Acessibilidade / Reportar erro

Escherichia coli vacuolating factor, involved in avian cellulitis, induces actin contraction and binds to cytoskeleton proteins in fibroblasts

Abstract

Background

Avian pathogenic Escherichia coli (APEC) isolated from avian cellulitis lesions produces a toxin, named Escherichia coli vacuolating factor (ECVF), that causes cell vacuolization and induces inflammatory response in broiler chicken.

Methods

We investigated the intracellular activities of ECVF in avian fibroblasts using fluorescence staining, electron microscopy, MTT and LDH measurements. As ECVF act specifically in avian cells, we performed blotting assay followed by mass spectrometry to better understand its initial intracellular protein recognition.

Results

ECVF induced actin contraction, mitochondrial damage and membrane permeability alterations. Ultrastructural analysis showed intracellular alterations, as nuclear lobulation and the presence of degraded structures inside the vacuoles. Moreover, ECVF induced cell death in fibroblasts. ECVF-biotin associates to at least two proteins only in avian cell lysates: alpha-actinin 4 and vinculin, both involved in cytoskeleton structure.

Conclusion

These findings demonstrated that ECVF plays an important role in avian cellulitis, markedly in initial steps of infection. Taken together, the results place this toxin as a target for drug and/or vaccine development, instead of the use of large amounts antibiotics.

Keywords
Escherichia coli ; Escherichia coli vacuolating factor (ECVF); Avian pathogenic Escherichia coli (APEC); Cytoskeleton

Background

Avian pathogenic Escherichia coli is a group of E. coli that cause extra-intestinal colibacillosis in poultry that is responsible for important worldwide economic losses [11. Dho-Moulin M, Fairbrother JM. Avian pathogenic Escherichia coli (APEC). Vet Res. 1999 Mar-Jun;30(2-3):299-316. -33. Guabiraba R, Schouler C. Avian colibacillosis: still many black holes. FEMS Microbiol Lett. 2015 Aug;362(15):fnv118. ]. APEC is also relevant to humans because it shares some similarities with human extra-intestinal pathogenic E. coli (ExPEC), which claims attention to a possible zoonotic risk [44. Moulin-Schouleur M, Reperant M, Laurent S, Bree A, Mignon-Grasteau S, Germon P, et al. Extraintestinal pathogenic Escherichia coli strains of avian and human origin: link between phylogenetic relationships and common virulence patterns. J Clin Microbiol. 2007 Oct;45(10):3366-76. -77. Mellata M. Human and avian extraintestinal pathogenic Escherichia coli: infections, zoonotic risks, and antibiotic resistance trends. Foodborne Pathog Dis. 2013 Nov;10(11):916-32. ]. The similarities in their genomic sequences, serogroups, antibiotic resistance and other abilities to cause disease suggest that APECs are a reservoir of virulence factors (VF) to ExPEC [66. Bauchart P, Germon P, Brée A, Oswald E, Hacker J, Dobrindt U. Pathogenomic comparison of human extraintestinal and avian pathogenic Escherichia coli - Search for factors involved in host specificity or zoonotic potential. Microb Pathog. 2010 Sep;49(3):105-15. ,88. Rodriguez-Siek KE, Giddings CW, Doetkott C, Johnson TJ, Fakhr MK, Nolan LK. Comparison of Escherichia coli isolates implicated in human urinary tract infection and avian colibacillosis. Microbiology. 2005 Jun;151(Pt 6):2097-110. ,99. Zhao L, Gao S, Huan H, Xu X, Zhu X, Yang W, et al. Comparison of virulence factors and expression of specific genes between uropathogenic Escherichia coli and avian pathogenic E. coli in a murine urinary tract infection model and a chicken challenge model. Microbiology. 2009 May;155(Pt 5):1634-44. ]. Adhesins, capsules, iron acquisition systems and secreted toxins [1010. Janben T, Schwarz C, Preikschat P, Voss M, Philipp HC, Wieler LH. Virulence-associated genes in avian pathogenic Escherichia coli (APEC) isolated from internal organs of poultry having died from colibacillosis. Int J Med Microbiol. 2001 Nov;291(5):371-8. ] are VF implicated in avian diseases.

Previously, a heat-labile cytotoxin produced by an APEC isolated from avian cellulitis lesions was described inducing vacuolization only in avian cells [1111. Salvadori MR, Yano T, Carvalho HF, Parreira VR, Gyles CL. Vacuolating cytotoxin produced by avian pathogenic Escherichia coli. Avian Dis. 2001 Jan-Mar;45(1):43-51. ], this cytotoxin was called ECVF. Parreira and Gyles [1212. Parreira VR, Gyles CL. A novel pathogenicity island integrated adjacent to the thrW tRNA gene of avian pathogenic Escherichia coli encodes a vacuolating autotransporter toxin. Infect Immun. 2003 Sep;71(9):5087-96. ] also described a pathogenicity island (PAI) adjacent to the thrW tRNA, which encode an autotransporter toxin, that was named vacuolating autotransporter toxin (Vat). Furthermore, cellular vacuolization induced by Vat and ECVF are similar to VacA, a cytotoxin produced by Helicobacter pylori [1313. Cover TL, Blaser MJ. Purification and characterization of the vacuolating toxin from Helicobacter pylori. J Biol Chem. 1992 May;267(15):10570-5. ]. Although VacA is specific for mammalian cells [1414. Cover TL, Blanke SR. Helicobacter pylori VacA, a paradigm for toxin multifunctionality. Nat Rev Microbiol. 2005 Apr;3(4):320-32. ], ECVF is toxic only to avian cells [1111. Salvadori MR, Yano T, Carvalho HF, Parreira VR, Gyles CL. Vacuolating cytotoxin produced by avian pathogenic Escherichia coli. Avian Dis. 2001 Jan-Mar;45(1):43-51. ].

Later, Quel et al. [1515. Quel NG, Aragão AZB, Salvadori MR, Farias AS, Joazeiro PP, Santos LMB, et al. Cellulitis lesions in broiler chickens are induced by Escherichia coli Vacuolating Factor (ECVF). Vet Microbiol. 2013 Mar 23;162(2-4):866-72. ] observed that purified ECVF induces acute inflammatory response in the epidermis, dermis and panniculus of broiler chickens, similar to cellulitis inflammation induced by E. coli AC53 (isolated from cellulitis lesions). However, the molecular action of the toxin in the avian cells has not yet been clarified. In this study, we proposed to better understand the multiple biological damage that ECVF induces in avian cells, leading them to death. We believe that ECVF could be a potential target to drug development against cellulitis development in poultry.

Methods

ECVF production and purification

ECVF was produced by E. coli AC 53 (serogroup O21,83) kindly provided by Dr. Carlton Gyles (University of Guelph, Ontario, Canada), isolated from cellulitis lesions [1616. Peighambari SM, Julian RJ, Vaillancourt JP, Gyles CL. Escherichia coli cellulitis: experimental infections in broiler chickens. Avian Dis. 1995 Jan-Mar;39(1):125-34. ]. The toxin was produced and purified as described previously [1515. Quel NG, Aragão AZB, Salvadori MR, Farias AS, Joazeiro PP, Santos LMB, et al. Cellulitis lesions in broiler chickens are induced by Escherichia coli Vacuolating Factor (ECVF). Vet Microbiol. 2013 Mar 23;162(2-4):866-72. ]. After purification, ECVF toxin concentration was measured using Bio-Rad Protein Assay and the cytotoxic dose for 50% of viability (CD50) was determined at 40 µg mL-1 (data not shown).

Cell culture

In this study we used two avian fibroblasts: a primary chicken embryo fibroblasts (CEF), kindly prepared and donated by Fort Dodge Animal Health (Campinas, São Paulo, Brazil) and CEC-32, that was kindly provided by Dr. Fabiana Horn (Biosciences Institute of Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil). Both cells were maintained at Eagle´s minimal essential medium (EMEM - Nutricell) supplemented with 10% fetal bovine serum (FBS - Sigma) and 1% of an antibiotic solution (100,000 U/L penicillin and 10 mg/L streptomycin - Sigma). To all experiments, the controls were treated with 60mM Tris-HCl pH7.4, which was the purification buffer, in the same volume that was applied in the ECVF-treated cells. Additionally, HUVEC and Vero cells were cultured at the same conditions as described above, until the use in ECVF-biotin assay.

Fluorescence staining

Fluorescence actin staining (FAS)

FAS was performed as previously described [1717. Navarro-García F, Sears C, Eslava C, Cravioto A, Nataro JP. Cytoskeletal effects induced by pet, the serine protease enterotoxin of enteroaggregative Escherichia coli. Infect Immun. 1999 May;67(5):2184-92. ]. Fibroblasts were incubated with purified ECVF (2.5 × CD50) and observed during 24 h. After incubation, avian cells were fixed with 2% formalin, washed, permeabilized by adding 0.1% Triton X-100, and stained with 0.05 mg/mL of phalloidin-TRITC (Sigma) in PBS. CEC-32 nuclei were additionally stained with 4’,6-diamidino-2-phenylindole (DAPI). Slides were mounted with 90% glycerol, covered with a glass cover slide, and examined at a fluorescence microscope (LEICA DM 2500).

Acridine orange (AO)

Dye accumulation was assessed by exposing living CEF cells, after vacuolization induced by exposure to ECVF (CD50) for 6, 18 and 24 h, to 5 μg/mL of AO (Sigma) for 2 min in EMEM as described by Catrenich and Chestnut [1818. Catrenich CE, Chestnut MH. Character and origin of vacuoles induced in mammalian cells by the cytotoxin of Helicobacter pylori. J Med Microbiol. 1992 Dec;37(6):389-95. ]. Cover-slips bearing cells were then rinsed three times in PBS, mounted on microscope slides and observed immediately (LEICA DM 2500). All observations were recorded until 10-15 min after AO exposure, to retain the cell morphology.

Transmission electron microscopy (TEM)

To exploit the endomembrane system under effects of ECVF, vacuolization was induced in CEC-32, which were observed using TEM. The avian fibroblasts (2 x 106 cells/mL) were treated with purified toxin (CD50), at different times: 2, 4, 6, 18 and 24 hours. Cells were fixed for 1h at RT with 2% glutaraldehyde in 0.1 M sodium cacodylate buffer (pH 7.4), post-fixed with 1% osmium tetroxide for 1h at 4°C and, subsequently with 1% uranyl acetate for 15 min at 4°C. The avian cells were then dehydrated in ascending concentrations of ethanol and embedded in Epon resin (Polysciences Inc.). Ultrathin sections were stained with 0.5% lead citrate and examined in a transmission electron microscope (LEO-906) at 60kV.

Cytotoxicity and cell death monitoring

MTT assay

The mitochondrial viability assay [1919. Borenfreund E, Babich H, Martin-Alguacil N. Comparisons of two in vitro cytotoxicity assays-The neutral red (NR) and tetrazolium MTT tests. Toxicol In Vitro. 1988;2(1):1-6. ], detects the cellular reduction of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) to formazan, by the mitochondrial dehydrogenase. Briefly, CEF cells were plated in 96-well plate (2 x 104 cells/mL) for 24 h and treated with ECVF (CD50) for 2, 4, 6, 15, 24, 36 and 48h. Cells were washed and fresh medium containing MTT solution (0.8 mg/mL) was added to each well. After 3 h at 37°C, in the dark, the medium was removed and the formazan was solubilized in HCl 1N - isopropanol (1:24 v/v). The plates were shaken for 10 min and the absorbance measured at 550 nm (reference to 700 nm). Two independent experiments were performed in triplicates.

Lactate dehydrogenase (LDH) release assay

Changes in cell membrane integrity of CEF exposed to ECVF (CD50) was determined as previously described [2020. Mitchell DB, Santone KS, Acosta D. Evaluation of cytotoxicity in cultured cells by enzyme leakage. J Tissue Cult Methods. 1980 Sep;6(3-4):113-6. ]. Briefly, CEF cells (2 x 104 cells/mL) were treated with ECVF at different time intervals (2, 4, 6 and 12h), the supernatant was collected and centrifuged at 400 × g per 5 min at 4°C (UniCenMR Herolab). An aliquot of 0.1 mL of the supernatant was placed in a cuvette containing 2.5 mL of phosphate buffer at 37°C combined with 0.2 mL of NADH (2.5 mg/mL) and 0.2 mL of sodium pyruvate (1 mg/mL). Kinect was measured at intervals of 30s until reach 3 min (end point) at 340 nm. CEF cells exposed to 10% SDS was used as a positive control for total LDH release.

DNA fragmentation detection by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)

To better understand the cell death process induced by ECVF, CEC-32 cells (2 × 106 cells/mL) were treated with the toxin (CD50), at different times: 2, 4, 6, 18 and 24 hours. Cells were fixed with 1.5% paraformaldehyde in PBS for 1h at 4°C. After that, fibroblasts were rinsed with PBS and subsequently permeabilized (0.1% sodium citrate and 0.1% Triton X-100) for 2 minutes at 4°C. Then, cells were incubated with the commercially available In Situ Cell Death Detection Kit (Roche Molecular Biochemicals), following the manufacturer´s instructions. After counter-stained with DAPI, slides were mounted with 80% glycerol and observed immediately at fluorescence microscope (LEICA DM 2500). Total and death cells were counted in 5 different fields per condition and the results were subjected to a statistical analysis using GraphPad Prism, version 5.

Host proteins recognition by ECVF

ECVF biotinylation

Lyophilized ECVF was resuspended in PBS containing 10 mg mL-1 of biotin (EZ-link sulfo-NHS-LC-biotin - Thermo Fisher Scientific) and incubated for 1 hour at 37°C. Then, the excess of biotin was removed in a desalting column (ZebaTM Spin Desalting Column - Thermo Fisher Scientific) and biotinylated ECVF was used to detect proteins in a blotting assay according to the methodology below. The entire ECVF biotinylation process was performed according to the manufacturer's recommendations.

Interaction with ECVF-biotin and host cell proteins

Cell monolayers (HUVEC, CEF and Vero) were washed twice with sterile PBS, pH 7.4, and resuspended by scraping in the same buffer. Cell disruption was made using ultrasound (amplitude of 20% for 2 min.). Then, the insoluble pellet was removed by centrifugation (10000 × g for 15 min at 4 °C) and the supernatant was used as described below. Protein dosage was performed as described by Bradford [2121. Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976 May 7;72(1-2):248-54. ]. SDS-PAGE was performed according to the methodology proposed by Laemmli [2222. Laemmli UK. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature. 1970 Aug 15;227(5259):680-5. ]. Protein extracts (50 µg for each cell line) were combined with the sample buffer (v/v) and boiled for 5 min. Proteins separation was carried out with 2 gels, one was stained by Coomassie bright blue (R 250) and the other was used for western blotting (performed as described by Renesto et al. [2323. Renesto P, Samson L, Ogata H, Azza S, Fourquet P, Gorvel JP, et al. Identification of two putative rickettsial adhesins by proteomic analysis. Res Microbiol. 2006 Sep;157(7):605-12. ], with modifications). Proteins separated in the SDS-PAGE were transferred to nitrocellulose membrane (Bio-rad), subsequently blocked in the TBS-T solution (Tris buffer, supplemented with 0.1% Tween 20 and 5% BSA) for 1 h, at 4 °C. Then, the membrane was incubated in the TBS-T solution containing ECVF-biotin, in the proportion of 1: 200 (v/v), overnight, at 4 °C. Membranes were washed three times with TBS-T and incubated with peroxidase-conjugated streptavidin (1:1000, GE Healthcare). The detection of ECVF binding to cellular proteins was revealed by chemiluminescence.

Protein identification by mass spectrometry (MS)

In order to identify the proteins that compose the interaction with the ECVF, the band was analyzed by mass spectrometry, performed by CEFAP (Center for Research Facilities, ICB-USP) in a LTQ Orbitrap Velos (Thermo Fisher). Raw data was processed in Mascot against Gallus gallus database.

Results

ECVF induces contraction of the actin filaments

CEC-32 cells were treated with high amounts of ECVF toxin (2.5 × CD50). After only 2 hours of incubation (Figure 1B), clearly changes in cell morphology were observed, such as cytoplasmic alterations followed by nuclear and chromatin modifications. Actin was under retraction and it was possible to observe a consequent loss of adhesion to the cell support and neighboring cells, leading to monolayer disruption (Figure 1). These effects were more evident after 18h (Figure 1E) and 24h (data not shown) of exposure to ECVF, when progressive loss of nuclei per field was observed. CEF cells, treated with a lower concentration of ECVF, also showed changes in the cytoskeleton compared to control cells. After incubation for 6 hours, some of these primary fibroblasts were rounded, due to the contraction of the actin filaments (Additional file 1 Additional file 1. CEF cells showing actin stained in green (Phalloidin-FITC). After 6h of ECVF treatment, an increased number of round shaped cells were observed. ).

Figure 1.
Alterations in cell morphology due to the contraction of the actin filaments. (A) CEC-32 cells without ECVF, 24 hours. (B, C, D and E) CEC-32 after ECVF incubation for 2, 4, 6, and 18 hours, respectively. Red: Phalloidin-TRITC; blue: DAPI. Arrows indicate normal actin filaments and arrowheads indicate contracted actin filaments. Scale bars = 50 μm.

AO uptake indicates cytoplasmic acidification caused by ECVF

CEF cells were treated with ECVF toxin during 6, 18 and 24h and then were stained with AO, a fluorescent probe used to follow the pH changes in vacuoles or acid vesicles. Control cells showed only the nucleus stained in green (Figure 2A), as expected. After the first 6h in contact with ECVF toxin, bright orange spots appeared in the cytosol (Figure 2B), probably due to AO protonation and accumulation into the acid lysosomes. ECVF-induced damage is time-dependent, as after 18h it was possible to observe a loss in the acid pH compartmentalization, since the entire cell appears diffusely yellow-orange fluorescent (Figure 2C and D). After 24h, the vacuoles induced by ECVF remain unstained, suggesting that these compartments are non-acid (Figure 2D).

Figure 2.
AO pattern changes inside CEF cells after ECVF treatment. (A) Control without ECVF, 24 hours. (B, C and D) CEF after ECVF incubation for 6, 18 and 24 hours, respectively. n: nucleus; c: cytoplasm. Arrows indicate lysosomes in orange and arrowheads indicate unstained vacuoles. Scale bars = 100 μm (A); 50 μm (B, C and D).

ECVF induces nuclear and cytoplasmic alterations

The control cell has an intact nucleus, well-organized nucleoli and lightly packed chromatin (Figure 3A and B). On treated cells, ECVF induced intense cytoplasmic vacuolization, along with disorganization of the endomembrane system, mitochondria as well as cytoskeleton, when compared with untreated cells. After 6 hours of treatment, it was possible to see some chromatin condensation within the nucleus, nuclear shrinkage and apoptotic bodies; moreover, these effects became more pronounced with time (Figure 3C). In addition, after 18 hours of exposure to ECVF, it was possible to observe membranous components inside the vacuoles (Figure 3D).

Figure 3.
Intracellular alterations in CEC-32 fibroblasts induced by ECVF treatment. (A, B) Control, untreated fibroblasts, after 24 hours. Electron micrographs of untreated cells show endomembrane system distributed in a less compact cytoplasm and a round-shaped nucleus with orderly euchromatin, typical of nucleus at the interphase. (C, D) Electron micrographs of treated cells show endomembrane system with numerous vacuoles of irregularly shapes, filled with membranous components. Vacuoles also show signs of swelling and are distributed in compacted cytoplasm. Nuclei exhibit irregular shape and display an abundance of lobulations and also a high content of heterochromatin associated to nuclear envelope. n: nucleus; nu: nucleolus; c: cytoplasm; v: vacuoles. *Asterisks indicate chromatin condensation, arrowheads indicate nuclear lobulation and arrows indicate intravacuolar structures. Scale bars = 5 μm (A), 2 μm (B, D), 1 μm (C).

Mitochondrial activity and plasma membrane integrity are affected

CEF cells were evaluated for mitochondrial viability and membrane integrity. Results of mitochondrial viability are shown in Figure 4A. Cell viability was affected since the first 2 hours after ECVF inoculation, and the mitochondrial activity was reduced to less than 60% after 24 hours. Membrane integrity of CEF cells was assessed by LDH release assay. Membrane injury started at 2 hours after ECVF treatment. After 6 hours, the maximum release of the LDH enzyme was observed (Figure 4B).

Figure 4.
ECVF affects the mitochondrial activity and causes injury in membrane integrity in CEF cells. (A) MTT assay shows that CEF viability decreases to 60% after 2 hours of incubation with ECVF. Performed in triplicates. (B) Membrane integrity was lost and LDH had a maximum release after 6 hours of ECVF incubation (C+ represents the total LDH release after SDS treatment).

Fibroblast death was detected by TUNEL

The TUNEL assay, performed in CEC-32 cells, showed classic condensation and fragmentation of nuclei (Figure 5). The quantity of TUNEL-positive cells is represented in Figure 5A and 5B. The significant number of labeled cells in this assay was difficult to assess since we noticed a marked decrease in the number of cells, which became detached from the cover slips, after 18-24 hours of incubation with the toxin.

Figure 5.
ECVF induces death in avian fibroblasts. (A) CEC-32 control, 24 hours. (B, C, D, E and F) CEC-32 after ECVF exposure, assayed using TUNEL. 2, 4, 6, 18 and 24 hours, respectively. Representative fields of each condition. (G) Total and death cells were counted for each time of ECVF treatment in 5 different fields. Bars represent the sum of these 5 fields. (H) Percentage of the ratio between death/total cells (a, b and c represent statistical different groups). ANOVA one-way, followed by Tukey’s test.

ECVF recognizes only avian proteins

Western blotting demonstrated that the ECVF-biotin shows specificity to recognize proteins of avian cells (Figure 6), since there was only interaction with CEF's proteins, and no signal was detected for the proteins extracted from mammalian cell lines (HUVEC and Vero).

Figure 6.
Detection of avian proteins, possibly involved in the effects of ECVF on cells. (A) SDS-PAGE showing the proteins extracted from HUVEC, CEF and Vero cells. (B) ECVF-biotin incubated membrane was revealed using peroxidase-conjugated streptavidin. Arrow indicates the CEF’s proteins band for which interaction with ECVF was detected.

Discussion

Many poultry diseases are caused by E. coli, including the yolk sac infection syndrome, head-swelling, septicemia and cellulitis. Avian cellulitis is asymptomatic in the living bird and it is found only after slaughter, causing important economic losses. E. coli have been commonly isolated from avian cellulitis lesions [2424. Messier S, Quessy S, Robinson Y, Devriese LA, Hommez J, Fairbrother JM. Focal dermatitis and cellulitis in broiler chickens: bacteriological and pathological findings. Avian Dis. 1993 Jul-Sep;37(3):839-44. -2626. Barbieri NL, Oliveira AL de, Tejkowski TM, Pavanelo DB, Rocha DA, Matter LB, et al. Genotypes and pathogenicity of cellulitis isolates reveal traits that modulate APEC virulence. PLoS One. 2013 Aug 19;8(8):e72322. ] and its relevance in this disease has been suggested by several authors [1515. Quel NG, Aragão AZB, Salvadori MR, Farias AS, Joazeiro PP, Santos LMB, et al. Cellulitis lesions in broiler chickens are induced by Escherichia coli Vacuolating Factor (ECVF). Vet Microbiol. 2013 Mar 23;162(2-4):866-72. ,2727. Gomis SM, Watts T, Riddell C, Potter AA, Allan BJ. Experimental reproduction of Escherichia coli cellulitis and septicemia in broiler chickens. Avian Dis. 1997 Jan-Mar;41(1):234-40. -2929. de Brito BG, Gaziri LCJ, Vidotto MC. Virulence factors and clonal relationships among Escherichia coli strains isolated from broiler chickens with cellulitis. Infect Immun. 2003 Jul;71(7):4175-7. ]. Moreover, the presence of aerobactin, fimbriae and cytotoxins may increase the ability of E. coli to colonize the subcutaneous tissues of chicken [3030. Ewers C, Jansen T, Kiessling S, Philipp HC, Wieler LH. Molecular epidemiology of avian pathogenic Escherichia coli (APEC) isolated from colisepticemia in poultry. Vet Microbiol. 2004 Nov 30;104(1-2):91-101. ].

Other bacteria are also capable to produce vacuolating cytotoxins. H. pylori, Vibrio cholera and Aeromonas veronii biovar sobria, produce toxins as VacA [3131. Cover TL, Puryear W, Perez-Perez GI, Blaser MJ. Effect of urease on HeLa cell vacuolation induced by Helicobacter pylori cytotoxin. Infect Immun. 1991 Apr;59(4):1264-70. ], a hemolysin [3232. Figueroa-Arredondo P, Heuser JE, Akopyants NS, Morisaki JH, Giono-Cerezo S, Enrı́quez-Rincón F, et al. Cell vacuolation caused by Vibrio cholerae hemolysin. Infect Immun. 2001 Mar;69(3):1613-24. ] and VCF [3333. Martins LM, Catani CF, Falcón RM, Carbonell GV, Azzoni AA, Yano T. Induction of apoptosis in Vero cells by Aeromonas veronii biovar sobria vacuolating cytotoxic factor. FEMS Immunol Med Microbiol. 2007 Mar;49(2):197-204. ], respectively. In addition to the characteristic vacuolization previously described, in this study we took advantage of microscopy and in vitro assays to clarify how ECVF induces changes in avian fibroblasts.

Cytotoxins can work by modulating host cells to promote colonization and maintenance of the bacteria in host tissues. According to Cover and Blanke [1414. Cover TL, Blanke SR. Helicobacter pylori VacA, a paradigm for toxin multifunctionality. Nat Rev Microbiol. 2005 Apr;3(4):320-32. ] bacteria that produce multifunctional toxins can use a single protein to obtain a range of actions in different tissues and host cells, enabling these pathogens to remodel tissues, allowing them to escape the host immune system defenses. Our results indicate that ECVF is a multifunctional toxin, damaging cell membrane, cytoplasm, organelles and nucleus.

Cytoskeleton alterations could be accessed using actin stained by labeled Phalloidin. Both CEC-32 and CEF cells show retraction of actin filaments when treated with ECVF. After 18h of cytotoxin exposure, fibroblasts had an intense contraction of the actin cytoskeleton, which had led to loss of adherence with the cell support and with neighboring cells (Figure 1), disrupting the cell monolayer. Noteworthy, cytoskeleton retraction is an important event during the early stages of apoptosis [3434. Ndozangue-Touriguine O, Hamelin J, Bréard J. Cytoskeleton and apoptosis. Biochem Pharmacol. 2008 Jul 1;76(1):11-8. ,3535. Desouza M, Gunning PW, Stehn JR. The actin cytoskeleton as a sensor and mediator of apoptosis. BioArchitecture. 2012 May 1;2(3):75-87. ] and it could also contribute to a nuclear response via mechanoregulation (see below). Another alteration observed during ECVF treatment in living CEF cells, is related to the vacuole fluorescence. Vacuoles induced by VacA are strongly stained by AO [1818. Catrenich CE, Chestnut MH. Character and origin of vacuoles induced in mammalian cells by the cytotoxin of Helicobacter pylori. J Med Microbiol. 1992 Dec;37(6):389-95. ], although ECVF’s vacuoles are unstained. Moreover, it is possible to see an evolution during the time course, as after the initial 6h, few orange spots are observed (Figure 2B), probably due to the AO penetration on lysosomes, full of acid content. However, after 18h and 24h the whole cytoplasm becomes red orange (Figure 2), indicating a loss in compartmentalization of these acid organelles.

To better understand what was going on inside the cells, CEC-32 were incubated with ECVF and observed by electron microscopy. It was possible to detect the presence of structures inside the vacuoles (Figure 3D). Similar structures were observed by Catrenich and Chestnut [1818. Catrenich CE, Chestnut MH. Character and origin of vacuoles induced in mammalian cells by the cytotoxin of Helicobacter pylori. J Med Microbiol. 1992 Dec;37(6):389-95. ] in vacuoles induced by VacA in HeLa cells. These authors suggested that these structures are degraded cytoplasmic components, suggesting that the vacuoles are, actually, autophagosomes.

Our results for the MTT test are similar to previously described for HeLa cells and VacA toxin [3636. Willhite DC, Cover TL, Blanke SR. Cellular vacuolation and mitochondrial cytochrome c release are independent outcomes of Helicobacter pylori vacuolating cytotoxin activity that are each dependent on membrane channel formation. J Biol Chem. 2003 Nov 28;278(48):48204-9. ]. VacA requested more time for loss in mitochondrial activity than to the appearance of vacuoles. As well, here we showed that most fibroblasts (over 50%, data not shown) were vacuolated after 6 hours, while the acute loss on mitochondrial viability occurred after 36 hours of ECVF incubation (Figure 4A). Willhite et al. [3636. Willhite DC, Cover TL, Blanke SR. Cellular vacuolation and mitochondrial cytochrome c release are independent outcomes of Helicobacter pylori vacuolating cytotoxin activity that are each dependent on membrane channel formation. J Biol Chem. 2003 Nov 28;278(48):48204-9. ], demonstrated that the development of mitochondrial alterations in response to intracellular actions of VacA, caused cytochrome c release and activation of pro-apoptotic caspases. Likewise, our data shows that cytoplasmic membrane of CEF cells lost integrity, releasing approximately 67 units of LDH.mL-1, at 12 hours of treatment (Figure 4B). This means that ECVF induces several damages in avian fibroblasts, increasing cell death over time.

In addition to the evidences mentioned above, we used In situ detection of fragmented DNA (TUNEL assay) to confirm the cell death process induced by ECVF. Furthermore, electron micrograph showed a significant nuclear disorganization, with euchromatin condensation and nuclear lobulation (Figure 3C-D), whilst remarkably DNA fragmentation was confirmed by TUNEL (Figure 5A and 5B). Some authors [3737. Farber A, Connors JP, Friedlander RM, Wagner RJ, Powell RJ, Cronenwett JL. A specific inhibitor of apoptosis decreases tissue injury after intestinal ischemia-reperfusion in mice. J Vasc Surg. 1999 Oct;30(4):752-60. -3939. Horn F, Corrêa AMR, Barbieri NL, Glodde S, Weyrauch KD, Kaspers B, et al. Infections with avian pathogenic and fecal Escherichia coli strains display similar lung histopathology and macrophage apoptosis. PLoS One. 2012;25;7(7):e41031. ] reported the relationship between apoptosis and tissue damage, suggesting that this type of cell death may be involved in the pathogenesis of some diseases caused by bacteria. VacA-induced cell death has been classified as an apoptotic process by several authors [4040. Cho SJ, Kang NS, Park SY, Kim BO, Rhee DK, Pyo S. Induction of apoptosis and expression of apoptosis related genes in human epithelial carcinoma cells by Helicobacter pylori VacA toxin. Toxicon. 2003 Nov;42(6):601-11. -4242. Yamasaki E, Wada A, Kumatori A, Nakagawa I, Funao J, Nakayama M, et al. Helicobacter pylori vacuolating cytotoxin induces activation of the proapoptotic proteins Bax and Bak, leading to cytochrome c release and cell death, independent of vacuolation. J Biol Chem. 2006 Apr 21;281(16):11250-9. ]. However, it was later demonstrated that VacA, in fact, induced a programmed necrosis in gastric epithelial cells [4343. Radin JN, González-Rivera C, Ivie SE, McClain MS, Cover TL. Helicobacter pylori VacA induces programmed necrosis in gastric epithelial cells. Infect Immun. 2011 Jul;79(7):2535-43. ]. Indeed, apoptosis and programmed necrosis, share mitochondrial-induced alterations and DNA damage, which were both observed in this study. As a matter of fact, mitochondria, play a central role in both modes of induced cell death, despite different cascades needs to be activated to lead the cell to death by either programmed necrosis or apoptosis [4444. Lamkanfi M, Dixit VM. Manipulation of host cell death pathways during microbial infections. Cell Host Microbe. 2010 Jul 22;8(1):44-54. ,4545. Rudel T, Kepp O, Kozjak-Pavlovic V. Interactions between bacterial pathogens and mitochondrial cell death pathways. Nat Rev Microbiol. 2010 Oct;8(10):693-705. ]. ECVF induces death in avian fibroblasts, albeit the pathway by which it occurs remains to be elucidated.

The specificity between toxins and their host cells is extremely important to understanding the mechanisms involved in bacterial pathogenicity. According to Sewald et al. [4646. Sewald X, Fischer W, Haas R. Sticky socks: Helicobacter pylori VacA takes shape. Trends Microbiol. 2008 Mar;16(3):89-92. ], VacA binds to specific receptors present in human cells that allows the toxin to bind to the cytoplasmic membrane, to be internalized and translocated, promoting vacuolization. Considering that ECVF could have a biological activity similar to that of VacA, it is reasonable to suggest that ECVF also has a specific receptor in avian cells that needed to be investigated. Hence, in this study, cytoplasmic proteins were identified in CEF (Figure 6), possibly involved with the intracellular damage caused by the toxin (Table 1). ECVF associates to alpha-actinin 4 and vinculin, both involved in cytoskeleton structure and maintenance. In this context, it is possible that ECVF acts on the nucleus via mechanoregulation, even before it is internalized. As reviewed by Hah and Kim [4747. Hah J, Kim DH. Deciphering nuclear mechanobiology in laminopathy. Cells. 2019 Mar 11;8(3):231. ], the nucleus can be regulated via changes in the cytoskeleton (by mechanosensing and mechanotransduction). Alpha-actinin is described as anchoring actin to a variety of intracellular structures and it is probably associated to vesicular trafficking [4848. Honda K, Yamada T, Endo R, Ino Y, Gotoh M, Tsuda H, et al. Actinin-4, a novel actin-bundling protein associated with cell motility and cancer invasion. J Cell Biol. 1998 Mar 23;140(6):1383-93. -5050. Nakatsuji H, Nishimura N, Yamamura R, Kanayama H, Sasaki T. Involvement of Actinin-4 in the recruitment of JRAB/MICAL-L2 to cell-cell junctions and the formation of functional tight junctions. Mol Cell Biol. 2008 May;28(10):3324-35. ]. Vinculin, is an F-actin binding protein involved in cell-matrix adhesion and cell-cell adhesion [5151. Zhang Z, Izaguirre G, Lin SY, Lee HY, Schaefer E, Haimovich B. The phosphorylation of vinculin on tyrosine residues 100 and 1065, mediated by Src kinases, affects cell spreading. Mol Biol Cell. 2004 Sep;15(9):4234-47.]. Although chicken alpha-actinin 4 and vinculin are very similar to their human orthologs (more than 90% of identity), ECVF was able to bind only to proteins presents in the avian cell lysate, suggesting that these protein-protein interactions could be important to the early stages of cellulitis infection and internalization of ECVF. This hypothesis is supported by the fact that this toxin does not induce vacuolization in mammalian cells [1111. Salvadori MR, Yano T, Carvalho HF, Parreira VR, Gyles CL. Vacuolating cytotoxin produced by avian pathogenic Escherichia coli. Avian Dis. 2001 Jan-Mar;45(1):43-51. ], indicating that ECVF has a species-specific mode of action. Our results pave the way for deciphering the complete mechanism by which ECVF helps the bacteria to invade the avian cell and open the possibility to explore the relevance of ECVF in the pathogenesis of avian cellulitis, by future screening studies to deciphering which gene is responsible for encoding this toxin.

Table 1.
List of the proteins identified by MS, probably involved in ECVF pathogenicity.

Conclusion

In summary, our data confirms that ECVF is a multifunctional toxin, which causes a range of damages, culminating in cell death. The cytoskeleton disorganization and morphological changes induced by ECVF may play an important role in the initial steps of bacterial infection in avian cellulitis. Therefore, we consider that the ECVF toxin could be a target to drugs and/or vaccines against cellulitis infection in poultry, alternatively to the use of antibiotics.

Abbreviations

AO: acridine orange; APEC: avian pathogenic E. coli; BSA: bovine serum albumin; CEC-32: chicken embryo cell-32; CEF: chicken embryo fibroblast; DAPI: 4’,6-diamidino-2-phenylindole; ECVF: E. coli vacuolating factor; EMEM: Eagle´s minimal essential medium; ExPEC: extra intestinal pathogenic E. coli; FBS: fetal bovine serum; FITC: fluorescein-5-isothiocyanate; HUVEC: human umbilical vein endothelial cells; LDH: lactate dehydrogenase; MS: mass spectrometry; MTT: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; PAI: pathogenicity island; PBS: phosphate buffer saline; PI: propidium iodide; RT: room temperature; SDS: sodium dodecyl sulfate; SDS-PAGE: sodium dodecyl sulfate and polyacrylamide gel electrophoresis; TEM: transmission electron microscopy; TRITC: tetramethylrhodamine B isothiocyanate; TUNEL: terminal deoxynucleotidyl transferase dUTP nick end labeling; VacA: vacuolating cytotoxin A; Vat: vacuolating autotransporter toxin; VF: virulence factor.

Acknowledgments

We would like to thank the technical assistance of Ana Stella Menegon Degrossoli, Ana Lucia Rodrigues da Soledade and Shirlei (from Fort Dodge Saúde Animal). Thanks are also due to (1) Laboratório de Microscopia Eletrônica (LME IB-Unicamp); (2) Laboratório de Matriz Extracelular e Regulação da Expressão Gênica (IB-Unicamp); and (3) Center for Research Facilities (CEFAP ICB-USP) for the assistance in the TEM, FAS and MS procedures, respectively.

References

  • 1. Dho-Moulin M, Fairbrother JM. Avian pathogenic Escherichia coli (APEC). Vet Res. 1999 Mar-Jun;30(2-3):299-316.
  • 2. Dziva F, Stevens MP. Colibacillosis in poultry: unravelling the molecular basis of virulence of avian pathogenic Escherichia coli in their natural hosts. Avian Pathol. 2008 Aug;37(4):355-66.
  • 3. Guabiraba R, Schouler C. Avian colibacillosis: still many black holes. FEMS Microbiol Lett. 2015 Aug;362(15):fnv118.
  • 4. Moulin-Schouleur M, Reperant M, Laurent S, Bree A, Mignon-Grasteau S, Germon P, et al. Extraintestinal pathogenic Escherichia coli strains of avian and human origin: link between phylogenetic relationships and common virulence patterns. J Clin Microbiol. 2007 Oct;45(10):3366-76.
  • 5. Ewers C, Antão EM, Diehl I, Philipp HC, Wieler LH. Intestine and environment of the chicken as reservoirs for extraintestinal pathogenic Escherichia coli strains with zoonotic potential. Appl Environ Microbiol. 2009 Jan 1;75(1):184-92.
  • 6. Bauchart P, Germon P, Brée A, Oswald E, Hacker J, Dobrindt U. Pathogenomic comparison of human extraintestinal and avian pathogenic Escherichia coli - Search for factors involved in host specificity or zoonotic potential. Microb Pathog. 2010 Sep;49(3):105-15.
  • 7. Mellata M. Human and avian extraintestinal pathogenic Escherichia coli: infections, zoonotic risks, and antibiotic resistance trends. Foodborne Pathog Dis. 2013 Nov;10(11):916-32.
  • 8. Rodriguez-Siek KE, Giddings CW, Doetkott C, Johnson TJ, Fakhr MK, Nolan LK. Comparison of Escherichia coli isolates implicated in human urinary tract infection and avian colibacillosis. Microbiology. 2005 Jun;151(Pt 6):2097-110.
  • 9. Zhao L, Gao S, Huan H, Xu X, Zhu X, Yang W, et al. Comparison of virulence factors and expression of specific genes between uropathogenic Escherichia coli and avian pathogenic E. coli in a murine urinary tract infection model and a chicken challenge model. Microbiology. 2009 May;155(Pt 5):1634-44.
  • 10. Janben T, Schwarz C, Preikschat P, Voss M, Philipp HC, Wieler LH. Virulence-associated genes in avian pathogenic Escherichia coli (APEC) isolated from internal organs of poultry having died from colibacillosis. Int J Med Microbiol. 2001 Nov;291(5):371-8.
  • 11. Salvadori MR, Yano T, Carvalho HF, Parreira VR, Gyles CL. Vacuolating cytotoxin produced by avian pathogenic Escherichia coli Avian Dis. 2001 Jan-Mar;45(1):43-51.
  • 12. Parreira VR, Gyles CL. A novel pathogenicity island integrated adjacent to the thrW tRNA gene of avian pathogenic Escherichia coli encodes a vacuolating autotransporter toxin. Infect Immun. 2003 Sep;71(9):5087-96.
  • 13. Cover TL, Blaser MJ. Purification and characterization of the vacuolating toxin from Helicobacter pylori J Biol Chem. 1992 May;267(15):10570-5.
  • 14. Cover TL, Blanke SR. Helicobacter pylori VacA, a paradigm for toxin multifunctionality. Nat Rev Microbiol. 2005 Apr;3(4):320-32.
  • 15. Quel NG, Aragão AZB, Salvadori MR, Farias AS, Joazeiro PP, Santos LMB, et al. Cellulitis lesions in broiler chickens are induced by Escherichia coli Vacuolating Factor (ECVF). Vet Microbiol. 2013 Mar 23;162(2-4):866-72.
  • 16. Peighambari SM, Julian RJ, Vaillancourt JP, Gyles CL. Escherichia coli cellulitis: experimental infections in broiler chickens. Avian Dis. 1995 Jan-Mar;39(1):125-34.
  • 17. Navarro-García F, Sears C, Eslava C, Cravioto A, Nataro JP. Cytoskeletal effects induced by pet, the serine protease enterotoxin of enteroaggregative Escherichia coli Infect Immun. 1999 May;67(5):2184-92.
  • 18. Catrenich CE, Chestnut MH. Character and origin of vacuoles induced in mammalian cells by the cytotoxin of Helicobacter pylori J Med Microbiol. 1992 Dec;37(6):389-95.
  • 19. Borenfreund E, Babich H, Martin-Alguacil N. Comparisons of two in vitro cytotoxicity assays-The neutral red (NR) and tetrazolium MTT tests. Toxicol In Vitro. 1988;2(1):1-6.
  • 20. Mitchell DB, Santone KS, Acosta D. Evaluation of cytotoxicity in cultured cells by enzyme leakage. J Tissue Cult Methods. 1980 Sep;6(3-4):113-6.
  • 21. Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976 May 7;72(1-2):248-54.
  • 22. Laemmli UK. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature. 1970 Aug 15;227(5259):680-5.
  • 23. Renesto P, Samson L, Ogata H, Azza S, Fourquet P, Gorvel JP, et al. Identification of two putative rickettsial adhesins by proteomic analysis. Res Microbiol. 2006 Sep;157(7):605-12.
  • 24. Messier S, Quessy S, Robinson Y, Devriese LA, Hommez J, Fairbrother JM. Focal dermatitis and cellulitis in broiler chickens: bacteriological and pathological findings. Avian Dis. 1993 Jul-Sep;37(3):839-44.
  • 25. Ghanbarpour R, Salehi M, Oswald E. Virulence genotyping of Escherichia coli isolates from avian cellulitis in relation to phylogeny. Comp Clin Pathol. 2010 Apr;19(2):147-53.
  • 26. Barbieri NL, Oliveira AL de, Tejkowski TM, Pavanelo DB, Rocha DA, Matter LB, et al. Genotypes and pathogenicity of cellulitis isolates reveal traits that modulate APEC virulence. PLoS One. 2013 Aug 19;8(8):e72322.
  • 27. Gomis SM, Watts T, Riddell C, Potter AA, Allan BJ. Experimental reproduction of Escherichia coli cellulitis and septicemia in broiler chickens. Avian Dis. 1997 Jan-Mar;41(1):234-40.
  • 28. Norton RA, Bilgili SF, McMurtrey BC. A reproducible model for the induction of avian cellulitis in broiler chickens. Avian Dis. 1997 Apr-Jun;41(2):422-8.
  • 29. de Brito BG, Gaziri LCJ, Vidotto MC. Virulence factors and clonal relationships among Escherichia coli strains isolated from broiler chickens with cellulitis. Infect Immun. 2003 Jul;71(7):4175-7.
  • 30. Ewers C, Jansen T, Kiessling S, Philipp HC, Wieler LH. Molecular epidemiology of avian pathogenic Escherichia coli (APEC) isolated from colisepticemia in poultry. Vet Microbiol. 2004 Nov 30;104(1-2):91-101.
  • 31. Cover TL, Puryear W, Perez-Perez GI, Blaser MJ. Effect of urease on HeLa cell vacuolation induced by Helicobacter pylori cytotoxin. Infect Immun. 1991 Apr;59(4):1264-70.
  • 32. Figueroa-Arredondo P, Heuser JE, Akopyants NS, Morisaki JH, Giono-Cerezo S, Enrı́quez-Rincón F, et al. Cell vacuolation caused by Vibrio cholerae hemolysin. Infect Immun. 2001 Mar;69(3):1613-24.
  • 33. Martins LM, Catani CF, Falcón RM, Carbonell GV, Azzoni AA, Yano T. Induction of apoptosis in Vero cells by Aeromonas veronii biovar sobria vacuolating cytotoxic factor. FEMS Immunol Med Microbiol. 2007 Mar;49(2):197-204.
  • 34. Ndozangue-Touriguine O, Hamelin J, Bréard J. Cytoskeleton and apoptosis. Biochem Pharmacol. 2008 Jul 1;76(1):11-8.
  • 35. Desouza M, Gunning PW, Stehn JR. The actin cytoskeleton as a sensor and mediator of apoptosis. BioArchitecture. 2012 May 1;2(3):75-87.
  • 36. Willhite DC, Cover TL, Blanke SR. Cellular vacuolation and mitochondrial cytochrome c release are independent outcomes of Helicobacter pylori vacuolating cytotoxin activity that are each dependent on membrane channel formation. J Biol Chem. 2003 Nov 28;278(48):48204-9.
  • 37. Farber A, Connors JP, Friedlander RM, Wagner RJ, Powell RJ, Cronenwett JL. A specific inhibitor of apoptosis decreases tissue injury after intestinal ischemia-reperfusion in mice. J Vasc Surg. 1999 Oct;30(4):752-60.
  • 38. Parolin MB, Reason IJM. Apoptose como mecanismo de lesão nas doenças hepatobiliares. Arq Gastroenterol. 2001 Apr;38(2):138-44.
  • 39. Horn F, Corrêa AMR, Barbieri NL, Glodde S, Weyrauch KD, Kaspers B, et al. Infections with avian pathogenic and fecal Escherichia coli strains display similar lung histopathology and macrophage apoptosis. PLoS One. 2012;25;7(7):e41031.
  • 40. Cho SJ, Kang NS, Park SY, Kim BO, Rhee DK, Pyo S. Induction of apoptosis and expression of apoptosis related genes in human epithelial carcinoma cells by Helicobacter pylori VacA toxin. Toxicon. 2003 Nov;42(6):601-11.
  • 41. Cover TL, Krishna US, Israel DA, Peek Jr RM. Induction of gastric epithelial cell apoptosis by Helicobacter pylori vacuolating cytotoxin. Cancer Res. 2003 Mar 1;63(5):951-7.
  • 42. Yamasaki E, Wada A, Kumatori A, Nakagawa I, Funao J, Nakayama M, et al. Helicobacter pylori vacuolating cytotoxin induces activation of the proapoptotic proteins Bax and Bak, leading to cytochrome c release and cell death, independent of vacuolation. J Biol Chem. 2006 Apr 21;281(16):11250-9.
  • 43. Radin JN, González-Rivera C, Ivie SE, McClain MS, Cover TL. Helicobacter pylori VacA induces programmed necrosis in gastric epithelial cells. Infect Immun. 2011 Jul;79(7):2535-43.
  • 44. Lamkanfi M, Dixit VM. Manipulation of host cell death pathways during microbial infections. Cell Host Microbe. 2010 Jul 22;8(1):44-54.
  • 45. Rudel T, Kepp O, Kozjak-Pavlovic V. Interactions between bacterial pathogens and mitochondrial cell death pathways. Nat Rev Microbiol. 2010 Oct;8(10):693-705.
  • 46. Sewald X, Fischer W, Haas R. Sticky socks: Helicobacter pylori VacA takes shape. Trends Microbiol. 2008 Mar;16(3):89-92.
  • 47. Hah J, Kim DH. Deciphering nuclear mechanobiology in laminopathy. Cells. 2019 Mar 11;8(3):231.
  • 48. Honda K, Yamada T, Endo R, Ino Y, Gotoh M, Tsuda H, et al. Actinin-4, a novel actin-bundling protein associated with cell motility and cancer invasion. J Cell Biol. 1998 Mar 23;140(6):1383-93.
  • 49. Yan Q, Sun W, Kujala P, Lotfi Y, Vida TA, Bean AJ. CART: an Hrs/actinin-4/BERP/myosin V protein complex required for efficient receptor recycling. Mol Biol Cell. 2005 May;16(5):2470-82.
  • 50. Nakatsuji H, Nishimura N, Yamamura R, Kanayama H, Sasaki T. Involvement of Actinin-4 in the recruitment of JRAB/MICAL-L2 to cell-cell junctions and the formation of functional tight junctions. Mol Cell Biol. 2008 May;28(10):3324-35.
  • 51. Zhang Z, Izaguirre G, Lin SY, Lee HY, Schaefer E, Haimovich B. The phosphorylation of vinculin on tyrosine residues 100 and 1065, mediated by Src kinases, affects cell spreading. Mol Biol Cell. 2004 Sep;15(9):4234-47.
  • Availability of data and materials

    The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.
  • Funding

    This work was supported by the State of São Paulo Research Foundation (FAPESP - proc. n. 2011/03586-0 to NGQ), the Coordination for the Improvement of Higher Education Personnel (CAPES - to AZBA).
  • Ethics approval

    Not applicable.
  • Consent for publication

    Not applicable.

Publication Dates

  • Publication in this collection
    05 Mar 2021
  • Date of issue
    2021

History

  • Received
    17 July 2020
  • Accepted
    25 Jan 2021
Centro de Estudos de Venenos e Animais Peçonhentos (CEVAP/UNESP) Av. Universitária, 3780, Fazenda Lageado, Botucatu, SP, CEP 18610-034, Brasil, Tel.: +55 14 3880-7693 - Botucatu - SP - Brazil
E-mail: editorial.jvatitd@unesp.br