected to remain limited on the target cell membranes, and whose mode of action is commonly attributed to the membrane-damaging pore-forming ability. Taken together, our study provides critical new insights regarding distinct implications of the two important virulence functionalities of VCC for the V. cholerae pathogenesis process: hemolytic activity for iron acquisition and cytotoxicity for tissue damage by the bacteria.


Asunto(s)
Toxinas Biológicas , Vibrio cholerae , Animales , Caspasas/metabolismo , Muerte Celular , Citotoxinas/metabolismo , Hierro/metabolismo , Mamíferos/metabolismo , Toxinas Biológicas/metabolismo , Vibrio cholerae/metabolismo
7.
Soft Matter ; 18(28): 5293-5301, 2022 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-35790122

RESUMEN

Pore-forming toxins (PFTs) produced by pathogenic bacteria serve as prominent virulence factors with potent cell-killing activity. Most of the ß-barrel PFTs form transmembrane oligomeric pores in the membrane lipid bilayer in the presence of cholesterol. The pore-formation mechanisms of the PFTs highlight well-orchestrated regulated events in the membrane environment, which involve dramatic changes in the protein structure and organization. Also, concerted crosstalk between protein and membrane lipid components appears to play crucial roles in the process. Membrane-damaging lesions formed by the pore assembly of the PFTs would also be expected to impose drastic alterations in the membrane organization, details of which remain obscure in most of the cases. Prior reports have established that aqueous interfaces of liquid crystals (LCs) offer promise as responsive interfaces for biomolecular events (at physiologically relevant concentrations), which can be visualized as optical signals. Inspired by this, herein, we sought to understand the lipid membrane interactions of a ß-barrel PFT i.e., Vibrio cholerae cytolysin (VCC), using LC-aqueous interfaces. Our results show the formation of dendritic patterns upon the addition of VCC to the lipid embedded with cholesterol over the LC film. In contrast, we did not observe any LC reorientation upon the addition of VCC to the lipid-laden LC-aqueous interface in the absence of cholesterol. An array of techniques such as polarizing optical microscopy (POM), atomic force microscopy (AFM), and fluorescence measurements were utilized to decipher the LC response to the lipid interactions of VCC occurring at these interfaces. Altogether, the results obtained from our study provide a novel platform to explore the mechanistic aspects of the protein-membrane interactions, in the process of membrane pore-formation by the membrane-damaging PFTs.


Asunto(s)
Cristales Líquidos , Vibrio cholerae , Membrana Celular/química , Colesterol , Citotoxinas/química , Citotoxinas/metabolismo , Citotoxinas/farmacología , Membrana Dobles de Lípidos/química , Vibrio cholerae/química , Vibrio cholerae/metabolismo , Agua/metabolismo
8.
Biochim Biophys Acta Biomembr ; 1864(11): 184013, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-35908609

RESUMEN

Pore-forming toxins (PFTs) rupture plasma membranes and kill target cells. PFTs are secreted as soluble monomers that undergo drastic structural rearrangements upon interacting with the target membrane and generate transmembrane oligomeric pores. A detailed understanding of the molecular mechanisms of the pore-formation process remains unclear due to limited structural insights regarding the transmembrane oligomeric pore states of the PFTs. However, recent advances in the field of cryo-electron microscopy (cryo-EM) have led to the high-resolution structure determination of the oligomeric pore forms of diverse PFTs. Here, we discuss the pore-forming mechanisms of various PFTs, specifically the mechanistic details contributed by the cryo-EM-based structural studies.


Asunto(s)
Toxinas Bacterianas , Toxinas Bacterianas/química , Membrana Celular/metabolismo , Microscopía por Crioelectrón , Proteínas Citotóxicas Formadoras de Poros/química
9.
J Membr Biol ; 255(2-3): 161-173, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35305136

RESUMEN

Pore-forming protein toxins (PFTs) represent a diverse class of membrane-damaging proteins that are produced by a wide variety of organisms. PFT-mediated membrane perforation is largely governed by the chemical composition and the physical properties of the plasma membranes. The interaction between the PFTs with the target membranes is critical for the initiation of the pore-formation process, and can lead to discrete membrane reorganization events that further aids in the process of pore-formation. Punching holes on the plasma membranes by the PFTs interferes with the cellular homeostasis by disrupting the ion-balance inside the cells that in turn can turn on multiple signalling cascades required to restore membrane integrity and cellular homeostasis. In this review, we discuss the physicochemical attributes of the plasma membranes associated with the pore-formation processes by the PFTs, and the subsequent membrane remodelling events that may start off the membrane-repair mechanisms.


Asunto(s)
Toxinas Biológicas , Membrana Celular/metabolismo , Membranas , Proteínas Citotóxicas Formadoras de Poros/química , Toxinas Biológicas/metabolismo
10.
Adv Protein Chem Struct Biol ; 128: 241-288, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35034720

RESUMEN

Pore-forming proteins (PFPs) of the diverse life forms have emerged as the potent cell-killing entities owing to their specialized membrane-damaging properties. PFPs have the unique ability to perforate the plasma membranes of their target cells, and they exert this functionality by creating oligomeric pores in the membrane lipid bilayer. Pathogenic bacteria employ PFPs as toxins to execute their virulence mechanisms, whereas in the higher vertebrates PFPs are deployed as the part of the immune system and to generate inflammatory responses. PFPs are the unique dimorphic proteins that are generally synthesized as water-soluble molecules, and transform into membrane-inserted oligomeric pore assemblies upon interacting with the target membranes. In spite of sharing very little sequence similarity, PFPs from diverse organisms display incredible structural similarity. Yet, at the same time, structure-function mechanisms of the PFPs document remarkable versatility. Such notions establish PFPs as the fascinating model system to explore variety of unsolved issues pertaining to the structure-function paradigm of the proteins that interact and act in the membrane environment. In this article, we discuss our current understanding regarding the structural basis of the pore-forming functions of the diverse class of PFPs. We attempt to highlight the similarities and differences in their structures, membrane pore-formation mechanisms, and their implications for the various biological processes, ranging from the bacterial virulence mechanisms to the inflammatory immune response generation in the higher animals.


Asunto(s)
Bacterias , Animales , Membrana Celular
11.
J Cell Biol ; 220(12)2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34617964

RESUMEN

Vibrio cholerae cytolysin (VCC) is a water-soluble, membrane-damaging, pore-forming toxin (PFT) secreted by pathogenic V. cholerae, which causes eukaryotic cell death by altering the plasma membrane permeability. VCC self-assembles on the cell surface and undergoes a dramatic conformational change from prepore to heptameric pore structure. Over the past few years, several high-resolution structures of detergent-solubilized PFTs have been characterized. However, high-resolution structural characterization of small ß-PFTs in a lipid environment is still rare. Therefore, we used single-particle cryo-EM to characterize the structure of the VCC oligomer in large unilamellar vesicles, which is the first atomic-resolution cryo-EM structure of VCC. From our study, we were able to provide the first documented visualization of the rim domain amino acid residues of VCC interacting with lipid membrane. Furthermore, cryo-EM characterization of lipid bilayer-embedded VCC suggests interesting conformational variabilities, especially in the transmembrane channel, which could have a potential impact on the pore architecture and assist us in understanding the pore formation mechanism.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/ultraestructura , Microscopía por Crioelectrón , Membrana Dobles de Lípidos/química , Perforina/química , Perforina/ultraestructura , Multimerización de Proteína , Vibrio cholerae/metabolismo , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Modelos Moleculares , Perforina/metabolismo , Estructura Secundaria de Proteína
12.
Biomed Mater ; 16(6)2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34517359

RESUMEN

The rationale behind the success of nickel free or with extremely low nickel austenitic high manganese and nitrogen stabilized stainless steels is adverse influences of nickel ion on human body. Replacement of nickel by nitrogen and manganese provides a stable microstructure and facilitates better biocompatibility in respect of the conventional 316L austenitic stainless steel (316L SS). In this investigation, biocompatibility of the high-manganese and nitrogen stabilized (Fe-18Cr-22Mn-0.65N) austenitic stainless steel was studied and found highly promising.In vitrocell culture and cell proliferation (MTT) assays were performed on this stainless steel and assessed in respect of the 316L SS. Both the steels exhibited similar cell growth behavior. Furthermore, an enhancement was observed in cell proliferation on the Fe-18Cr-22Mn-0.65N SS after surface modification by ultrasonic shot peening (USP). The mean percent proliferation of the MG-63 cells increased from ≈88% for Un-USP to 98% and 105% for USP 3-2 and USP 2-2 samples, respectively for 5 d of incubation. Interestingly,in vivoanimal study performed in rabbits for 3 and 6 weeks showed callus formation and sign of union without any allergic reaction.


Asunto(s)
Materiales Biocompatibles , Aleaciones Dentales , Prótesis e Implantes , Acero Inoxidable , Materiales Biocompatibles/química , Materiales Biocompatibles/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Aleaciones Dentales/química , Aleaciones Dentales/toxicidad , Humanos , Manganeso/química , Ensayo de Materiales , Nitrógeno/química , Acero Inoxidable/química , Acero Inoxidable/toxicidad
13.
Front Mol Biosci ; 8: 717147, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34368235

RESUMEN

Thermostable direct hemolysin (TDH) is the major virulence determinant of the gastroenteric bacterial pathogen Vibrio parahaemolyticus. TDH is a membrane-damaging pore-forming toxin (PFT). TDH shares remarkable structural similarity with the actinoporin family of eukaryotic PFTs produced by the sea anemones. Unlike most of the PFTs, it exists as tetramer in solution, and such assembly state is crucial for its functionality. Although the structure of the tetrameric assembly of TDH in solution is known, membrane pore structure is not available yet. Also, the specific membrane-interaction mechanisms of TDH, and the exact role of any receptor(s) in such process, still remain unclear. In this mini review, we discuss some of the unique structural and physicochemical properties of TDH, and their implications for the membrane-damaging action of the toxin. We also present our current understanding regarding the membrane pore-formation mechanism of this atypical bacterial PFT.

14.
Biochem Soc Trans ; 49(1): 455-465, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33492383

RESUMEN

The integrity of the plasma membranes is extremely crucial for the survival and proper functioning of the cells. Organisms from all kingdoms of life employ specialized pore-forming proteins and toxins (PFPs and PFTs) that perforate cell membranes, and cause detrimental effects. PFPs/PFTs exert their damaging actions by forming oligomeric pores in the membrane lipid bilayer. PFPs/PFTs play important roles in diverse biological processes. Many pathogenic bacteria secrete PFTs for executing their virulence mechanisms. The immune system of the higher vertebrates employs PFPs to kill pathogen-infected cells and transformed cancer cells. The most obvious consequence of membrane pore-formation by the PFPs/PFTs is the killing of the target cells due to the disruption of the permeability barrier function of the plasma membranes. PFPs/PFTs can also activate diverse cellular processes that include activation of the stress-response pathways, induction of programmed cell death, and inflammation. Upon attack by the PFTs, host cells may also activate pathways to repair the injured membranes, restore cellular homeostasis, and trigger inflammatory immune responses. In this article, we present an overview of the diverse cellular responses that are triggered by the PFPs/PFTs, and their implications in the process of pathogen infection and immunity.


Asunto(s)
Inmunidad , Infecciones/patología , Proteínas Citotóxicas Formadoras de Poros/farmacología , Toxinas Biológicas/farmacología , Animales , Membrana Celular/efectos de los fármacos , Humanos , Inmunidad/efectos de los fármacos , Inmunidad/fisiología , Infecciones/inmunología , Membrana Dobles de Lípidos/metabolismo , Virulencia/fisiología
15.
Front Microbiol ; 12: 809782, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35140698

RESUMEN

Vibrio cholerae cytolysin (VCC) is a ß-barrel pore-forming toxin (ß-PFT). Upon encountering the target cells, VCC forms heptameric ß-barrel pores and permeabilizes the cell membranes. Structure-function mechanisms of VCC have been extensively studied in the past. However, the existence of any natural inhibitor for VCC has not been reported yet. In the present study, we show that curcumin can compromise the membrane-damaging activity of VCC. Curcumin is known to modulate a wide variety of biological processes and functions. However, the application of curcumin in the physiological scenario often gets limited due to its extremely poor solubility in the aqueous environment. Interestingly, we find that VCC can associate with the insoluble fraction of curcumin in the aqueous medium and thus gets separated from the solution phase. This, in turn, reduces the availability of VCC to attack the target membranes and thus blocks the membrane-damaging action of the toxin. We also observe that the soluble aqueous extract of curcumin, generated by the heat treatment, compromises the pore-forming activity of VCC. Interestingly, in the presence of such soluble extract of curcumin, VCC binds to the target membranes and forms the oligomeric assembly. However, such oligomers appear to be non-functional, devoid of the pore-forming activity. The ability of curcumin to bind to VCC and neutralize its membrane-damaging activity suggests that curcumin has the potential to act as an inhibitor of this potent bacterial ß-PFT.

16.
Mol Microbiol ; 115(4): 508-525, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33089544

RESUMEN

ß-barrel pore-forming toxins perforate cell membranes by forming oligomeric ß-barrel pores. The most crucial step is the membrane-insertion of the pore-forming motifs that create the transmembrane ß-barrel scaffold. Molecular mechanism that regulates structural reorganization of these pore-forming motifs during ß-barrel pore-formation still remains elusive. Using Vibrio cholerae cytolysin as an archetypical example of the ß-barrel pore-forming toxin, we show that a key tyrosine residue (Y321) in the hinge region of the pore-forming motif plays crucial role in this process. Mutation of Y321 abrogates oligomerization of the membrane-bound toxin protomers, and blocks subsequent steps of pore-formation. Our study suggests that the presence of Y321 in the hinge region of the pore-forming motif is crucial for the toxin molecule to sense membrane-binding, and to trigger essential structural rearrangements required for the subsequent oligomerization and pore-formation process. Such a regulatory mechanism of pore-formation by V. cholerae cytolysin has not been documented earlier in the structurally related ß-barrel pore-forming toxins.


Asunto(s)
Secuencias de Aminoácidos , Perforina/química , Perforina/fisiología , Tirosina/química , Vibrio cholerae/química , Vibrio cholerae/fisiología , Proteínas Bacterianas/química , Proteínas Bacterianas/fisiología , Línea Celular , Membrana Celular/metabolismo , Células Cultivadas , Citotoxinas/química , Citotoxinas/fisiología , Humanos , Microscopía Electrónica de Transmisión , Simulación de Dinámica Molecular , Mutación , Perforina/ultraestructura , Conformación Proteica , Multimerización de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Vibrio cholerae/ultraestructura
17.
J Membr Biol ; 253(5): 469-478, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32955633

RESUMEN

Pore-forming proteins/toxins (PFPs/PFTs) are the distinct class of membrane-damaging proteins. They act by forming oligomeric pores in the plasma membranes. PFTs and PFPs from diverse organisms share a common mechanism of action, in which the designated pore-forming motifs of the membrane-bound protein molecules insert into the membrane lipid bilayer to create the water-filled pores. One common characteristic of these pore-forming motifs is that they are amphipathic in nature. In general, the hydrophobic sidechains of the pore-forming motifs face toward the hydrophobic core of the membranes, while the hydrophilic residues create the lining of the water-filled pore lumen. Interestingly, pore-forming motifs of the distinct subclass of PFPs/PFTs share very little sequence similarity with each other. Therefore, the common guiding principle that governs the sequence-to-structure paradigm in the mechanism of action of these PFPs/PFTs still remains an enigma. In this article, we discuss this notion using the examples of diverse groups of membrane-damaging PFPs/PFTs.


Asunto(s)
Secuencia de Aminoácidos , Variación Genética , Proteínas Citotóxicas Formadoras de Poros/química , Proteínas Citotóxicas Formadoras de Poros/genética , Toxinas Biológicas/química , Toxinas Biológicas/genética , Animales , Membrana Celular/química , Membrana Celular/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Relación Estructura-Actividad , Toxinas Biológicas/metabolismo
18.
Ultrasonics ; 104: 106110, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32146383

RESUMEN

Surface mechanical attrition treatment (SMAT) of metallic biomaterials has gained significant importance due to its ability to develop nano structure in the surface region. In the present study, the microstructural changes and corrosion behavior of the commercially pure titanium (cp-Ti), following different durations of ultrasonic shot peening (USSP) has been investigated. cp-Ti was shot peened for different durations from 0 to 120 s and the treated samples were examined for microstructural changes in the surface region, cell viability and corrosion behavior. Cell viability was considerably increased after USSP for 60-120 s, exhibiting maximum for the 90 s of USSP. The passivation tendency was also improved with peening duration up to 90 s, however, it declined for longer duration of USSP. The beneficial effects of USSP may be attributed to nano structuring in the surface region and development of higher positive potentials at the USSP treated surface. Transmission Electron Microscope (TEM) examination of the USSPed surface revealed dislocation entanglement and substructure. Also, higher surface volta potential was observed over the USSPed sample exhibiting better cell proliferation. The present work is corollary to previous work of the group and mainly discusses the role of USSP duration, as a process parameter, on the cell viability and corrosion resistance of cp-Ti.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Nanoestructuras/química , Titanio/química , Ultrasonido/métodos , Proliferación Celular , Supervivencia Celular , Corrosión , Técnicas Electroquímicas , Humanos , Ensayo de Materiales , Microscopía de Fuerza Atómica , Microscopía Electrónica , Propiedades de Superficie , Difracción de Rayos X
19.
Biochemistry ; 59(4): 605-614, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31808340

RESUMEN

Thermostable direct hemolysin (TDH) of Vibrio parahemolyticus is a membrane-damaging pore-forming toxin with potent cytolytic/cytotoxic activity. TDH exists as a tetramer consisting of protomers with a core ß-sandwich domain, flanked by an 11-amino acid long N-terminal region (NTR). This NTR could not be modeled in the previously determined crystal structure of TDH. Moreover, the functional implication of NTR for the membrane-damaging action of TDH remains unknown. In the present study, we have explored the implications of NTR for the structure-function mechanism of TDH. Our data show that the presence of NTR modulates the physicochemical property of TDH in terms of augmenting the amyloidogenic propensity of the protein. Deletion of NTR compromises the binding of TDH toward target cell membranes and drastically affects the membrane-damaging cytolytic/cytotoxic activity of the toxin. Mutations of aromatic/hydrophobic residues within NTR also confer compromised cell-killing activity. Moreover, covalent trapping of NTR, via an engineered disulfide bond, against the core ß-sandwich domain also abrogates the cytolytic/cytotoxic activity of TDH. This observation suggests that an unrestrained configuration of NTR is crucial for the membrane-damaging action of TDH. On the basis of our study, we propose a model explaining the role of NTR in the membrane-damaging function of TDH.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/ultraestructura , Proteínas Bacterianas/química , Toxinas Bacterianas/metabolismo , Fenómenos Bioquímicos/genética , Transporte Biológico/genética , Proteínas Hemolisinas/química , Proteínas Hemolisinas/fisiología , Hemólisis , Humanos , Mutación/genética , Subunidades de Proteína/metabolismo , Vibrio parahaemolyticus/química , Vibrio parahaemolyticus/genética , Vibrio parahaemolyticus/metabolismo
20.
Biochemistry ; 59(2): 163-170, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31608629

RESUMEN

A wide variety of bacterial pathogens secrete a unique class of proteins that attack target cell membranes and form transmembrane oligomeric pores with distinct ß-barrel structural scaffolds. Owing to their specific mode of action and characteristic structural assembly, these proteins are termed as ß-barrel pore-forming toxins (ß-PFTs). The most obvious consequence of such pore-forming activity of bacterial ß-PFTs is the permeabilization of cell membranes, which eventually leads to cell death. Bacterial ß-PFTs have been studied extensively for nearly past four decades, and their mechanisms of actions have revealed some of the most enigmatic aspects of the protein structure-function paradigm. In most of the cases, ß-PFTs are released by the bacteria as water-soluble monomeric precursors, which upon encountering target cell membranes assemble into membrane-inserted oligomeric pores. Structural descriptions are now documented for the water-soluble precursor forms, as well as for the membrane-anchored oligomeric pores of many ß-PFTs. These studies have revealed that ß-PFTs undergo a series of well-orchestrated structural rearrangements during membrane pore formation. Nevertheless, mechanisms that trigger and regulate distinct steps of the pore-formation processes still remain obscure. Here, we discuss our current understanding regarding structure-function mechanisms in the ß-PFT family, with particular emphasis on some of the unsolved issues associated with the ß-barrel pore-formation mechanism.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Membrana Celular/efectos de los fármacos , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Bacterias/química , Proteínas Bacterianas/química , Toxinas Bacterianas/química , Proteínas Citotóxicas Formadoras de Poros/química , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...