Waiting
Login processing...

Trial ends in Request Full Access Tell Your Colleague About Jove

Biology

Detecting Migration and Infiltration of Neutrophils in Mice

Published: February 6, 2020 doi: 10.3791/60543
* These authors contributed equally

Summary

Here, we present three methods to assess neutrophil migration and infiltration both in vivo and in vitro. These methods can be used to discover promising therapeutics targeting neutrophil migration.

Abstract

Neutrophils are a major member of the innate immune system and play pivotal roles in host defense against pathogens and pathologic inflammatory reactions. Neutrophils can be recruited to inflammation sites via the guidance of cytokines and chemokines. Overwhelming infiltration of neutrophils can lead to indiscriminate tissue damage, such as in rheumatoid arthritis (RA). Neutrophils isolated from peritoneal exudate respond to a defined chemoattractant, N-formyl-Met-Leu-Phe (fMLP), in vitro in Transwell or Zigmond chamber assays. The air pouch experiment can be used to evaluate the chemotaxis of neutrophils towards lipopolysaccharide (LPS) in vivo. The adjuvant-induced arthritis (AA) mouse model is frequently used in RA research, and immunohistochemical staining of joint sections with anti-myeloperoxidase (MPO) or anti-neutrophil elastase (NE) antibodies is a well-established method to measure neutrophil infiltration. These methods can be used to discover promising therapies targeting neutrophil migration.

Introduction

Neutrophils are the most abundant white blood cell and account for 50−70% of the whole white blood cell population in humans1. Neutrophils are one of the primary responders during acute inflammation. Neutrophils can be recruited to inflammation sites via the guidance of cytokines and chemokines released by tissue-resident cells2,3,4, which is mediated by the interactions between cell adhesion molecules on the surface of neutrophils and vascular endothelium cells5. Neutrophils are fundamental to host defense and play a role in pathologic inflammatory reactions due to their powerful capacity to damage tissue via the release of reactive oxygen species (ROS) and other tissue-damaging molecules3,6.

Previous studies have described several neutrophil isolation protocols from mice or humans. Oh et al. demonstrated a density gradient separation method to isolate human neutrophils from whole human blood7. However, the isolation of sufficient neutrophils from mouse blood is difficult because of the small blood volume. Alternatively, large numbers of pure and viable mouse neutrophils can be elicited from mouse peritoneal fluid, and these purified neutrophils can be used ex vivo to examine several aspects of cellular functions ex vivo, including neutrophil infiltration, migration, chemotaxis, oxidative burst, cytokine and neutrophil extracellular trap (NET) production8. Transwell assays9 or Zigmond chamber assays10,11 can be used to evaluate neutrophil migration in vitro. The air pouch model is used to evaluate the migration and infiltration of neutrophils in vivo. The subcutaneous air pouch model is a convenient in vivo animal model to study the migration of inflammatory cells.

Traditionally, neutrophils were considered as pathogen eliminators in acute phases of inflammation. However, recent findings have shown that neutrophils are complicated cells that perform a significant variety of specialized functions. Neutrophils can regulate many processes such as acute injury and repair, tumorigenesis, autoimmune response, and chronic inflammation12,13. Neutrophils also modulate adaptive immune responses and can regulate B cells and T cells14,15. Substantial shortage of neutrophils leads to mortality or severe immunodeficiency in humans and neutrophil depletion in mice leads to fatality, while excessive activation or recruitment of neutrophils in organs causes several immune diseases, such as rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE)6. Neutrophils are the most abundant cells in the synovial fluid of RA patients. Neutrophils produce excessive amounts of myeloperoxidase (MPO) and neutrophil elastase (NE) via degradation, which exacerbates cartilage erosion. MPO is a peroxidase enzyme mainly expressed in the granules of neutrophils16. NE is associated with articular cartilage destruction17. MPO and NE could be used to evaluate the status of neutrophil migration and infiltration in the tissue of RA patients.

This article provides three conventional methods to evaluate the migration of normal neutrophils induced both in vivo and in vitro, as well as the infiltration of pathological neutrophils in a mouse joint-specific inflammation model.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

All experimental procedures were reviewed and approved by the Beijing University of Chinese Medicine Animal Care and Use Committee.

NOTE: C57BL/6 mice (7-8 weeks old) were used.

1. Neutrophil isolation

  1. Acquisition of peritoneal exudate cells
    1. Prepare fresh 10% proteose peptone solution in ddH2O. Calculate the volume needed according to the number of mice.
      NOTE: Set the number of mice as N, (2N+1) mL of solution must be dissolved and filtered in advance.
    2. Spray the workspace with 70% ethanol. Use an insulin injector to draw 1 mL of peptone solution and discharge bubbles.
    3. Conduct the first intraperitoneal injection of 1 mL of peptone solution per mouse.
      1. Grab the mouse in a head-down position with one hand. Disinfect the injection spot with alcohol-soaked cotton balls.
        NOTE: The preferred injection position lies in the lateral aspect of the lower left or right quadrant of the abdomen.
      2. Infuse the reagents rapidly. Inject 1 mL into the peritoneal cavity of each mouse with the insulin injector.
        NOTE: The angle between the needle and the skin should be approximately 15−30° to avoid injuring the intestine or other organs.
    4. Allow inflammatory response to develop overnight. After 12 h, conduct the second injection in the same manner as the first injection.
    5. Three hours after the second injection, fully anesthetize mice with 5% isoflurane for 5 min in a gas anesthesia chamber at a speed of 2 L/min. Remove the anesthetized mice from the chamber and sacrifice them by cervical dislocation.
      NOTE: Sufficient depth of anesthesia is ensured by pinching the toes before moving mice from the chamber to the single breathing unit. The legs should not move when the toes are pinched.
      NOTE: All the following steps should be processed in a tissue culture hood.
    6. Spray the mouse with 70% ethanol. Lay the mouse on a sterile plastic pad and fix the limbs with needles.
    7. Use a sterile set of surgical tools to make a horizontal incision (~1 cm) in the middle of the lower abdomen. Lift the skin of the upper abdomen with forceps and cut along the midline of the abdomen and expose the intact peritoneal wall.
    8. Inject 5 mL of sterile RPMI-1640 complete medium into the abdominal cavity with a 30 G x 1/2" needle. Insert the needle through the peritoneal wall with the beveled edge of the needle facing up and inject the entire volume.
    9. Shake the pad horizontally for 5 min. Massage the abdomen gently several times.
    10. Inject a 23 G x 1 1/4" needle into the lateral space of the abdomen. Extract the abdominal liquid (~5 mL) and collect it in a 50 mL centrifuge tube.
      NOTE: Place the tubes on ice as soon as possible in case of neutrophil activation.
    11. Inject another 5 mL of complete medium and repeat the procedure to remove the remaining cells from the peritoneum. Pool the peritoneal fluid in the 50 mL centrifuge tube.
    12. Centrifuge the pooled peritoneal fluid at 400 x g for 10 min at room temperature (RT).
    13. Discard the supernatant. Resuspend the cells in 1 mL of RPMI-1640 complete medium.
      NOTE: Do not vortex to avoid neutrophil activation.
  2. Isolation of neutrophils
    1. Add 4 mL of freshly prepared 70.2% density gradient medium (e.g., Percoll) in a 15 mL centrifuge tube.
    2. Carefully overlay 4 mL of freshly prepared 54.8% density gradient medium on the 70.2% density gradient medium slowly along the edge of the tube with sharp pipette tips in the 15 mL centrifuge tube.
      NOTE: Exercise caution to avoid disturbing the interface between the 54.8% density gradient medium and 70.2% density gradient medium.
    3. Carefully overlay the 1 mL peritoneal cell suspension on top of the 54.8% density gradient medium layer slowly with sharp pipette tips (Figure 1A).
      NOTE: Exercise caution to avoid disturbing the interface between the cell suspension and 54.8% density gradient medium.
    4. Centrifuge at 1,500 x g for 30 min at 22 °C without braking.
    5. Collect the neutrophils at the interface of the 54.8% density gradient medium and 70.2% density gradient medium layers (Figure 1B) to a new tube.
    6. Add 1 mL of RPMI-1640 complete medium to the collected cells and carefully resuspend cells by gently pipetting several times. Centrifuge at 100 x g for 10 min at RT and carefully remove the supernatant.
    7. Repeat the wash step (step 1.2.6) once.
    8. Add 0.5 mL of culture medium to the pellet and resuspend cells by gently pipetting several times. Take a 50 µL aliquot to count the cells using an automatic hematology analyzer.

2. Neutrophil migration assay

  1. Measure neutrophil migration by Transwell assay9 or Zigmond chamber assay as previously described10,11.

3. Air pouch assay

  1. First air injection
    1. On day 0, fully anesthetize mice with 5% isoflurane for 3 min in a gas anesthesia chamber at a speed of 2 L/min, and maintain the anesthesia of each mouse in a single breathing unit with 2% isoflurane at a speed of 0.5 L/min.
      NOTE: Sufficient depth of anesthesia is ensured by pinching the toes before moving mice from the chamber to the single breathing unit. The legs should not move when the toes are pinched.
    2. Use a 0.22 µm filter attached to a 5 mL syringe to obtain a 3 mL volume of sterilized air.
    3. Lift the back skin of the anesthetized mouse with tweezers and subcutaneously inject 3 mL of sterilized air using a 26 G x 3/8" needle.
    4. After treatment, remove the mice from the breathing unit. Monitor the mice to ensure they are alive until they start to move around.
  2. Second air injection
    1. On day 3, inject an additional 3 mL of sterilized air into the previously established air pocket to sustain the air pouch as described in section 3.1.
  3. Treatment
    1. On day 6, 6 h before sacrifice, inject different treatments into the air pouch. Inject 1 mL of phosphate-buffered saline (PBS) as a negative control. Inject 1 mL of 1 µg/mL LPS as the positive control to induce local inflammation.
    2. Fully anesthetize mice with 5% isoflurane for 3 min in a gas anesthesia chamber at a speed of 2 L/min, and maintain the anesthesia of each mouse in a single breathing unit with 2% isoflurane at a speed of 0.5 L/min. Prepare wash buffer according to Table of Materials.
      NOTE: Sufficient depth of anesthesia is ensured by pinching the toes before moving mice from the chamber to the single breathing unit. The legs should not move when the toes are pinched.
    3. For each air pouch, wash the air pouch with 1 mL of wash buffer and collect the inflammatory exudate in a 15 mL centrifuge tube. Wash the air pouch with 2 mL of wash buffer 2x and collect the inflammatory exudate in the same centrifuge tube.
    4. Centrifuge at 100 x g for 10 min at RT. Discard the supernatant and resuspend cells in 1 mL of wash buffer. Count the cells to quantify the neutrophil ratio using the automatic hematology analyzer.
      NOTE: See representative results in Figure 2.

4. Induction of the adjuvant-induced arthritis (AA) mouse model

  1. Suspend complete Freund's adjuvant (CFA) by vortexing at least 5 s, then draw 100 µL of suspension into an insulin injector.
    NOTE: We recommend using completely new CFA in experiments to ensure sterility.
  2. Anesthetize mice as described in step 3.1.1.
  3. Mark the chosen paw and inject 20 µL of CFA into the ankle joint space. Inject 20 µL of suspension into four periarticular spots on the chosen paw (80 µL in total).
  4. Remove mice from the breathing unit and put the processed mice in a new chamber. Monitor mice to ensure that they are breathing until they regain the ability to move.
  5. Every 3 days, assess the joint diameter by measuring the ankle joint diameter using a pocket thickness gauge (Figure 3A).
  6. Every 3 days, assess arthritis severity by arthritis scoring criterion (Figure 3C): 0, normal, no evidence of erythema and swelling; 1, the mildest arthritis, erythema and mild swelling confined to the tarsals or ankle joint; 2, moderate arthritis, erythema and mild swelling extending from the ankle to the tarsals; 3, severe arthritis, erythema and moderate swelling extending from the ankle to metatarsal joints; 4, the most severe arthritis, erythema and severe swelling encompass the ankle, foot and digits, or ankylosis of the limb.

5. Immunohistochemical staining of joint sections

  1. Joint isolation
    1. Sacrifice the mouse in section 4 using cervical dislocation after anesthesia with isoflurane. Spray the mouse with 70% ethanol.
    2. Remove the skin and part of the muscle from the hind leg with tweezers and scissors. Spray the joint with 70% ethanol and remove the rest of the muscles using a paper towel.
    3. Fix the ankle joint in 4% paraformaldehyde for 2 days at RT. Decalcify the joint in 10% EDTA for 1 month at RT and change the medium weekly.
    4. Embed the tissue in paraffin and prepare 4-µm-thick tissue sections.
      1. Place tissue in a marked mold with certain volume of liquid paraffin. Cool briefly.
      2. Set the thickness at 4 µm and cut slices on a microtome. Float sections in a 43 °C water bath.
      3. Mount the sections onto slides and put the slides into the oven at 70 °C for 2 h. For future use, conserve the slides at -20 °C.
  2. Safranin O and fast green staining of joint sections
    NOTE: The following staining steps are conducted at RT.
    1. Place the slides from step 5.1.4.3 in a rack and perform the following washes to rehydrate at RT: xylene for 5 min (3x), 100% ethanol for 2 min (2x), 95% ethanol for 2 min (2x), 70% ethanol for 2 min, and 50% ethanol for 15 min. Wash in running tap water for 3 min.
    2. Stain in 0.1% fast green solution for 5 min. Rinse in 1% acetic acid for 10 s.
    3. Stain in 0.1% safranin O staining solution for 20 min. Immerse the slides in the following washes: 95% ethanol for 2 min (2x), 100% ethanol for 2 min (2x), and xylene for 2 min (2x).
    4. Mount the tissue sections and observe the tissues under a microscope.
      NOTE: See representative images in Figure 4A.
  3. Immunohistochemical staining to visualize neutrophils
    1. Bake the paraffin sections for 2 h at 78 °C. Place the slides in a rack and perform the following washes to rehydrate at RT: xylene for 15 min (2x), 100% ethanol for 5 min (2x), 95% ethanol for 5 min, 80% ethanol for 5 min, H2O for 3 min, and PBS for 3 min.
      NOTE: Do not let slides dry at any time during this step.
    2. Add one drop of permeabilization buffer to cover the tissue. Incubate sections in a humidity-controlled tray at 37 °C.
    3. Rinse slides in PBS for 3 min (3x). Avoid rinsing the tissue directly.
    4. Perform heat-induced antigen epitope retrieval using a pressure boiler.
      1. Arrange slides in a rack. Immerse slides in the pressure boiler filled with retrieval buffer.
      2. Put the pressure boiler on a microwave oven. Set the microwave oven at 600 W and heat the slides for 10 min.
      3. After boiling, keep slides in the boiler to cool to 90 °C. Take out the slides and rinse them in PBS for 3 min (3x).
    5. Quench endogenous peroxidase activity in freshly prepared 3% H2O2 at RT for 15 min. Rinse slides in PBS for 3 min (3x).
    6. Outline a large circle around the sample with a hydrophobic pen, avoid touching the sample. Block with 3% bovine serum albumin (BSA) in a humidity-controlled chamber at 37 °C for 60 min.
    7. Remove blocking solution. Add 50 µL of PBS-diluted primary antibody to each section quickly. Then, incubate the slides in a humidity-controlled tray at 4 °C overnight.
      NOTE: Different dilution ratios are used for different antibodies (1:25 for MPO and 1:20 for NE).
    8. On the second day, take out the tray and let it stand at RT for 30 min. Then, rinse the slides in PBS for 3 min (3x).
    9. Add 50 µL of PBS-diluted secondary antibodies to the tissue.
      NOTE: Different dilution ratios were applied: 1:1,000 for MPO and 1:1,500 for NE.
    10. Incubate slides in a humidity-controlled tray at 37 °C for 30 min. Then, rinse the slides in PBS for 3 min (3x).
    11. Develop in diluted 3,3'-diaminobenzidine (DAB) solution for 5 min. Keep an eye on the reaction in case of the development of a dark color. Rinse the slides in distilled water.
    12. Counterstain the slides in hematoxylin for 10 s. Rinse the slides in tap water for 5 min.
    13. Rinse in acid alcohol superfast differentiation solution for 3 s. Then rinse in tap water for 10 min.
    14. Immerse the slides in the following washes at RT: 80% ethanol for 5 min, 95% ethanol for 5 min, 100% ethanol for 5 min, and xylene for 15 min (2x). Fix the coverslip with mounting solution. Observe the tissue under a microscope.
      NOTE: See representative images in Figure 4B.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

Peritoneal exudate cells were collected from lavage fluid of mice. Cells were resuspended in 1 mL of RPMI-1640 complete medium, layered onto a two-step (54.8%/70.2%) discontinuous density gradient (Figure 1A), and centrifuged at 1,500 x g for 30 min. Neutrophils (≥95%, ~1 x 107 neutrophils/mouse) were recovered from the lower interface (Figure 1B).

Air pouch experiments were performed to investigate the neutrophil recruitment stimulated by LPS in vivo (Figure 2A). The leukocyte subsets in the air pouch exudates were measured (Figure 2B).

Neutrophil migration in RA was evaluated via the CFA-induced arthritis murine model. Compared with the control group, the AA group showed significant edema in the paw. In the AA group, the ankle joint diameter increased (Figure 3B) and the arthritis score rose consistently (Figure 3D).

Cartilage damage is the representative syndrome of RA, safranin O-fast green cartilage staining was performed to assess the cartilage damage in AA mouse. As shown in Figure 4A, CFA challenge induced a large amount of leukocyte infiltration, significant cartilage erosion and synovial hyperplasia. MPO and NE expression levels are representative markers of neutrophil infiltration. Immunohistochemical assays were performed to observe neutrophil infiltration in joints. MPO and NE expression was significantly upregulated in the joint section (Figure 4B).

Figure 1
Figure 1: Neutrophil isolation. (A) Peritoneal exudate cells resuspended in 1 mL of RPMI-1640 complete medium were layered onto a two-step (54.8%/70.2%) discontinuous density gradient. (B) After centrifugation at 1,500 x g for 30 min, neutrophils (≥95%, ~1 x 107 neutrophils/mouse) were recovered from the lower interface. Please click here to view a larger version of this figure.

Figure 2
Figure 2: Representative results of the air pouch assay. (A) Illustration of the air pouch assay. (B) Representative results of leukocyte subset infiltration in the air pouch assay. PBS: control; LPS: 1 µg/mL LPS. Data are presented as the mean ± SD. Please click here to view a larger version of this figure.

Figure 3
Figure 3: Representative results of the adjuvant-induced arthritis (AA) mouse model. (A) The ankle joint diameter was measured using a pocket thickness gauge. (B) Joint swelling was assessed based on the ankle joint diameter (n = 7). (C) Pictures of each arthritis score. (D) The severity of arthritis was graded on a scale of 0−4 points (n = 7). Data are presented as the mean ± SD. Please click here to view a larger version of this figure.

Figure 4
Figure 4: Representative results of safranin O-fast green cartilage staining and immunohistochemical assay of joint sections from control and AA mice. (A) Representative results of safranin O-fast green cartilage staining of joint sections. (B) Representative results of the expression level of MPO and NE in the ankle joints. Please click here to view a larger version of this figure.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

Detailed protocols of highly-purified neutrophils from peripheral blood7, bone marrow and tissues18 have been available for a long time. Here we adopt a method of isolating neutrophils from peritoneal fluid19 in which mature neutrophils remain inactivated for further anti-inflammatory and antioxidant studies.

We used the air pouch experiment to explore the LPS-induced infiltration of neutrophils in vivo. This method has been proposed as a potential method for directly measuring cell infiltration in the general inflammatory environment in vivo.

It is noteworthy that the air pouch assay only demonstrates the neutrophil's function in an organ-nonspecific manner and removes several steps of the leukocyte recruitment cascade. Since neutrophil recruitment to specific organs can rely on different organ properties, adhesion molecules, and chemokines, exploring the neutrophil functional state in organ-specific conditions is of great importance for studying the role that neutrophils potentially play in certain diseases3. It has been suggested that endpoint models are required to investigate neutrophil infiltration. Therefore, performing immunohistochemical staining on the joint sections of mice in the AA model can provide an insightful perspective on the neutrophils in the joint space. According to our data, vast numbers of neutrophils are recruited into the joint tissue and serve as fundamental evidence for further research on interrupting the infiltration of neutrophils to treat RA. In addition, comprehensive assays, for example, safranin O-fast green staining, are required to evaluate the disease model for further study.

Neutrophils are the major subset of infiltrating inflammatory cells and work as the first line of defense against invading pathogens or tissue injury20,21. If neutrophils infiltrate tissues in large numbers, high levels of cytokines and NETs are secreted, which together may overwhelm the protective mechanisms in tissues and lead to tissue damage. Tissue injury further stimulates neutrophil infiltration, thus forming a vicious cycle22. Interfering with cell migration by means of trapping activated cells in lymphoid organs has been proposed as an important therapeutic approach and has been applied in clinical trials with various side effects23. Discovering a novel treatment to concurrently regulate neutrophil migration and inflammatory activity is a promising strategy for treating inflammatory diseases in the future. Furthermore, if motility-regulating agents are combined, neutrophil-mediated drug delivery24 can increase drug specificity, thus decreasing side effects.

However, further modifications can be combined to broaden the application of the above-mentioned assays. For example, in the AA mouse model, to obtain the overall impression of infiltration of inflammatory cells in the joint, researchers are encouraged to enzymatically digest joints to release the resident leukocyte populations and then apply flow cytometry to count the populations.

The protocols herein include three ways to assess neutrophil migration and infiltration. The application of these protocols is useful for discovering potential treatments for RA and other inflammatory diseases involving neutrophils.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

The authors have nothing to disclose.

Acknowledgments

This work was supported by the National Natural Science Foundation of China (grant numbers 81430099 and 31500704), International Cooperation and Exchange Projects (grant number 2014DFA32950), and the research program of Beijing University of Chinese Medicine (grant numbers BUCM-2019-JCRC006 and 2019-JYB-TD013).

Materials

Name Company Catalog Number Comments
0.1% Fast Green Solution Solarbio 8348b Transfer 20 mg of fast grene FCF in one vial into another 100 mL beaker. Add 20 mL of H2O into the beaker and dissolve the stain by stirring to make 0.1% fast green solution, and filter it using a Nalgene PES 75mm filter
0.1% Safranin O Staining Solution Solarbio 8348a Transfer 20 mg of safranin O stain in one vial into a 100 ml beaker. Add 20 mL of H2O into the beaker and dissolve the stain by stirring to make 0.1% safranin O staining solution, and filter it using a Nalgene PES 75mm filter
0.5 M Ethylenediaminetetraacetic acid solution (EDTA), pH 8.0 Sigma 324506 Sterile
100% Ethanol Beijing Chemical Works
100% Methanol Beijing Chemical Works
15 mL Conical Polypropylene Centrifuge Tube Falcon 14-959-53A
23 G x 1 1/4" Needle BD 305120
26 G x 3/8" Needle BD 305110
3% bovine serum albumin (BSA) Dissolve 0.3 g BSA in 10 mL PBS
3% H2O2 Mix 1 mL 30% H2O2 with methanol with 9 mL methanol
3,3'-diaminobenzidine (DAB) kit ZSGB-BIO ZLI-9018
30 G x 1/2" Needle BD 305106
30% H2O2 Beijing Chemical Works
5 mL Syringe BD Z683574
50 mL Conical Polypropylene Centrifuge Tube Falcon 14-432-22
50% Ethanol Mix 500 mL 100% ethanol with 500 mL dH2O
54.8% Percoll Mix 2.74 mL SIP with 2.26 mL 1×PBS, stand still
70% Ethanol Mix 700 mL 100% ethanol with 300 mL dH2O
70.2% Percoll Mix 3.51 mL SIP with 1.49 mL 1×PBS, stand still
80% Ethanol Mix 800 mL 100% ethanol with 200 mL dH2O
95% Ethanol Mix 950 mL 100% ethanol with 50 mL dH2O
Acid Alcohol Superfast Differentiation Solution Beyotime C0165S
ANTIBODIES
Anti-Myeloperoxidase Antibody Abcam ab208670
Anti-Neutrophil Elastase Antibody Abcam ab21595
Automatic Hematology Analyzer Sysmex XS-800i
Bovine Serum Albumin (BSA) VWR 0332-100G
Complete Freund's Adjuvant, 10 mg/ml sigma 1002036152
Cover Slip CITOGLAS 10212432C
Dial Thickness Gauge Mitutoyo 7301
Eppendorf Microtubes, 1.5 mL Sigma Z606340
Foetal Bovine Serum (FBS) Premium PAN P30-1302
Gas Anesthesia System ZS Dichuang ZS-MV-IV
Goat Anti-Rabbit IgG H&L (HRP) PPLYGEN C1309 This is the secondary antibody used in the immunohistochemical staining.
Hank's Balanced Salt Solution (HBSS) Biological Industries, Beth HaEmek, Israel 02-016-1A Sterile
Hematoxylin Staining Solution ZSGB-BIO ZLI-9609
Lipopolysaccharide (LPS) Sigma L3012
MEDIA AND SUPPLEMENTS
Modified Safranin O-fast Green FCF Cartilage Stain Kit Solarbio G1371
N-formyl-Met-Leu-Phe (fMLP) Sigma 47729
Penicillin Streptomycin Solution, 100× Invitrogen 1514022
Percoll GE Healthcare 10245207 Density gradient medium
Permeabilization Buffer Mix 100 μL Triton X-100 with 1 L dH2O to get 0.01% Triton X-100
Phosphate Buffer Saline (PBS), 1× Mix 90% ddH2O with 10% (v/v) 10×PBS, autoclaved
Phosphate Buffer Saline (PBS), 10× Dissolve 16 g NaCl, 0.4 g KCl, 2.88 g Na2HPO2H2O, 0.48 g KH2PO4 (anhydrous) in 200 mL ddH2O, adjust pH 7.4, autoclaved
PLASTIC WARES AND EQUIPMENTS
POWDER
Proteose Peptone Oxoid 1865317
Retrieval Buffer Mix 18 mL retrieval buffer A with 82 mL retrieval buffer B, add dH2O to 1000 mL, adjust pH to 6.0
Retrieval Buffer A Stock for IHC Dissolve 4.2 g citric acid (C6H5OH2O) in 200 mL dH2O
Retrieval Buffer B Stock for IHC Dissolve 5.88 g trisodium citrate dihydrate (C6H5Na3O7·2H20) in 200 mL dH2O
Roswell Park Memorial Institute (RPMI)-1640 medium Sigma R8758
RPMI-1640 Complete Medium RPMI-1640 medium is supplemented with 10% FBS and 1% penicillin/streptomycin.
Shu Rui U40 Disposable Sterile Insulin Injection Needle 1 mL BD 328421
Slide CITOGLAS 10127105P-G
SOLUTION
Stock Isotonic Percoll (SIP) Mix 90% (v/v) of percoll with 10% (v/v) 10×PBS, stand still for 20 min
Wash Buffer in Air Pouch Assay Dilute 0.5M EDTA to 10mM with HBSS
Xylene Beijing Chemical Works

DOWNLOAD MATERIALS LIST

References

  1. Klein, C. Genetic defects in severe congenital neutropenia: emerging insights into life and death of human neutrophil granulocytes. Annual Review of Immunology. 29, 399-413 (2011).
  2. Nuzzi, P. A., Lokuta, M. A., Huttenlocher, A. Analysis of Neutrophil Chemotaxis. Adhesion Protein Protocols. Coutts, A. S. , Humana Press. Totowa, NJ. 23-35 (2007).
  3. Margraf, A., Ley, K., Zarbock, A. Neutrophil Recruitment: From Model Systems to Tissue-Specific Patterns. Trends in Immunology. 40 (7), 613-634 (2019).
  4. Bardoel, B. W., Kenny, E. F., Sollberger, G., Zychlinsky, A. The balancing act of neutrophils. Cell Host & Microbe. 15 (5), 526-536 (2014).
  5. Wright, H. L., Moots, R. J., Bucknall, R. C., Edwards, S. W. Neutrophil function in inflammation and inflammatory diseases. Rheumatology. 49 (9), 1618-1631 (2010).
  6. Thieblemont, N., Wright, H. L., Edwards, S. W., Witko-Sarsat, V. Human neutrophils in auto-immunity. Seminars in Immunology. 28 (2), 159-173 (2016).
  7. Oh, H., Siano, B., Diamond, S. Neutrophil isolation protocol. Journal of Visualized Experiments. (17), e745 (2008).
  8. Filippi, M. D. Neutrophil transendothelial migration: updates and new perspectives. Blood. 133 (20), 2149-2158 (2019).
  9. Kouspou, M. M., Price, J. T. Analysis of cellular migration using a two-chamber methodology. Methods in Molecular Biology. 787, 303-317 (2011).
  10. Muinonen-Martin, A. J., Veltman, D. M., Kalna, G., Insall, R. H. An improved chamber for direct visualisation of chemotaxis. PLoS One. 5 (12), e15309 (2010).
  11. Walheim, C. C., Zanin, J. P., de Bellard, M. E. Analysis of trunk neural crest cell migration using a modified Zigmond chamber assay. Journal of Visualized Experiments. (59), e3330 (2012).
  12. Liew, P. X., Kubes, P. The Neutrophil's Role During Health and Disease. Physiological Reviews. 99 (2), 1223-1248 (2019).
  13. Ley, K., et al. Neutrophils: New insights and open questions. Science Immunology. 3 (30), eaat4579 (2018).
  14. Tillack, K., Breiden, P., Martin, R., Sospedra, M. T lymphocyte priming by neutrophil extracellular traps links innate and adaptive immune responses. Journal of Immunology. 188 (7), 3150-3159 (2012).
  15. Puga, I., et al. B cell-helper neutrophils stimulate the diversification and production of immunoglobulin in the marginal zone of the spleen. Nature Immunology. 13 (2), 170-180 (2011).
  16. Strzepa, A., Pritchard, K. A., Dittel, B. N. Myeloperoxidase: A new player in autoimmunity. Cellular Immunology. 317, 1-8 (2017).
  17. Momohara, S., Kashiwazaki, S., Inoue, K., Saito, S., Nakagawa, T. Elastase from polymorphonuclear leukocyte in articular cartilage and synovial fluids of patients with rheumatoid arthritis. Clinical Rheumatology. 16 (2), 133-140 (1997).
  18. Swamydas, M., Luo, Y., Dorf, M. E., Lionakis, M. S. Isolation of Mouse Neutrophils. Current Protocols in Immunology. 110 (1), 3.20.1-3.20.15 (2015).
  19. Luo, Y., Dorf, M. E. Isolation of Mouse Neutrophils. Current Protocols in Immunology. 22 (1), 3.20.1-3.20.6 (1997).
  20. Carlson, M., et al. Human neutrophil lipocalin is a unique marker of neutrophil inflammation in ulcerative colitis and proctitis. Gut. 50 (4), 501-506 (2002).
  21. Nathan, C. Neutrophils and immunity: challenges and opportunities. Nature Reviews Immunology. 6 (3), 173-182 (2006).
  22. Zhang, S., et al. Tanshinone IIA ameliorates chronic arthritis in mice by modulating neutrophil activities. Clinical and Experimental Immunology. 190 (1), 29-39 (2017).
  23. Forster, R., Sozzani, S. Emerging aspects of leukocyte migration. European Journal of Immunology. 43 (6), 1404-1406 (2013).
  24. Hu, L., et al. Neutrophil-Mediated Delivery of Dexamethasone Palmitate-Loaded Liposomes Decorated with a Sialic Acid Conjugate for Rheumatoid Arthritis Treatment. Pharmaceutical Research. 36 (7), 97 (2019).

Tags

Migration Infiltration Neutrophils Mice In Vivo In Vitro Arthritis Model Researchers Methodological Details Inflammation Sites Air Pouch Assay Adjuvant-induced Arthritis Arthritis Therapy Model Group Treatment Group Inflammatory Disease Inflammatory Bowel Disease Anesthetization 0.22 Micron Filter Sterilized Air Anesthetized Mouse 26 Gauge Needle Breathing Unit Well Set Cage Monitor
Detecting Migration and Infiltration of Neutrophils in Mice
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Lu, Q., Yuan, K., Li, X., Jiang, H., More

Lu, Q., Yuan, K., Li, X., Jiang, H., Huo, G., Jia, W., Huang, G., Xu, A. Detecting Migration and Infiltration of Neutrophils in Mice. J. Vis. Exp. (156), e60543, doi:10.3791/60543 (2020).

Less
Copy Citation Download Citation Reprints and Permissions
View Video

Get cutting-edge science videos from JoVE sent straight to your inbox every month.

Waiting X
Simple Hit Counter