A defrosted aliquot of bacteria was mixed with an equal volume of n-butanol (Merck) under stirring for 30 min at room temperature (RT). After centri-fugation at 13,000 g for 20 min, the aquatic phase was lyophilized, resuspended with chromatography start buffer (15% n-propanol in 0.1 M ammonium acetate, pH 4.7), and centrifuged at 45,000 g for 15 min. The supernatant was subjected to hydrophobic interaction chromatography (HIC) on octyl-Sepharose.
>
Chemicals & Drugs
>
Organic Chemical
>
Lipoteichoic acid
Lipoteichoic acid
Lipoteichoic acid is a major component of the cell walls of Gram-positive bacteria.
It plays a key role in bacterial adhesion, immune response modulation, and other cellular processes.
PubCompare.ai's AI-driven tools can help optimize your Lipoteichoic acid research by streamlining protocol comparisons, identifying the best methods and products, and accelerating your studies.
Experiecnce a more efficient research process today with PubCompare.ai.
It plays a key role in bacterial adhesion, immune response modulation, and other cellular processes.
PubCompare.ai's AI-driven tools can help optimize your Lipoteichoic acid research by streamlining protocol comparisons, identifying the best methods and products, and accelerating your studies.
Experiecnce a more efficient research process today with PubCompare.ai.
Most cited protocols related to «Lipoteichoic acid»
1-Propanol
ammonium acetate
Bacteria
Buffers
Butyl Alcohol
Chromatography
Hydrophobic Interactions
octyl-sepharose CL-4B
Mice. C57BL/6J specific pathogen free (SPF) mice were purchased from The Jackson Laboratory (Bar Harbor, ME, USA). Gender matched mice between 8–12 weeks of age were used for each experiment.
Reagents and antibodies. Lipoteichoic acid (LTA, S. aureus origin), peptidoglycan (PGN, S. aureus origin), Pam3CSK4, sparstolonin B (SsnB), phorbol 12-myristate 13-acetate (PMA), ionomycin, ovalbumin (OVA) and α-galactosylceramide (α-GalCer) were all purchased from Sigma Aldrich (St Louis, MO, USA). Dispase and collagenase were purchased from Thermo Fisher Scientific (Waltham, MA, USA). Percoll was purchased from GE Healthcare (Chicago, IL, US). The Cytofix/Cytoperm kit was purchased from BD Bioscience (Franklin Lakes, NJ, USA). Recombinant murine granulocyte macrophage-colony stimulating factor (rmGM-CSF) was purchased from Peprotech (Rocky Hill, NJ, USA). Anti-CD11c (N418), anti-CD11b (M1/70), anti-CD207 (4C7), anti-CD103 (2E7), anti-CCR7 (4B12), anti-CLA (HECA-452), anti-CD80 (16-010A1), anti-CD86 (GL-1), anti-CD45 (30-F11), anti-CD3 (17A2), anti-CD4 (GK1.5), anti-IL-17A (BL168), anti-CD16/CD32 (2.4G2) (93), and 5-(and -6)-carboxyfluorescein diacetate succinimidyl ester (CFSE) were all purchased from Biolegend (San Diego, CA, USA). Anti-MHC-II (M5/114.15.2), anti-CD11b (M1/70), anti-TCRβ (H57–597), anti-ɤδTCR (GL3), anti-IFN-γ (XMG1.2), anti-IL-4 (8D4–8), and anti-Toll-like receptor 2 (TLR2) were all purchased from Thermo Fisher Scientific (Waltham, MA, USA). CD1d-tetramer was purchased from Proimmune (Oxford, UK). Anti-MHC class I (HB159) and anti-MHC class II (M5/114) was purchased from Bio X Cell (West Lebanon, NH, USA). The isotype-matched control for each antibody was purchased from the same company.
S. aureus culture. The frozen S. aureus (MRSA; USA300) stock was thawed on ice, then transferred to a tryptic soy broth (TSB; BD bioscience, Franklin Lakes, NJ, USA) and cultured at 37 °C for 18 hr with shaking. The colony forming units (CFU) were calculated in each culture. Heat-killed S. aureus (HK-SA) was prepared with heating at 95 °C for 30 min. The heated S. aureus suspension was centrifuged at 10,000 rpm for 1 min to harvest the bacteria cells, then the cell pellet was resuspended in phosphate buffered saline (PBS) or 0.9% NaCl.
Lipoprotein isolation and preparation of the cell wall component from S. aureus. Lipoprotein was isolated from S. aureus by following a method described in previous reports with modifications [18 (link),19 (link),23 (link)]. Briefly, cultured S. aureus (107–8 CFU/mL) was harvested by centrifuging at 5000× g for 20 min. The pellet was washed twice by 20 mM Tris-HCl (pH 8.0). The pellet was resuspended in 20 mM Tris–HCl (pH 8.0), then the bacterial cell was crushed with 0.3 mm stainless beads. The treated suspension was centrifuged at 5000× g for 20 min, then the supernatant was harvested as the protein suspension. The suspension was mixed with 100% ethanol and kept at −20 °C overnight. The sample was centrifuged at 12,000× g for 15 min, then the precipitated pellet was washed with 80% ethanol and centrifuged again at 12,000× g for 5 min. The precipitated pellet was dissolved with 1 M urea/50 mM Tris–HCl, 50 mM ethylenediaminetetraacetic acid (EDTA) (pH 8.0) (Crude Protein Extract; CPE). Triton X-114 was added to the protein suspension (final 1%), then the suspension was incubated at 4 °C with gentle mixing. The incubated suspension was heated at 37 °C, forming the micelle phase-containing lipoprotein. The micelle phase was extracted and lipoprotein (Clude S. aureus-lipoprotein; SA-LP) was harvested by following a method for CPE precipitation. The SA-LP was separated to each fraction (L1 to L4) in a size dependent manner by using a molecular weight cut-off filter (Amicon ultra; Darmstadt, Germany). For preparation of the cell wall extract (CWE), the twice-washed S. aureus pellet was resuspended in 20 mM Tris–HCl (pH 8.0). The suspension was kept at −80 °C for 30 min, then sonicated for 20 min. The suspension was centrifuged at 5000× g for 20 min, and the pellet was harvested as CWE.
Sodium dodecyl sulfate-poly-acrylamide gel electrophoresis (SDS-PAGE) and silver stain. The extracted protein solution was diluted with 5 × SDS sample buffer (2% SDS, 62.5 mM Tris–HCl (pH 6.8), 10% glycerol, 0.01% bromophenol blue, 50 mM dithiothreitol (DTT). The proteins, separated by SDS-PAGE, were visualized with a Silver Stain Kit (Thermo Fisher Scientific, Waltham, MA, USA). Whole staining procedure was followed with the manual.
Mouse primary cell isolation. Skin leukocytes were isolated by following a method described in a previous report with modification [47 (link)]. Briefly, the extracted ear was washed with tissue washing buffer (RPMI 1640 supplemented with 10% fetal bovine serum (FBS), 10 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) and 100 U/mL penicillin, 100 mg/mL streptomycin) at 37 °C for 30 min with gentle shaking. The ear was separated into the ventral and dorsal sheets from the cartilage, and incubated at 4 °C overnight with dispase working solution (tissue washing buffer containing 0.25 mg/mL of dispase) to separate the epidermal and dermal sheets. These sheets were chopped with scissors, then incubated at 37 °C for 30 min in collagenase working solution (tissue washing buffer containing 1 mg/mL collagenase and 0.01% DNase). The digested ear pieces were passed through a 5 mL syringe with a 22 G needle to make single cell suspensions. Lymph node cells were prepared from skin-draining LN (dLN) by following a method described in a previous report [48 (link)]. Briefly, isolated dLN was crushed on a dish and suspended in cell culture medium. The cell suspension was filtered through a 70 μm cell strainer, then twice washed with cell culture medium (RPMI 1640 supplemented with 10% fetal bovine serum (FBS), 100 U/mL penicillin and 100 mg/mL streptomycin). Splenocytes were obtained from the spleen by following a method described in a previous report [48 (link)]. Briefly, isolated spleen was crushed on a 70 μm cell strainer, and the cells were suspended in cell culture medium. After being washed once with the cell culture medium, the cells were further resuspended in an erythrocyte lysis solution (155 mM NH4Cl, 10 mM KHCO3, 1 mM Na-EDTA, and 17 mM Tris–HCl (pH 7.3)). After being washed twice with cell culture medium, the cells were used as splenocytes. Mouse bone marrow leukocytes were obtained from the tibia and femur. After extracting the tibia and femur, bone marrow leukocytes were flushed out with a syringe containing cell culture medium. The cell suspension was filtered through a 70 μm cell strainer and washed once with cell culture medium, then the cells were treated with the erythrocytes lysis solution. After lysis, the cells were washed twice with cell culture medium, and the cells were used as bone marrow leukocytes. Pan-naïve T cells were isolated from the splenocyte by using an EasySep Mouse Pan-Naïve T Cell Isolation Kit (Stemcell Technology; Vancouver, BC, Canada). Naïve CD4+ and CD8+T cells were isolated from the splenocyte by using a MagniSort mouse CD4 naïve T cell or mouse CD8 naïve T cell Enrichment kit (Thermo Fisher Scientific, Waltham, MA, USA), respectively. LN and splenic dendritic cells were isolated by using MagniSort Mouse CD11c Positive Selection kit (Thermo Fisher Scientific, Waltham, MA, USA). The whole procedure for the cell isolation kit was performed by following the manual.
Mouse BMDCs preparation. Mouse BMDCs were prepared by following a method described in a previous report [49 (link)]. At day 0, 2.0 × 106 of bone marrow leukocytes were suspended in 10 mL of DC culture medium (cell culture medium containing 20 ng/mL of rmGM-CSF), and the cells were seeded on a 100 mm dish. At day 3, 10 mL of the fresh DC culture medium was added to the cultured cells. At days 6 and 8, half of the cultured medium was collected and centrifuged, then the cell pellets were resuspended in 10 mL of the fresh DC culture medium. The cell suspension was put back into the original plate. At day 10, cells were ready to use for each experiment.
BMDC stimulation assay. Naïve BMDCs were seeded on a 6-well plate with DC culture medium, then the cells were stimulated by the S. aureus-derived component, LTA (10 μg/mL), PGN (10 μg/mL), SA-LP (10 μg/mL or identical amounts with isolated from 106 of S. aureus), CWE (identical amount with isolated from 106 of S. aureus), CPE (10 μg/m or identical amount with isolated from 106 of S. aureus) and live-SA (106) and HK-SA (106) for 24 hr. The TLR2 agonist Pam3CSK4 (500 ng/mL) and TLR4 agonist LPS (100 ng/mL) were used for the positive control. TLR2 was blocked by the anti-mouse TLR2 monoclonal antibody (mAB) (10 μg/mL) or inhibited by Ssn B (100 μM). The stimulated cells were analyzed by flow cytometry for the detection of activation markers (CD80, DC86 and MHC class II). Cultured medium was harvested for the measurement of cytokine production by ELISA.
Flow cytometry. Cell surface markers and intracellular cytokines were analyzed by a flow cytometer (FACScalibur and LSR-II; BD Biosciences, Franklin Lakes, NJ, USA) with the fluorochrome-conjugated monoclonal antibodies described in reagents and antibodies. The cells were initially incubated with FcR blocker (anti-CD16/32; 2.4G2) at 4 °C for 10 min. For surface marker staining, the cells were incubated with the antibody at 4 °C for 30 min. Intracelluler cytokine staining was performed by using a Cytofix/CytoPerm Kit (BD Biosciences, Franklin Lakes, NJ, USA) by following the manual. Briefly, the cells stained with the antibody for the surface marker were fixed and permilized. The cells were incubated with the antibody for cytokine staining at 4 °C for 30 min. The dead cells were excluded by forward scatter, side scatter, and propidium iodide gating. All data were analyzed by BD FACS Diva (BD bioscience, Franklin Lakes, NJ, USA) or FlowJo (Tree Star; Ashland, OR, USA).
Murine skin inflammation model. To establish the skin inflammation model, anesthetized mice were treated with the antigen by intradermal (ID) injection into the ear (Figure S2 ). The antigen was dissolved or diluted with 0.9% saline, then applied into the dorsal side of the ear. After 48 or 120 hr of the treatment, the mice were sacrificed and the treated ear and skin-dLN was excised for use in each analysis.
In vivo antigen tracking. For antigen tracking, FITC-labeled SA-LP and OVA or non-labeled OVA (10 μg in each) were ID-injected into the mouse ear. After 48 hr of the treatment, the skin dLN was extracted from the treated mice, then the isolated LN cells were analyzed by flow cytometry.
Ex vivo antigen re-stimulation. The skin leukocytes were isolated from the SA-LP ID-injected mice ears, then the cells were labeled by CFSE. The labeled cells were cocultured with stimulated splenic DCs (SA-LP 1 μg/mL or LPS 100 ng/mL + OVA 10 μg/mL) or naïve splenic DCs at 37 °C for 24 hr. The proliferated cells were analyzed by flow cytometry.
In vitro antigen presentation. Isolated primary DCs were stimulated with SA-LP (10 μg/mL) overnight. The stimulated or naive DCs were cocultured with pan-naïve T, naïve CD4+, or CD8+T cells for 72 hr. At the last 6 hr of the coculture, the proliferated cells were re-stimulated with 100 ng/mL of PMA, 1 μg/mL of ionomycin, and protein transportation was inhibited with Golgi stop (BD Bioscience). The proliferated cells were analyzed by flow cytometry.
In vitro MHC blocking assay. MHC class I and II molecules on the DCs were blocked with antibodies by following a method described in a previous report with modification [50 (link)]. Briefly, LN isolated DCs were pre-incubated with the blocking antibody for MHC class I or MHC class II (10 μg/mL in each) for 1 hr. An isotype antibody was also used for the control. Then, the cells were stimulated with SA-LP (10 μg/mL) overnight. The stimulated DCs were cocultured with splenic naïve CD4+ or CD8+T cells for 72 hr in the presence of the blocking antibody or isotype antibody. At the last 6 hr of the coculture, the proliferated cells were re-stimulated with 100 ng/mL of PMA, 1 μg/mL of ionomycin, and protein transportation was inhibited with Golgi stop (BD Bioscience). The cells were analyzed by flow cytometry.
SA-LP-activated dendritic cell transfer. The primary DCs were isolated from WT mouse LN, then the cells were labeled with CFSE and stimulated with SA-LP (10 μg/mL) at 37 °C overnight. The treated DCs were washed with cell culture medium three times, then the cells (2.0 × 106) were transferred into WT mice ears by ID injection. After 48 and 120 hr of the treatment, the skin-dLN and ear were used for each analysis.
Cytokine measurement by Enzyme-Linked Immuno Sorbent Assay (ELISA). The cytokine (TNF-α, IL-12p40 and IL-6) produced from the stimulated cell was measured by using a Mouse ELISA kit (Thermo Fisher Scientific, Waltham, MA, USA) for each target. The whole procedure was performed by following the manual.
Statistical analyses. A Student’s t-test was used to analyze the data for significant differences. Values of * p < 0.05, ** p < 0.01, and *** p < 0.001 were regarded as significant.
Reagents and antibodies. Lipoteichoic acid (LTA, S. aureus origin), peptidoglycan (PGN, S. aureus origin), Pam3CSK4, sparstolonin B (SsnB), phorbol 12-myristate 13-acetate (PMA), ionomycin, ovalbumin (OVA) and α-galactosylceramide (α-GalCer) were all purchased from Sigma Aldrich (St Louis, MO, USA). Dispase and collagenase were purchased from Thermo Fisher Scientific (Waltham, MA, USA). Percoll was purchased from GE Healthcare (Chicago, IL, US). The Cytofix/Cytoperm kit was purchased from BD Bioscience (Franklin Lakes, NJ, USA). Recombinant murine granulocyte macrophage-colony stimulating factor (rmGM-CSF) was purchased from Peprotech (Rocky Hill, NJ, USA). Anti-CD11c (N418), anti-CD11b (M1/70), anti-CD207 (4C7), anti-CD103 (2E7), anti-CCR7 (4B12), anti-CLA (HECA-452), anti-CD80 (16-010A1), anti-CD86 (GL-1), anti-CD45 (30-F11), anti-CD3 (17A2), anti-CD4 (GK1.5), anti-IL-17A (BL168), anti-CD16/CD32 (2.4G2) (93), and 5-(and -6)-carboxyfluorescein diacetate succinimidyl ester (CFSE) were all purchased from Biolegend (San Diego, CA, USA). Anti-MHC-II (M5/114.15.2), anti-CD11b (M1/70), anti-TCRβ (H57–597), anti-ɤδTCR (GL3), anti-IFN-γ (XMG1.2), anti-IL-4 (8D4–8), and anti-Toll-like receptor 2 (TLR2) were all purchased from Thermo Fisher Scientific (Waltham, MA, USA). CD1d-tetramer was purchased from Proimmune (Oxford, UK). Anti-MHC class I (HB159) and anti-MHC class II (M5/114) was purchased from Bio X Cell (West Lebanon, NH, USA). The isotype-matched control for each antibody was purchased from the same company.
S. aureus culture. The frozen S. aureus (MRSA; USA300) stock was thawed on ice, then transferred to a tryptic soy broth (TSB; BD bioscience, Franklin Lakes, NJ, USA) and cultured at 37 °C for 18 hr with shaking. The colony forming units (CFU) were calculated in each culture. Heat-killed S. aureus (HK-SA) was prepared with heating at 95 °C for 30 min. The heated S. aureus suspension was centrifuged at 10,000 rpm for 1 min to harvest the bacteria cells, then the cell pellet was resuspended in phosphate buffered saline (PBS) or 0.9% NaCl.
Lipoprotein isolation and preparation of the cell wall component from S. aureus. Lipoprotein was isolated from S. aureus by following a method described in previous reports with modifications [18 (link),19 (link),23 (link)]. Briefly, cultured S. aureus (107–8 CFU/mL) was harvested by centrifuging at 5000× g for 20 min. The pellet was washed twice by 20 mM Tris-HCl (pH 8.0). The pellet was resuspended in 20 mM Tris–HCl (pH 8.0), then the bacterial cell was crushed with 0.3 mm stainless beads. The treated suspension was centrifuged at 5000× g for 20 min, then the supernatant was harvested as the protein suspension. The suspension was mixed with 100% ethanol and kept at −20 °C overnight. The sample was centrifuged at 12,000× g for 15 min, then the precipitated pellet was washed with 80% ethanol and centrifuged again at 12,000× g for 5 min. The precipitated pellet was dissolved with 1 M urea/50 mM Tris–HCl, 50 mM ethylenediaminetetraacetic acid (EDTA) (pH 8.0) (Crude Protein Extract; CPE). Triton X-114 was added to the protein suspension (final 1%), then the suspension was incubated at 4 °C with gentle mixing. The incubated suspension was heated at 37 °C, forming the micelle phase-containing lipoprotein. The micelle phase was extracted and lipoprotein (Clude S. aureus-lipoprotein; SA-LP) was harvested by following a method for CPE precipitation. The SA-LP was separated to each fraction (L1 to L4) in a size dependent manner by using a molecular weight cut-off filter (Amicon ultra; Darmstadt, Germany). For preparation of the cell wall extract (CWE), the twice-washed S. aureus pellet was resuspended in 20 mM Tris–HCl (pH 8.0). The suspension was kept at −80 °C for 30 min, then sonicated for 20 min. The suspension was centrifuged at 5000× g for 20 min, and the pellet was harvested as CWE.
Sodium dodecyl sulfate-poly-acrylamide gel electrophoresis (SDS-PAGE) and silver stain. The extracted protein solution was diluted with 5 × SDS sample buffer (2% SDS, 62.5 mM Tris–HCl (pH 6.8), 10% glycerol, 0.01% bromophenol blue, 50 mM dithiothreitol (DTT). The proteins, separated by SDS-PAGE, were visualized with a Silver Stain Kit (Thermo Fisher Scientific, Waltham, MA, USA). Whole staining procedure was followed with the manual.
Mouse primary cell isolation. Skin leukocytes were isolated by following a method described in a previous report with modification [47 (link)]. Briefly, the extracted ear was washed with tissue washing buffer (RPMI 1640 supplemented with 10% fetal bovine serum (FBS), 10 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) and 100 U/mL penicillin, 100 mg/mL streptomycin) at 37 °C for 30 min with gentle shaking. The ear was separated into the ventral and dorsal sheets from the cartilage, and incubated at 4 °C overnight with dispase working solution (tissue washing buffer containing 0.25 mg/mL of dispase) to separate the epidermal and dermal sheets. These sheets were chopped with scissors, then incubated at 37 °C for 30 min in collagenase working solution (tissue washing buffer containing 1 mg/mL collagenase and 0.01% DNase). The digested ear pieces were passed through a 5 mL syringe with a 22 G needle to make single cell suspensions. Lymph node cells were prepared from skin-draining LN (dLN) by following a method described in a previous report [48 (link)]. Briefly, isolated dLN was crushed on a dish and suspended in cell culture medium. The cell suspension was filtered through a 70 μm cell strainer, then twice washed with cell culture medium (RPMI 1640 supplemented with 10% fetal bovine serum (FBS), 100 U/mL penicillin and 100 mg/mL streptomycin). Splenocytes were obtained from the spleen by following a method described in a previous report [48 (link)]. Briefly, isolated spleen was crushed on a 70 μm cell strainer, and the cells were suspended in cell culture medium. After being washed once with the cell culture medium, the cells were further resuspended in an erythrocyte lysis solution (155 mM NH4Cl, 10 mM KHCO3, 1 mM Na-EDTA, and 17 mM Tris–HCl (pH 7.3)). After being washed twice with cell culture medium, the cells were used as splenocytes. Mouse bone marrow leukocytes were obtained from the tibia and femur. After extracting the tibia and femur, bone marrow leukocytes were flushed out with a syringe containing cell culture medium. The cell suspension was filtered through a 70 μm cell strainer and washed once with cell culture medium, then the cells were treated with the erythrocytes lysis solution. After lysis, the cells were washed twice with cell culture medium, and the cells were used as bone marrow leukocytes. Pan-naïve T cells were isolated from the splenocyte by using an EasySep Mouse Pan-Naïve T Cell Isolation Kit (Stemcell Technology; Vancouver, BC, Canada). Naïve CD4+ and CD8+T cells were isolated from the splenocyte by using a MagniSort mouse CD4 naïve T cell or mouse CD8 naïve T cell Enrichment kit (Thermo Fisher Scientific, Waltham, MA, USA), respectively. LN and splenic dendritic cells were isolated by using MagniSort Mouse CD11c Positive Selection kit (Thermo Fisher Scientific, Waltham, MA, USA). The whole procedure for the cell isolation kit was performed by following the manual.
Mouse BMDCs preparation. Mouse BMDCs were prepared by following a method described in a previous report [49 (link)]. At day 0, 2.0 × 106 of bone marrow leukocytes were suspended in 10 mL of DC culture medium (cell culture medium containing 20 ng/mL of rmGM-CSF), and the cells were seeded on a 100 mm dish. At day 3, 10 mL of the fresh DC culture medium was added to the cultured cells. At days 6 and 8, half of the cultured medium was collected and centrifuged, then the cell pellets were resuspended in 10 mL of the fresh DC culture medium. The cell suspension was put back into the original plate. At day 10, cells were ready to use for each experiment.
BMDC stimulation assay. Naïve BMDCs were seeded on a 6-well plate with DC culture medium, then the cells were stimulated by the S. aureus-derived component, LTA (10 μg/mL), PGN (10 μg/mL), SA-LP (10 μg/mL or identical amounts with isolated from 106 of S. aureus), CWE (identical amount with isolated from 106 of S. aureus), CPE (10 μg/m or identical amount with isolated from 106 of S. aureus) and live-SA (106) and HK-SA (106) for 24 hr. The TLR2 agonist Pam3CSK4 (500 ng/mL) and TLR4 agonist LPS (100 ng/mL) were used for the positive control. TLR2 was blocked by the anti-mouse TLR2 monoclonal antibody (mAB) (10 μg/mL) or inhibited by Ssn B (100 μM). The stimulated cells were analyzed by flow cytometry for the detection of activation markers (CD80, DC86 and MHC class II). Cultured medium was harvested for the measurement of cytokine production by ELISA.
Flow cytometry. Cell surface markers and intracellular cytokines were analyzed by a flow cytometer (FACScalibur and LSR-II; BD Biosciences, Franklin Lakes, NJ, USA) with the fluorochrome-conjugated monoclonal antibodies described in reagents and antibodies. The cells were initially incubated with FcR blocker (anti-CD16/32; 2.4G2) at 4 °C for 10 min. For surface marker staining, the cells were incubated with the antibody at 4 °C for 30 min. Intracelluler cytokine staining was performed by using a Cytofix/CytoPerm Kit (BD Biosciences, Franklin Lakes, NJ, USA) by following the manual. Briefly, the cells stained with the antibody for the surface marker were fixed and permilized. The cells were incubated with the antibody for cytokine staining at 4 °C for 30 min. The dead cells were excluded by forward scatter, side scatter, and propidium iodide gating. All data were analyzed by BD FACS Diva (BD bioscience, Franklin Lakes, NJ, USA) or FlowJo (Tree Star; Ashland, OR, USA).
Murine skin inflammation model. To establish the skin inflammation model, anesthetized mice were treated with the antigen by intradermal (ID) injection into the ear (
In vivo antigen tracking. For antigen tracking, FITC-labeled SA-LP and OVA or non-labeled OVA (10 μg in each) were ID-injected into the mouse ear. After 48 hr of the treatment, the skin dLN was extracted from the treated mice, then the isolated LN cells were analyzed by flow cytometry.
Ex vivo antigen re-stimulation. The skin leukocytes were isolated from the SA-LP ID-injected mice ears, then the cells were labeled by CFSE. The labeled cells were cocultured with stimulated splenic DCs (SA-LP 1 μg/mL or LPS 100 ng/mL + OVA 10 μg/mL) or naïve splenic DCs at 37 °C for 24 hr. The proliferated cells were analyzed by flow cytometry.
In vitro antigen presentation. Isolated primary DCs were stimulated with SA-LP (10 μg/mL) overnight. The stimulated or naive DCs were cocultured with pan-naïve T, naïve CD4+, or CD8+T cells for 72 hr. At the last 6 hr of the coculture, the proliferated cells were re-stimulated with 100 ng/mL of PMA, 1 μg/mL of ionomycin, and protein transportation was inhibited with Golgi stop (BD Bioscience). The proliferated cells were analyzed by flow cytometry.
In vitro MHC blocking assay. MHC class I and II molecules on the DCs were blocked with antibodies by following a method described in a previous report with modification [50 (link)]. Briefly, LN isolated DCs were pre-incubated with the blocking antibody for MHC class I or MHC class II (10 μg/mL in each) for 1 hr. An isotype antibody was also used for the control. Then, the cells were stimulated with SA-LP (10 μg/mL) overnight. The stimulated DCs were cocultured with splenic naïve CD4+ or CD8+T cells for 72 hr in the presence of the blocking antibody or isotype antibody. At the last 6 hr of the coculture, the proliferated cells were re-stimulated with 100 ng/mL of PMA, 1 μg/mL of ionomycin, and protein transportation was inhibited with Golgi stop (BD Bioscience). The cells were analyzed by flow cytometry.
SA-LP-activated dendritic cell transfer. The primary DCs were isolated from WT mouse LN, then the cells were labeled with CFSE and stimulated with SA-LP (10 μg/mL) at 37 °C overnight. The treated DCs were washed with cell culture medium three times, then the cells (2.0 × 106) were transferred into WT mice ears by ID injection. After 48 and 120 hr of the treatment, the skin-dLN and ear were used for each analysis.
Cytokine measurement by Enzyme-Linked Immuno Sorbent Assay (ELISA). The cytokine (TNF-α, IL-12p40 and IL-6) produced from the stimulated cell was measured by using a Mouse ELISA kit (Thermo Fisher Scientific, Waltham, MA, USA) for each target. The whole procedure was performed by following the manual.
Statistical analyses. A Student’s t-test was used to analyze the data for significant differences. Values of * p < 0.05, ** p < 0.01, and *** p < 0.001 were regarded as significant.
Lipoteichoic acid and protein detection by Western blot was undertaken essentially as previously described (Gründling and Schneewind, 2007b ). In brief, for sodium dodecyl sulphate (SDS)-polyacrylamide gel electrophoresis (PAGE) and Western blot analysis of cell-associated LTA and His-tagged proteins, 1 ml of overnight culture was mixed with 0.5 ml of 0.1 mm glass beads and lysed by vortexing for 45 min in the cold. Glass beads were sedimented by centrifugation at 200 g for 1 min, and 0.5 ml of the resultant supernatant transferred to a fresh tube. Bacterial debris and LTA were sedimented by centrifugation at 17 000 g for 15 min and suspended in protein sample buffer containing 2% SDS normalized for OD600; that is, samples from a culture with an OD600 of 2 were suspended in 50 μl of sample buffer. Samples were boiled for 20 min, centrifuged at 17 000 g for 5 min and 10 μl of samples loaded onto SDS-PAA gels. To determine the amount of LTA shed into the culture medium, 500 μl of culture was first centrifuged at 17 000 g for 5 min to pellet bacteria. Culture supernatant (100 μl) was removed, mixed with 100 μl of 2× protein sample buffer, boiled for 30 min and insoluble material removed by centrifugation at 17 000 g for 5 min. Supernatant samples were normalized based on OD600 of 2, in that 10 μl of a culture of OD600 of 2 was loaded. To determine if the His-tagged proteins were shed into the supernatant, 1.4 ml of culture was centrifuged at 17 000 g for 10 min to pellet the bacteria. One millilitre of the supernatant was transferred to a new tube, mixed with 100 μl of 100% trichloroacetic acid (TCA), vortexed, incubated on ice for 1 h and centrifuged for 10 min at 17 000 g. The supernatant was aspirated and the TCA precipitated pellet was washed twice with 1 ml of ice-cold acetone. Between wash steps, samples were incubated on ice for 1 h and debris collected by centrifugation as described above. After the final centrifugation step, pellets were air dried and suspended in 2× protein sample buffer normalized for OD600; that is, samples from a culture with an OD600 of 2 were suspended in 100 μl of sample buffer. The samples were boiled for 30 min and 10 μl analysed by Western blot. LTA samples were routinely loaded onto 15% SDS-PAA gels and probed with polyglycerolphosphate-specific LTA antibody (Clone 55 from Hycult biotechnology) and HRP-conjugated anti-mouse IgG (Cell Signalling Technologies, USA) used at 1:2000 and 1:10 000 dilutions respectively. His-tagged protein samples were routinely loaded onto 10% SDS-PAA gels and probed with HRP-conjugated His-tag-specific antibody (Sigma) used at a 1:10 000 dilution and Western blots were developed by enhanced chemiluminesce (ECL). Western blots were performed with at least three independently grown cultures in at least two independent experiments and representative images are shown.
Mice were infected as described previously10 (link). Briefly, mice were anesthetized and intranasally inoculated with approximately 105 CFU of K. pneumoniae or S. pneumoniae in 50 μL of PBS. To determine bacterial load in the lung, mice were killed, lungs removed, homogenized in PBS, and plated on appropriate media. For survival curves, mice were killed when they exhibited two or more of the following signs of systemic pneumococcal infection: reduced movement, hunched posture, piloerection, shivering, dyspnea, or circling. No animal exhibited any of these signs of infection for more than 24 h. For innate stimulation of the lung, mice were anesthetized as above then intranasally administered with 1 μg of K. pneumoniae LPS (Sigma) or 50 μg lipoteichoic acid (Sigma). Alveolar macrophages were depleted using clodronate containing liposomes as described previously47 (link). Antibodies and recombinant proteins were administered at amounts/concentration and times stated in figure legends. For microbiota transfer, unanesthetized mice were intranasally inoculated with 10 μL of upper respiratory tract lavage fluid and orally inoculated with 200 μL of fecal suspension 3 days prior to lung infection. Upper respiratory tract lavage fluid was prepared by lavaging the upper airway of non-antibiotic-treated mice, as described in “Quantification of bacterial load in gastrointestinal and upper airway microbiota,” the lavage fluid was centrifuged at 16,000 × g to pellet bacteria in the lavage fluid, the supernatant discarded and the pellet resuspended in 10 μL of sterile PBS. The fecal suspension was prepared by suspending a single fecal pellet from non-antibiotic-treated mice in 1 mL of sterile PBS. For oral inoculation of bacterial consortia, mice were orally gavaged at indicated time points with either consortia resuspended in sterile PBS. All bacteria were grown to mid-log phase. For intranasal inoculation of “High” and “Low” NLR-stimulating bacteria, unanesthetized mice were administered indicated bacteria in 20 μL, 48 h prior to lung infection. Because of the small inoculation volume and lack of anesthesia these bacteria remain in the upper airway48 (link), 49 (link).
S. aureus (DSM 20233) was cultured aerobically in a 42-liter fermentor (MBR Bio Reactor) at 37°C and harvested at an OD578 of 15 (extrapolated) in a continuous flow centrifuge, resuspended in 0.1 M citrate buffer, pH 4.7, and disrupted with glass beads in a Braun disintegrator. Standard hot phenol/water extractions followed by fast performance liquid chromatography (FPLC) of aqueous extracts on octyl-Sepharose (Amersham Pharmacia Biotech) and DEAE–Sepharose (Amersham Pharmacia Biotech) were performed according to the procedure described in reference
2-diethylaminoethanol
Buffers
Citrate
Fermentors
Liquid Chromatography
octyl-sepharose CL-4B
Phenol
Sepharose
Staphylococcus aureus
Most recents protocols related to «Lipoteichoic acid»
Bacterial pellets of B. subtilis ATCC 6633 were harvested by centrifugation at 6,200 × g (8,000 rpm) for 10 min at 4°C and washed with PBS (pH 7.0). LTA was isolated as previously described [21] (link). The bacterial pellets of B. subtilis were resuspended in 0.1 M sodium citrate buffer (pH 4.7) and disrupted using ultrasonication for 2 h at a frequency of 20 kHz with stirring. Subsequently, the bacterial lysates were mixed vigorously with an equal volume of n-butanol and the aqueous phase was collected by centrifugation at 10,075 × g (13,000 rpm) for 15 min at room temperature. The collected aqueous phase was dialyzed using a semipermeable dialysis membrane of 1-kDa molecular cutoff (Spectrum Laboratories, Rancho Domingueguz, CA, USA) in endotoxin-free distilled water (Daihan Pahrm. Co. Ltd., Seoul, Korea). Following the dialysis process, the extract was prepared with a 15% n-propanol concentration in 0.1 M sodium acetate buffer and subjected to hydrophobic interaction chromatography using an octyl-Sepharose column (GE Healthcare, Chicago, IL, USA) to obtain fractions containing LTA. Unbound substances were removed through washing with 20% n-propanol in 0.1 M sodium acetate buffer, followed by the elution of LTA-containing fractions in 35% n-propanol with 0.1 M sodium acetate buffer using a fraction collector (Bio-Rad, Hercules, CA, USA). Then, the column fractions containing phosphates were consolidated, dialyzed, and prepared with 30% n-propanol in 0.1 M sodium acetate buffer for an ion-exchange chromatography with DEAE-Sepharose (Sigma-Aldrich). The fractions were subsequently eluted using a linear salt gradient ranging from 0 to 1 M NaCl in the equilibration buffer. The LTA-containing fractions were pooled, dialyzed, and subjected to lyophilization under vacuum (5 Torr; 24 h). The isolated LTA was quantified by measuring its dry weight, and experimental dose was established according to the previous study [22] (link).
Referred to the proteomic results, six related proteins, lipoteichoic acid synthase (Q2G093), signal peptidase I (Q2FZT7), teichoic acids export ATP-binding protein TagH (Q2G2L1), lipoprotein (Q2G0V0), UDP-N-acetylmuramoyl-tripeptide-D-alanyl-D-alanine ligase (Q2FWH4), and D-alanine-D-alanine ligase (Q2FWH3), were selected for further verifiable quantification for three samples from the azalomycin F group or the blank one, using the parallel reaction monitoring (PRM) technology. According to the similar procedure to proteome analysis, the peptides from tryptic hydrolysis were dissolved in 0.1% (v/v) formic acid and separated on an EASY-nLC 1000 UPLC system with a reversed phase analytical column. The gradient elution consisted of an increase from 7% to 25% solvent B in 40 min, 22% to 35% in 12 min, 35% to 80% in 4 min, and then holding at 80% for the last 4 min. The analysis process was achieved at a constant flow rate of 0.4 μL/min. The separated peptides were subjected to an NSI source followed by MS/MS analysis in a Q ExactiveTM Plus (Thermo Fisher Scientific, San Jose, CA, USA). The electrospray voltage applied was 2.0 kV. The full scan of m/z ranged from 400 to 1000, and intact peptides were detected in the Orbitrap at a resolution of 70,000. The peptides were then selected for MS/MS analysis using an NCE setting as 27, and the fragments were detected in the Orbitrap at a resolution of 17,500. The data-dependent procedure alternated between one MS scan followed by 20 MS/MS scans. The AGC was set at 3e6 for full MS and 1e5 for MS/MS, respectively. The maximum IT was set at 50 ms for full MS and 180 ms for MS/MS. The isolation window for MS/MS was set at 1.6 m/z.
The resulting MS data were processed using Skyline (v.3.6). Peptide parameters were set as trypsin (KR|P) for the enzyme, zero for the max missed cleavage, 7 to 25 residues for the peptide length, and alkylation on Cys for fixed modification. Transition parameters were set as 2 and 3 for precursor charges, 1 for ion charges, and b and y for ion types. The product ions were set from ion 3 to the last ion. The mass tolerance of ion match was set as 0.02 Da.
The resulting MS data were processed using Skyline (v.3.6). Peptide parameters were set as trypsin (KR|P) for the enzyme, zero for the max missed cleavage, 7 to 25 residues for the peptide length, and alkylation on Cys for fixed modification. Transition parameters were set as 2 and 3 for precursor charges, 1 for ion charges, and b and y for ion types. The product ions were set from ion 3 to the last ion. The mass tolerance of ion match was set as 0.02 Da.
Protocol full text hidden due to copyright restrictions
Open the protocol to access the free full text link
The paraffin-embedded tissues were sectioned into 3-μm-thick slides. According to standard histological protocols, hematoxylin and eosin (HE) staining was conducted. For immunohistochemistry (IHC), the following primary antibodies were used to incubate the slides, respectively, after blocking: bacterial lipopolysaccharide (LPS; HycultBiotech #HM6011, Netherlands), lipoteichoic acid (LTA; Santa Cruz, #sc-57752, United States), occludin (Servicebio #GB111401, China), and zonula occludens 1 (ZO-1; Servicebio #GB111981, China). IHC quantification involved the calculation of positive areas (occludin and ZO-1) using Aipathwell software (Version 2.0, Servicebio, China).
Cell culture Raw 264.7 monocytes (1×10 6 (link) ) were seeded into 12-well plates. Cells were incubated in media alone, or 1:1000 dilutions of sterile-filtered faecal homogenate (100 mg/mL) from CS-exposed or air-exposed mice for 24 hours. During the final 4 hours, LPS (1 µg/mL), monophosphoryl lipid A (4 µg/mL), lipoteichoic acid (1 µg/mL) were added to cell media. Tumour necrosis factor (TNF)-α protein in culture media was assessed using DuoSet ELISA kits (R&D Systems).
Top products related to «Lipoteichoic acid»
Sourced in United States, Germany
Lipoteichoic acid (LTA) is a cell wall component found in Gram-positive bacteria. It serves as a structural element and plays a role in maintaining the integrity of the bacterial cell wall. LTA is commonly used in research applications to study the immune response and cell signaling pathways in bacterial infections.
Sourced in United States, China, United Kingdom, Germany, Australia, Japan, Canada, Italy, France, Switzerland, New Zealand, Brazil, Belgium, India, Spain, Israel, Austria, Poland, Ireland, Sweden, Macao, Netherlands, Denmark, Cameroon, Singapore, Portugal, Argentina, Holy See (Vatican City State), Morocco, Uruguay, Mexico, Thailand, Sao Tome and Principe, Hungary, Panama, Hong Kong, Norway, United Arab Emirates, Czechia, Russian Federation, Chile, Moldova, Republic of, Gabon, Palestine, State of, Saudi Arabia, Senegal
Fetal Bovine Serum (FBS) is a cell culture supplement derived from the blood of bovine fetuses. FBS provides a source of proteins, growth factors, and other components that support the growth and maintenance of various cell types in in vitro cell culture applications.
Sourced in Norway, United States, France
Lipoteichoic acid is a component of the cell wall of certain Gram-positive bacteria. It plays a role in the structural integrity and function of the bacterial cell wall.
Sourced in United States, Germany, China, United Kingdom, Sao Tome and Principe, Macao, Italy, Japan, Canada, France, Switzerland, Israel, Australia, Spain, India, Ireland, Brazil, Poland, Netherlands, Sweden, Denmark, Hungary, Austria, Mongolia
The LPS laboratory equipment is a high-precision device used for various applications in scientific research and laboratory settings. It is designed to accurately measure and monitor specific parameters essential for various experimental procedures. The core function of the LPS is to provide reliable and consistent data collection, ensuring the integrity of research results. No further details or interpretations can be provided while maintaining an unbiased and factual approach.
Sourced in United States
Lipoteichoic acid (LTA) is a cell wall component found in Gram-positive bacteria. It serves as a structural element and plays a role in maintaining cell integrity.
Sourced in United States, France, Canada
Lipoteichoic acid is a component of the cell wall of Gram-positive bacteria. It functions as a structural component and plays a role in bacterial adhesion and host immune responses.
Sourced in United States, Germany, United Kingdom, Israel, Canada, Austria, Belgium, Poland, Lao People's Democratic Republic, Japan, China, France, Brazil, New Zealand, Switzerland, Sweden, Australia
GraphPad Prism 5 is a data analysis and graphing software. It provides tools for data organization, statistical analysis, and visual representation of results.
Sourced in United States
Flagellin is a structural protein found in the flagella of bacteria. It is a key component of the bacterial flagellum, which is responsible for the motility of many bacterial species. Flagellin plays a crucial role in the assembly and function of the flagellum, enabling bacteria to move and navigate their environment.
Sourced in United States, France, United Kingdom, Canada, Germany, Sweden, Italy, Macao
Pam3CSK4 is a synthetic triacylated lipopeptide that mimics the structure of the acylated amino terminus of bacterial lipoproteins. It acts as a potent agonist of Toll-like receptor 2 (TLR2) and can be used in cell-based assays to study TLR2-mediated cellular responses.
Sourced in United States, United Kingdom, Germany, China, France, Canada, Japan, Australia, Switzerland, Italy, Israel, Belgium, Austria, Spain, Brazil, Netherlands, Gabon, Denmark, Poland, Ireland, New Zealand, Sweden, Argentina, India, Macao, Uruguay, Portugal, Holy See (Vatican City State), Czechia, Singapore, Panama, Thailand, Moldova, Republic of, Finland, Morocco
Penicillin is a type of antibiotic used in laboratory settings. It is a broad-spectrum antimicrobial agent effective against a variety of bacteria. Penicillin functions by disrupting the bacterial cell wall, leading to cell death.
More about "Lipoteichoic acid"
Lipoteichoic acid (LTA) is a crucial component of the cell walls of Gram-positive bacteria, playing a vital role in various cellular processes.
As a major constituent of the bacterial cell wall, LTA is involved in adhesion, immune response modulation, and other important functions.
LTA shares similarities with lipopolysaccharide (LPS), another key bacterial cell wall component found in Gram-negative bacteria.
Both LTA and LPS are known to interact with immune cells, triggering responses that can have significant implications for human health and disease.
Researchers studying Lipoteichoic acid often utilize related compounds and techniques, such as fetal bovine serum (FBS), which is commonly used in cell culture media to support cell growth and proliferation.
Additionally, tools like GraphPad Prism 5, a statistical analysis software, can be helpful in analyzing data related to LTA experiments.
Other bacterial components that may be relevant to Lipoteichoic acid research include Flagellin, a protein found in bacterial flagella, and Pam3CSK4, a synthetic lipopeptide that mimics the structure of bacterial lipoproteins.
These molecules can interact with the immune system and contribute to our understanding of host-pathogen interactions.
Optimizing Lipoteichoic acid research protocols can be facilitated by AI-driven tools like those offered by PubCompare.ai.
These tools can streamline the process of identifying and comparing the best methods and products for your studies, helping to accelerate your research and workflow.
Experiecnce a more efficient research process today with PubCompare.ai.
As a major constituent of the bacterial cell wall, LTA is involved in adhesion, immune response modulation, and other important functions.
LTA shares similarities with lipopolysaccharide (LPS), another key bacterial cell wall component found in Gram-negative bacteria.
Both LTA and LPS are known to interact with immune cells, triggering responses that can have significant implications for human health and disease.
Researchers studying Lipoteichoic acid often utilize related compounds and techniques, such as fetal bovine serum (FBS), which is commonly used in cell culture media to support cell growth and proliferation.
Additionally, tools like GraphPad Prism 5, a statistical analysis software, can be helpful in analyzing data related to LTA experiments.
Other bacterial components that may be relevant to Lipoteichoic acid research include Flagellin, a protein found in bacterial flagella, and Pam3CSK4, a synthetic lipopeptide that mimics the structure of bacterial lipoproteins.
These molecules can interact with the immune system and contribute to our understanding of host-pathogen interactions.
Optimizing Lipoteichoic acid research protocols can be facilitated by AI-driven tools like those offered by PubCompare.ai.
These tools can streamline the process of identifying and comparing the best methods and products for your studies, helping to accelerate your research and workflow.
Experiecnce a more efficient research process today with PubCompare.ai.