Summary

Immunocompetent Alveolus-on-Chip Model for Studying Alveolar Mucosal Immune Responses

Published: May 31, 2024
doi:

Summary

Lung-on-chip models surpass traditional 2D cultures by mimicking the air-liquid interface and endothelial cell perfusion, simulating blood flow and nutrient exchange crucial for lung physiology studies. This enhances lung research relevance, offering a dynamic, physiologically accurate environment to advance the understanding and treatment of respiratory infections.

Abstract

We introduce an advanced immunocompetent lung-on-chip model designed to replicate the human alveolar structure and function. This innovative model employs a microfluidic-perfused biochip that supports an air-liquid interface mimicking the environment in the human alveoli. Tissue engineering is used to integrate key cellular components, including endothelial cells, macrophages, and epithelial cells, to create a representative tissue model of the alveolus. The model facilitates in-depth examinations of the mucosal immune responses to various pathogens, including viruses, bacteria, and fungi, thereby advancing our understanding of lung immunity. The primary goal of this protocol is to provide details for establishing this alveolus-on-chip model as a robust in vitro platform for infection studies, enabling researchers to closely observe and analyze the complex interactions between pathogens and the host’s immune system within the pulmonary environment. This is achieved through the application of microfluidic-based techniques to simulate key physiological conditions of the human alveoli, including blood flow and biomechanical stimulation of endothelial cells, alongside maintaining an air-liquid interface crucial for the realistic exposure of epithelial cells to air. The model system is compatible with a range of standardized assays, such as immunofluorescence staining, cytokine profiling, and colony-forming unit (CFU)/plaque analysis, allowing for comprehensive insights into immune dynamics during infection. The Alveolus-on-chip is composed of essential cell types, including human distal lung epithelial cells (H441) and human umbilical vein endothelial cells (HUVECs) separated by porous polyethylene terephthalate (PET) membranes, with primary monocyte-derived macrophages strategically positioned between the epithelial and endothelial layers. The tissue model enhances the ability to dissect and analyze the nuanced factors involved in pulmonary immune responses in vitro. As a valuable tool, it should contribute to the advancement of lung research, providing a more accurate and dynamic in vitro model for studying the pathogenesis of respiratory infections and testing potential therapeutic interventions.

Introduction

The human lung has a remarkable role in respiration and immune defense, with complex interactions between the immune responses of the alveolar mucosa1. The ability of the alveoli to create an efficient immune response is vital for preventing lung infections and securing pulmonary health. Since the lungs are constantly exposed to a wide range of potential risks, including bacteria, viruses, fungi, allergies, and particulate matter, understanding the complexities of alveolar mucosal immune responses is critical for discovering the mechanisms behind respiratory infections, inflammatory disorders and treating pulmonary diseases1.

To study infection and inflammation-related processes of the respiratory tract in vitro, models that could faithfully mimic the alveolar milieu and the immune responses are required. 2D cell culture and animal modules have been used for decades as essential tools for biomedical research on lung immune response. However, they often have limitations in their translational potential to human situations. Lung-on-chip models can contribute to filling the gap between traditional in vitro and in vivo models and provide a novel approach to studying human-specific immune responses2,3. Lung-on-chip models can mimic the air-liquid interface, which is required for lung cells to recapitulate physiological conditions of the respiratory tract and to develop a more accurate and robust tissue model. This culture technique enables a precise examination of cell differentiation, functioning, and responses to drugs or disease-related stimuli in vitro2.

In this study, we present a microfluidic-based model of the human alveolus as an effective tool to recapitulate the human alveolar milieu by applying perfusion to mimic blood flow and biomechanical stimulation of endothelial cells and incorporating an air-liquid interface with epithelial cells exposed towards an air phase4. We have developed a microfluidic perfused alveolus-on-chip that mimics the physical structure and biological interactions of the human alveolus, with a particular focus on the air-liquid interface. This interface plays a crucial role in the differentiation of respiratory epithelial cells, which is essential for accurately modeling the pulmonary environment. The model uses human distal lung epithelial cells (H441) and human umbilical vein endothelial cells (HUVECs), separated by porous polyethylene terephthalate (PET) membranes, with primary monocyte-derived macrophages positioned between the cell layers. This setup replicates the intricate cellular arrangement of the alveolus and is critical for accurately simulating the air-liquid interface, which is a significant factor in the physiological function of lung tissue.

The rationale behind the model development extends to integrating both circulating and tissue-resident immune cells. This approach is designed to accurately mimic the inflammatory host response to human respiratory infections, providing a dynamic environment to study pathogen-host interactions. The presence of macrophages allows for the examination of immediate immune responses and their interaction with pathogens, reflecting the first line of defense against respiratory infections. Furthermore, the design of the biochip platform facilitates convenient and precise manipulation of both biophysical and biochemical cues, which is crucial for replicating alveolus function in vitro. This flexibility is instrumental in dissecting the contributing factors to human infections, enabling researchers to adjust conditions to reflect various disease states or to test potential therapeutic interventions. The compatibility of the platform with multiple readout technologies, including advanced microscopy, microbiological analyses, and biochemical effluent analysis, enhances its utility. These capabilities allow for a comprehensive assessment of the tissue response to infections, including evaluating cellular behavior, pathogen proliferation, and the effectiveness of immune responses.

We present a detailed protocol and techniques to create and utilize a human alveolus-on-chip model focused on replicating the air-liquid interface and integrating immune cells to study human infections in vitro.

Protocol

HUVEC cells are isolated from umbilical cords and used up to passage 4. Primary monocytes are isolated from healthy donors from whole blood. The study was approved by the ethics committee of the Jena University Hospital, Jena, Germany (3939-12/13). According to the Declaration of Helsinki, all individuals donating cells for the study gave their informed consent. 1. Day 1: Preparation of the biochip The biochips are available in different sizes and models. For exper…

Representative Results

An examination of morphological alterations and the expression of marker proteins could be performed using immunofluorescence staining. After co-culturing for 14 days, the vascular and epithelial sides are analyzed for expression of respective cell markers. This method is useful for studying the interactions and integrity of vascular and epithelial components, which is essential for disease modeling as a functional biological readout related to infection. Immunofluorescence staining could be supported by quantifying anal…

Discussion

The alveolus-on-chip model represents a multilayered tissue model of the human alveolus, integrating essential cell types of the lower respiratory tract, including lung epithelial cells, endothelial cells, and macrophages, cultured in an organotypic arrangement at an ALI with medium perfusion of the endothelial lining. Cells of different layers express specific cell marker proteins such as E-cadherin, a calcium-dependent adhesion molecule of lung epithelial cells, which is central in establishing intercellular epithelial…

Divulgations

The authors have nothing to disclose.

Acknowledgements

H.K. and A.S.M. acknowledge funding from the Leibniz Science-Campus InfectoOptics Jena, financed by the funding line Strategic Networking of the Leibniz Association. M.A. and A.S.M. were supported by the IGF project IMPROVE funded by the Federal Ministry for Economic Affairs and Energy on the basis of a resolution of the German Bundestag. A.S.M further acknowledges financial support by the Cluster of Excellence Balance of the Microverse under Germany's Excellence Strategy – EXC 2051 – Project-ID 690 390713860.

Materials

Consumables
Cellcounting chamber slides (Countess) Invitrogen C10283
Cell culture Multiwell Plates, 24 Well, steril Greiner Bio-One 662 160
Cell culture Multiwell Plates, 6 Well, steril Greiner Bio-One 657 160
Coverslips (24x40mm; #1.5) Menzel-Gläser 15747592
Eco wipes Dr. Schuhmacher 00-915-REW10003-01
Eppies 2.0 Sarstedt 72.691
Eppis 0.5 Sarstedt 72.699
Eppis 1.5 Sarstedt 72.690.001
Falcons 15mL Greiner Bio-One 188 271-TRI
Falcons 50mL Greiner Bio-One 227 261-TRI
Gauze swab Noba PZN 2417767
Gloves Nitril 3000 Meditrade 1280
Microscope slides Menzel-Gläser AAAA000001##12E
Multiwell Plates 24 Well, sterile Greiner Bio-One 662 160
Pasteur pipettes (glass) 150mm Assistent 40567001
Pasteur pipettes (glass) 230mm Assistent 40567002
Round-bottom tubes (PS, 5mL) Falcon 352052
Safety-Multifly-Set, 20G, 200mm Sarstedt 85.1637.235
Scalpels Dahlhausen 11.000.00.715
Serological pipettes 10mL Greiner Bio-One 607 160-TRI
Serological pipettes 25mL Greiner Bio-One 760 160-TRI
Serological pipettes 2mL Greiner Bio-One 710 160-TRI
Serological pipettes 50mL Greiner Bio-One 768 160-TRI
Serological pipettes 5mL Greiner Bio-One 606 160-TRI
S-Monovette, 7,5ml Z-Gel Sarstedt 1.1602
S-Monovette, 9,0ml K3E Sarstedt 02.1066.001
Softasept N Braun 3887138
T25 flask Greiner Bio-One 690 960
Tips sterile 10µL Greiner Bio-One 771 261
Tips sterile 1250µL Greiner Bio-One 750 261
Tips sterile 300µL Greiner Bio-One 738 261
Tips unsterile 10µL Greiner Bio-One 765 290
Tips unsterile 1000µL Greiner Bio-One 739 291
Tips unsterile 200µL Greiner Bio-One 686 290
Tweezers (Präzisionspinzette DUMONT abgewinkelt Inox08, 5/45, 0,06 mm) Roth K343.1
Chemicals
Descosept AF Dr. Schuhmacher N-20338
Ethanol 96% Nordbrand-Nordhausen 410
Fluorescein isothiocyanate (FITC)-dextran (3-5kDa) Sigma Aldrich FD4-100MG
Fluorescent Mounting Medium Dako S3023
Methanol VWR 20847.295
Saponin Fluka 47036
Tergazyme Alconox 1304-1
Cell culture
Collagen IV Sigma-Aldrich C5533-5MG
Dexametason Sigma-Aldrich D4902
DPBS (-/-) Lonza BE17-516F
DPBS (+/+) Lonza BE17-513F
EDTA solution Sigma-Aldrich E788S
Endothelial Cell Growth Medium Promocell C-22020
Endothelial Cell Growth Medium supplement mix Promocell C-39225
Fetal bovine Serum Sigma-Aldrich E2129-10g
H441 ATCC
Human recombinant GM-CSF Peprotech 300-30
Lidocain Sigma-Aldrich L5647-15G
Penicillin-Streptomycin (10,000 U/mL) Gibco 15140-122 /-163
RPMI Gibco 72400047
Trypane blue stain 0.4% Invitrogen T10282
Trypsin Gibco 15090-046
Primary antibodies
Cadherin-5 / VE-Cadherin (goat) BD 610252
CD68 (rabbit) CellSignaling 76437
E-Cadherin (goat) R&D AF748
SP-A (mouse) Abcam ab51891
Secondary antibodies
AF488 (donkey anti mouse) Invitrogen R37114
AF647 (donkey anti mouse) invitrogen A31571
AF647 (donkey anti rabbit) Invitrogen A31573
Cy3 (donkey anti goat) jackson research 705-165-147
DAPI (4',6-Diamidino-2-Phenylindole, Dilactate) Invitrogen D3571
Microfluidic
Chip Dynamic 42 BC002
Male Luer Lock (small) ChipShop 09-0503-0270-09
Male mini luer plugs, row of four,PP, green Microfluidic chipshop 09-0558-0336-11
Male mini luer plugs, row of four,PP, opaque Microfluidic chipshop 09-0556-0336-09
Male mini luer plugs, row of four,PP, red Microfluidic chipshop 09-0557-0336-10
Plugs Cole Parmer GZ-45555-56
Reservoir 4.5mL ChipShop 16-0613-0233-09
Tubing Dynamic 42 ST001
Equipment
Autoclave Tuttnauer 5075 ELV
Centrifuge Eppendorf 5424
CO2 Incubator Heracell 150i
Countess automated cell counter Invitrogen C10227
Flowcytometer BD FACS Canto II
Freezer (-20 °C) Liebherr LCexv 4010
Freezer (-80 °C) Heraeus Herafreeze HFU 686
Fridge Liebherr LCexv 4010
Heraeus Multifuge Thermo Scientific X3R
Microscope Leica DM IL LED
Orbital shaker Heidolph Reax2000
Peristaltic pump REGLO Digital MS-4/12 ISM597D
Pipettes 10µL Eppendorf Research plus 3123000020
Pipettes 100µL Eppendorf Research plus 3123000047
Pipettes 1000µL Eppendorf Research plus 3123000063
Pipettes 2.5µL Eppendorf Research plus 3123000012
Pipettes 20µL Eppendorf Research plus 3123000039
Pipettes 200µL Eppendorf Research plus 3123000055
Scale Sartorius 6101
Scale Sartorius TE1245
Sterile bench Kojair Biowizard SL-130
Waterbath Julabo SW-20C
Fluorescence Microscope Setup
Apotome.2 Zeiss
Illumination device Zeiss HXP 120 C
Microscope Zeiss Axio Observer 5
Optical Sectioning Zeiss ApoTome
Power Supply Microscope Zeiss Eplax Vp232
Software
ZEN Blue Edition Zeiss

References

  1. Mettelman, R. C., Allen, E. K., Thomas, P. G. Mucosal immune responses to infection and vaccination in the respiratory tract. Immunity. 55 (5), 749-780 (2022).
  2. Artzy-Schnirman, A., et al. Advanced in vitro lung-on-chip platforms for inhalation assays: From prospect to pipeline. Eur J Pharma Biopharma. 144, 11-17 (2019).
  3. Huh, D., et al. Reconstituting organ-level lung functions on a chip. Science. 328 (5986), 1662-1668 (2010).
  4. Benam, K. H., et al. Small airway-on-a-chip enables analysis of human lung inflammation and drug responses in vitro. Nat Meth. 13 (2), 151-157 (2016).
  5. Ronaldson-Bouchard, K., et al. A multi-organ chip with matured tissue niches linked by vascular flow. Nat Biomed Eng. 6 (4), 351 (2022).
  6. Deinhardt-Emmer, S., et al. Co-infection with staphylococcus aureus after primary influenza virus infection leads to damage of the endothelium in a human alveolus-on-a-chip model. Biofabrication. 12 (2), 025012 (2020).
  7. Schicke, E., et al. Staphylococcus aureus lung infection results in down-regulation of surfactant protein-a mainly caused by pro-inflammatory macrophages. Microorganisms. 8 (4), 577 (2020).
  8. King, S. D., Chen, S. Y. Recent progress on surfactant protein a: Cellular function in lung and kidney disease development. Am J Physiol Cell Physiol. 319 (2), C316-C320 (2020).
  9. Hoang, T. N. M., et al. Invasive aspergillosis-on-chip: A quantitative treatment study of human aspergillus fumigatus infection. Biomaterials. 283, 121420 (2022).
  10. Yuksel, H., Ocalan, M., Yilmaz, O. E-cadherin: An important functional molecule at respiratory barrier between defence and dysfunction. Front Physiol. 12, 720227 (2021).
  11. Van Roy, F., Berx, G. The cell-cell adhesion molecule e-cadherin. Cell Mol Life Sci. 65 (23), 3756-3788 (2008).
This article has been published
Video Coming Soon
Keep me updated:

.

Citer Cet Article
Koceva, H., Amiratashani, M., Rennert, K., Mosig, A. S. Immunocompetent Alveolus-on-Chip Model for Studying Alveolar Mucosal Immune Responses. J. Vis. Exp. (207), e66602, doi:10.3791/66602 (2024).

View Video