1
|
Floor E, Su J, Chatterjee M, Kuipers ES, IJssennagger N, Heidari F, Giordano L, Wubbolts RW, Mihăilă SM, Stapels DAC, Vercoulen Y, Strijbis K. Development of a Caco-2-based intestinal mucosal model to study intestinal barrier properties and bacteria-mucus interactions. Gut Microbes 2025; 17:2434685. [PMID: 39714032 DOI: 10.1080/19490976.2024.2434685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 11/15/2024] [Accepted: 11/21/2024] [Indexed: 12/24/2024] Open
Abstract
The intestinal mucosal barrier is a dynamic system that allows nutrient uptake, stimulates healthy microbe-host interactions, and prevents invasion by pathogens. The mucosa consists of epithelial cells connected by cellular junctions that regulate the passage of nutrients covered by a mucus layer that plays an important role in host-microbiome interactions. Mimicking the intestinal mucosa for in vitro assays, particularly the generation of a mucus layer, has proven to be challenging. The intestinal cell-line Caco-2 is widely used in academic and industrial laboratories due to its capacity to polarize, form an apical brush border, and reproducibly grow into confluent cell layers in different culture systems. However, under normal culture conditions, Caco-2 cultures lack a mucus layer. Here, we demonstrate for the first time that Caco-2 cultures can form a robust mucus layer when cultured under air-liquid interface (ALI) conditions on Transwell inserts with addition of vasointestinal peptide (VIP) in the basolateral compartment. We demonstrate that unique gene clusters are regulated in response to ALI and VIP single stimuli, but the ALI-VIP combination treatment resulted in a significant upregulation of multiple mucin genes and proteins, including secreted MUC2 and transmembrane mucins MUC13 and MUC17. Expression of tight junction proteins was significantly altered in the ALI-VIP condition, leading to increased permeability to small molecules. Commensal Lactiplantibacillus plantarum bacteria closely associated with the Caco-2 mucus layer and differentially colonized the surface of the ALI cultures. Pathogenic Salmonella enterica were capable of invading beyond the mucus layer and brush border. In conclusion, Caco-2 ALI-VIP cultures provide an accessible and straightforward way to culture an in vitro intestinal mucosal model with improved biomimetic features. This novel in vitro intestinal model can facilitate studies into mucus and epithelial barrier functions and in-depth molecular characterization of pathogenic and commensal microbe-mucus interactions.
Collapse
Affiliation(s)
- Evelien Floor
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jinyi Su
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Maitrayee Chatterjee
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- The TIM Company, Delft, the Netherlands
| | - Elise S Kuipers
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Noortje IJssennagger
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Danone Research and Innovation Center, Utrecht, The Netherlands
| | - Faranak Heidari
- Div. Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Laura Giordano
- Div. Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Richard W Wubbolts
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Silvia M Mihăilă
- Div. Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Daphne A C Stapels
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Yvonne Vercoulen
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Karin Strijbis
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
2
|
Park CH, Park JH, Suh YJ. Perspective of 3D culture in medicine: transforming disease research and therapeutic applications. Front Bioeng Biotechnol 2024; 12:1491669. [PMID: 39749112 PMCID: PMC11693738 DOI: 10.3389/fbioe.2024.1491669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
3D cell culture is gaining momentum in medicine due to its ability to mimic real tissues (in vivo) and provide more accurate biological data compared to traditional methods. This review explores the current state of 3D cell culture in medicine and discusses future directions, including the need for standardization and simpler protocols to facilitate wider use in research. Purpose 3D cell culture develops life sciences by mimicking the natural cellular environment. Cells in 3D cultures grow in three dimensions and interact with a matrix, fostering realistic cell behavior and interactions. This enhanced model offers significant advantages for diverse research areas. Methods By mimicking the cellular organization and functionalities found in human tissues, 3D cultures provide superior platforms for studying complex diseases like cancer and neurodegenerative disorders. This enables researchers to gain deeper insights into disease progression and identify promising therapeutic targets with greater accuracy. 3D cultures also play a crucial role in drug discovery by allowing researchers to effectively assess potential drugs' safety and efficacy. Results 3D cell culture's impact goes beyond disease research. It holds promise for tissue engineering. By replicating the natural tissue environment and providing a scaffold for cell growth, 3D cultures pave the way for regenerating damaged tissues, offering hope for treating burns, organ failure, and musculoskeletal injuries. Additionally, 3D cultures contribute to personalized medicine. Researchers can use patient-derived cells to create personalized disease models and identify the most effective treatment for each individual. Conclusion With ongoing advancements in cell imaging techniques, the development of novel biocompatible scaffolds and bioreactor systems, and a deeper understanding of cellular behavior within 3D environments, 3D cell culture technology stands poised to revolutionize various aspects of healthcare and scientific discovery.
Collapse
Affiliation(s)
- Chan Hum Park
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon, Republic of Korea
- Departments of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, School of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jung Ho Park
- Department of Breast and Endocrine Surgery, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| | - Yong Joon Suh
- Department of Breast and Endocrine Surgery, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| |
Collapse
|
3
|
Kim R, Allbritton NL. A Microphysiological System with an Anaerobic Air-Liquid Interface and Functional Mucus Layer for Coculture of Intestinal Bacteria and Primary Human Colonic Epithelium. ADVANCED MATERIALS INTERFACES 2024; 11:2400093. [PMID: 39386255 PMCID: PMC11460523 DOI: 10.1002/admi.202400093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Indexed: 10/12/2024]
Abstract
Coculture of intestinal bacteria with primary human intestinal epithelium provides a valuable tool for investigating host-colon bacterial interactions and for testing and screening therapeutics. However, most current intestinal model systems lack key physiological features of the in vivo colon, such as both a proper oxygen microenvironment and a mucus layer. In this work, a new in vitro colonic microphysiological system is demonstrated with a cell-derived, functional mucus that closely resembles the in vivo colonic mucosa and apical microenvironment by employing an anaerobic air-liquid interface culture. The human primary colon epithelial cells in this new in vitro system exhibit high cell viability (>98%) with ≈100 μm thick functional mucus layer on average. Successful coculture of model anaerobic gut bacterial strains Lactobacillus rhamnosus GG and Anaerobutyricum hallii without loss in human cell viability demonstrates that this new model can be an invaluable tool for future studies of the impact of commensal and pathogenic bacteria on the colon.
Collapse
Affiliation(s)
- Raehyun Kim
- Department of Biological and Chemical Engineering, Hongik University, Sejong-si 30016, Republic of Korea
| | - Nancy L Allbritton
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
4
|
Chen D, Xu L, Xuan M, Chu Q, Xue C. Unveiling the functional roles of patient-derived tumour organoids in assessing the tumour microenvironment and immunotherapy. Clin Transl Med 2024; 14:e1802. [PMID: 39245957 PMCID: PMC11381553 DOI: 10.1002/ctm2.1802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 09/10/2024] Open
Abstract
Recent studies have established the pivotal roles of patient-derived tumour organoids (PDTOs), innovative three-dimensional (3D) culture systems, in various biological and medical applications. PDTOs, as promising tools, have been established and extensively used for drug screening, prediction of immune response and assessment of immunotherapeutic effectiveness in various cancer types, including glioma, ovarian cancer and so on. The overarching goal is to facilitate the translation of new therapeutic modalities to guide personalised immunotherapy. Notably, there has been a recent surge of interest in the co-culture of PDTOs with immune cells to investigate the dynamic interactions between tumour cells and immune microenvironment. A comprehensive and in-depth investigation is necessary to enhance our understanding of PDTOs as promising testing platforms for cancer immunotherapy. This review mainly focuses on the latest updates on the applications and challenges of PDTO-based methods in anti-cancer immune responses. We strive to provide a comprehensive understanding of the potential and prospects of PDTO-based technologies as next-generation strategies for advancing immunotherapy approaches.
Collapse
Affiliation(s)
- Di Chen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lixia Xu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengjuan Xuan
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qingfei Chu
- Department of State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen Xue
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Franko R, de Almeida Monteiro Melo Ferraz M. Exploring the potential of in vitro extracellular vesicle generation in reproductive biology. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70007. [PMID: 39238549 PMCID: PMC11375532 DOI: 10.1002/jex2.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/11/2024] [Accepted: 08/15/2024] [Indexed: 09/07/2024]
Abstract
The interest in the growing field of extracellular vesicle (EV) research highlights their significance in intercellular signalling and the selective transfer of biological information between donor and recipient cells. EV studies have provided valuable insights into intercellular communication mechanisms, signal identification and their involvement in disease states, offering potential avenues for manipulating pathological conditions, detecting biomarkers and developing drug-delivery systems. While our understanding of EV functions in reproductive tissues has significantly progressed, exploring their potential as biomarkers for infertility, therapeutic interventions and enhancements in assisted reproductive technologies remains to be investigated. This knowledge gap stems partly from the difficulties associated with large-scale EV production relevant to clinical applications. Most existing studies on EV production rely on conventional 2D cell culture systems, characterized by suboptimal EV yields and a failure to replicate in vivo conditions. This results in the generation of EVs that differ from their in vivo counterparts. Hence, this review firstly delves into the importance of EVs in reproduction to then expand on current techniques for in vitro EV production, specifically examining diverse methods of culture and the potential of bioengineering technologies to establish innovative systems for enhanced EV production.
Collapse
Affiliation(s)
- Roksan Franko
- Clinic of Ruminants, Faculty of Veterinary Medicine Ludwig-Maximilians-Universität München Oberschleißheim Germany
- Gene Center Ludwig-Maximilians-Universität München Munich Germany
| | - Marcia de Almeida Monteiro Melo Ferraz
- Clinic of Ruminants, Faculty of Veterinary Medicine Ludwig-Maximilians-Universität München Oberschleißheim Germany
- Gene Center Ludwig-Maximilians-Universität München Munich Germany
| |
Collapse
|
6
|
Lagowala DA, Wally A, Wilmsen K, Kim B, Yeung-Luk B, Choi J, Swaby C, Luk M, Feller L, Ghosh B, Niedrkofler A, Tieng E, Sherman E, Chen D, Upadya N, Zhang R, Kim DH, Sidhaye V. Microphysiological Models of Lung Epithelium-Alveolar Macrophage Co-Cultures to Study Chronic Lung Disease. Adv Biol (Weinh) 2024; 8:e2300165. [PMID: 37840439 PMCID: PMC11995713 DOI: 10.1002/adbi.202300165] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/06/2023] [Indexed: 10/17/2023]
Abstract
The interactions between immune cells and epithelial cells influence the progression of many respiratory diseases, such as chronic obstructive pulmonary disease (COPD). In vitro models allow for the examination of cells in controlled environments. However, these models lack the complex 3D architecture and vast multicellular interactions between the lung resident cells and infiltrating immune cells that can mediate cellular response to insults. In this study, three complementary microphysiological systems are presented to delineate the effects of cigarette smoke and respiratory disease on the lung epithelium. First, the Transwell system allows the co-culture of pulmonary immune and epithelial cells to evaluate cellular and monolayer phenotypic changes in response to cigarette smoke exposure. Next, the human and mouse precision-cut lung slices system provides a physiologically relevant model to study the effects of chronic insults like cigarette smoke with the dissection of specific interaction of immune cell subtypes within the structurally complex tissue environment. Finally, the lung-on-a-chip model provides an adaptable system for live imaging of polarized epithelial tissues that mimic the in vivo environment of the airways. Using a combination of these models, a complementary approach is provided to better address the intricate mechanisms of lung disease.
Collapse
Affiliation(s)
- Dave A. Lagowala
- Department of Environmental Health and Engineering, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Arabelis Wally
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kai Wilmsen
- Department of Environmental Health and Engineering, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Byunggik Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Bonnie Yeung-Luk
- Department of Environmental Health and Engineering, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Jeongseob Choi
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Carter Swaby
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Matthew Luk
- Department of Environmental Health and Engineering, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Laine Feller
- Department of Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Baishakhi Ghosh
- Department of Environmental Health and Engineering, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Austin Niedrkofler
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ethan Tieng
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ethan Sherman
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daniel Chen
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nisha Upadya
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rachel Zhang
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Venkataramana Sidhaye
- Department of Environmental Health and Engineering, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
7
|
Wang P, Jin L, Zhang M, Wu Y, Duan Z, Guo Y, Wang C, Guo Y, Chen W, Liao Z, Wang Y, Lai R, Lee LP, Qin J. Blood-brain barrier injury and neuroinflammation induced by SARS-CoV-2 in a lung-brain microphysiological system. Nat Biomed Eng 2024; 8:1053-1068. [PMID: 37349391 DOI: 10.1038/s41551-023-01054-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 05/11/2023] [Indexed: 06/24/2023]
Abstract
In some patients, COVID-19 can trigger neurological symptoms with unclear pathogenesis. Here we describe a microphysiological system integrating alveolus and blood-brain barrier (BBB) tissue chips that recapitulates neuropathogenesis associated with infection by SARS-CoV-2. Direct exposure of the BBB chip to SARS-CoV-2 caused mild changes to the BBB, and infusion of medium from the infected alveolus chip led to more severe injuries on the BBB chip, including endothelial dysfunction, pericyte detachment and neuroinflammation. Transcriptomic analyses indicated downregulated expression of the actin cytoskeleton in brain endothelium and upregulated expression of inflammatory genes in glial cells. We also observed early cerebral microvascular damage following lung infection with a low viral load in the brains of transgenic mice expressing human angiotensin-converting enzyme 2. Our findings suggest that systemic inflammation is probably contributing to neuropathogenesis following SARS-CoV-2 infection, and that direct viral neural invasion might not be a prerequisite for this neuropathogenesis. Lung-brain microphysiological systems should aid the further understanding of the systemic effects and neurological complications of viral infection.
Collapse
Affiliation(s)
- Peng Wang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- University of Science and Technology of China, Hefei, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, China
| | - Lin Jin
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences-Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Min Zhang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yunsong Wu
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zilei Duan
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences-Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yaqiong Guo
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Chaoming Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences-Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yingqi Guo
- Core Technology Facility of Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Wenwen Chen
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Zhiyi Liao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences-Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yaqing Wang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences-Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
| | - Luke P Lee
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA, USA.
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, Korea.
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.
- University of Science and Technology of China, Hefei, China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
8
|
Lee RJ, Adappa ND, Palmer JN. Akt activator SC79 stimulates antibacterial nitric oxide generation in human nasal epithelial cells in vitro. Int Forum Allergy Rhinol 2024; 14:1147-1162. [PMID: 38197521 PMCID: PMC11219270 DOI: 10.1002/alr.23318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/05/2023] [Accepted: 12/26/2023] [Indexed: 01/11/2024]
Abstract
BACKGROUND The role of Akt in nasal immunity is unstudied. Akt phosphorylates and activates endothelial nitric oxide synthase (eNOS) expressed in epithelial ciliated cells. Nitric oxide (NO) production by ciliated cells can have antibacterial and antiviral effects. Increasing nasal NO may be a useful antipathogen strategy in chronic rhinosinusitis (CRS). We previously showed that small-molecule Akt activator SC79 induces nasal cell NO production and suppresses IL-8 via the transcription factor Nrf-2. We hypothesized that SC79 NO production may additionally have antibacterial effects. METHODS NO production was measured using fluorescent dye DAF-FM. We tested effects of SC79 during co-culture of Pseudomonas aeruginosa with primary nasal epithelial cells, using CFU counting and live-dead staining to quantify bacterial killing. Pharmacology determined the mechanism of SC79-induced NO production and tested dependence on Akt. RESULTS SC79 induced dose-dependent, Akt-dependent NO production in nasal epithelial cells. The NO production required eNOS and Akt. The NO released into the airway surface liquid killed P. aeruginosa. No toxicity (LDH release) or inflammatory effects (IL8 transcription) were observed over 24 h. CONCLUSIONS Together, these data suggest multiple immune pathways are stimulated by SC79, with antipathogen effects. This in vitro pilot study suggests that a small-molecule Akt activator may have clinical utility in CRS or respiratory other infection settings, warranting future in vivo studies.
Collapse
Affiliation(s)
- Robert J. Lee
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine
- Department of Physiology, University of Pennsylvania Perelman School of Medicine
| | - Nithin D. Adappa
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine
| | - James N. Palmer
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine
| |
Collapse
|
9
|
Wolters RM, Ferguson JA, Nuñez IA, Chen EE, Sornberger T, Myers L, Oeverdieck S, Raghavan SSR, Kona C, Handal LS, Esilu TE, Davidson E, Doranz BJ, Engdahl TB, Kose N, Williamson LE, Creech CB, Gibson-Corley KN, Ward AB, Crowe JE. Isolation of human antibodies against influenza B neuraminidase and mechanisms of protection at the airway interface. Immunity 2024; 57:1413-1427.e9. [PMID: 38823390 PMCID: PMC11440431 DOI: 10.1016/j.immuni.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/16/2024] [Accepted: 05/06/2024] [Indexed: 06/03/2024]
Abstract
Influenza B viruses (IBVs) comprise a substantial portion of the circulating seasonal human influenza viruses. Here, we describe the isolation of human monoclonal antibodies (mAbs) that recognized the IBV neuraminidase (NA) glycoprotein from an individual following seasonal vaccination. Competition-binding experiments suggested the antibodies recognized two major antigenic sites. One group, which included mAb FluB-393, broadly inhibited IBV NA sialidase activity, protected prophylactically in vivo, and bound to the lateral corner of NA. The second group contained an active site mAb, FluB-400, that broadly inhibited IBV NA sialidase activity and virus replication in vitro in primary human respiratory epithelial cell cultures and protected against IBV in vivo when administered systemically or intranasally. Overall, the findings described here shape our mechanistic understanding of the human immune response to the IBV NA glycoprotein through the demonstration of two mAb delivery routes for protection against IBV and the identification of potential IBV therapeutic candidates.
Collapse
Affiliation(s)
- Rachael M Wolters
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James A Ferguson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ivette A Nuñez
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Elaine E Chen
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ty Sornberger
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Luke Myers
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Svearike Oeverdieck
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sai Sundar Rajan Raghavan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Chandrahaas Kona
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Laura S Handal
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | - Taylor B Engdahl
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nurgun Kose
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lauren E Williamson
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - C Buddy Creech
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Katherine N Gibson-Corley
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - James E Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
10
|
Tóth G, Golubova A, Falk A, Lind SB, Nicholas M, Lanekoff I. Interleukin-13 Treatment of Living Lung Tissue Model Alters the Metabolome and Proteome-A Nano-DESI MS Metabolomics and Shotgun Proteomics Study. Int J Mol Sci 2024; 25:5034. [PMID: 38732251 PMCID: PMC11084154 DOI: 10.3390/ijms25095034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/04/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Asthma is a chronic respiratory disease with one of the largest numbers of cases in the world; thus, constant investigation and technical development are needed to unravel the underlying biochemical mechanisms. In this study, we aimed to develop a nano-DESI MS method for the in vivo characterization of the cellular metabolome. Using air-liquid interface (ALI) cell layers, we studied the role of Interleukin-13 (IL-13) on differentiated lung epithelial cells acting as a lung tissue model. We demonstrate the feasibility of nano-DESI MS for the in vivo monitoring of basal-apical molecular transport, and the subsequent endogenous metabolic response, for the first time. Conserving the integrity of the ALI lung-cell layer enabled us to perform temporally resolved metabolomic characterization followed by "bottom-up" proteomics on the same population of cells. Metabolic remodeling was observed upon histamine and corticosteroid treatment of the IL-13-exposed lung cell monolayers, in correlation with alterations in the proteomic profile. This proof of principle study demonstrates the utility of in vivo nano-DESI MS for characterizing ALI tissue layers, and the new markers identified in our study provide a good starting point for future, larger-scale studies.
Collapse
Affiliation(s)
- Gábor Tóth
- Department of Chemistry—BMC, Uppsala University, 75237 Uppsala, Sweden
| | | | - Alexander Falk
- Department of Chemistry—BMC, Uppsala University, 75237 Uppsala, Sweden
| | | | | | - Ingela Lanekoff
- Department of Chemistry—BMC, Uppsala University, 75237 Uppsala, Sweden
| |
Collapse
|
11
|
Bannier-Hélaouët M, Korving J, Ma Z, Begthel H, Giladi A, Lamers MM, van de Wetering WJ, Yawata N, Yawata M, LaPointe VLS, Dickman MM, Kalmann R, Imhoff SM, van Es JH, López-Iglesias C, Peters PJ, Haagmans BL, Wu W, Clevers H. Human conjunctiva organoids to study ocular surface homeostasis and disease. Cell Stem Cell 2024; 31:227-243.e12. [PMID: 38215738 DOI: 10.1016/j.stem.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/14/2023] [Accepted: 12/11/2023] [Indexed: 01/14/2024]
Abstract
The conjunctival epithelium covering the eye contains two main cell types: mucus-producing goblet cells and water-secreting keratinocytes, which present mucins on their apical surface. Here, we describe long-term expanding organoids and air-liquid interface representing mouse and human conjunctiva. A single-cell RNA expression atlas of primary and cultured human conjunctiva reveals that keratinocytes express multiple antimicrobial peptides and identifies conjunctival tuft cells. IL-4/-13 exposure increases goblet and tuft cell differentiation and drastically modifies the conjunctiva secretome. Human NGFR+ basal cells are identified as bipotent conjunctiva stem cells. Conjunctival cultures can be infected by herpes simplex virus 1 (HSV1), human adenovirus 8 (hAdV8), and SARS-CoV-2. HSV1 infection was reversed by acyclovir addition, whereas hAdV8 infection, which lacks an approved drug therapy, was inhibited by cidofovir. We document transcriptional programs induced by HSV1 and hAdV8. Finally, conjunctival organoids can be transplanted. Together, human conjunctiva organoid cultures enable the study of conjunctival (patho)-physiology.
Collapse
Affiliation(s)
- Marie Bannier-Hélaouët
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center, Utrecht, the Netherlands; Oncode Institute, Hubrecht Institute, Utrecht, the Netherlands.
| | - Jeroen Korving
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center, Utrecht, the Netherlands
| | - Ziliang Ma
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), and Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Harry Begthel
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center, Utrecht, the Netherlands; Oncode Institute, Hubrecht Institute, Utrecht, the Netherlands
| | - Amir Giladi
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center, Utrecht, the Netherlands; Oncode Institute, Hubrecht Institute, Utrecht, the Netherlands
| | - Mart M Lamers
- Viroscience Department, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Willine J van de Wetering
- Maastricht Multimodal Molecular Imaging Institute, Maastricht University, Maastricht, the Netherlands
| | - Nobuyo Yawata
- Department of Ocular Pathology and Imaging Science, Kyushu University, Fukuoka, Japan; Singapore Eye Research Institute, Singapore, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Makoto Yawata
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; National University Health System, Singapore, Singapore; Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore; NUSMED Immunology Translational Research Program, National University of Singapore, Singapore, Singapore; Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A(∗)STAR), Singapore, Singapore; International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Vanessa L S LaPointe
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht, the Netherlands
| | - Mor M Dickman
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht, the Netherlands; University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Rachel Kalmann
- Department of Ophthalmology, University Medical Center, Utrecht, the Netherlands
| | - Saskia M Imhoff
- Department of Ophthalmology, University Medical Center, Utrecht, the Netherlands
| | - Johan H van Es
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center, Utrecht, the Netherlands; Oncode Institute, Hubrecht Institute, Utrecht, the Netherlands
| | - Carmen López-Iglesias
- Maastricht Multimodal Molecular Imaging Institute, Maastricht University, Maastricht, the Netherlands
| | - Peter J Peters
- Maastricht Multimodal Molecular Imaging Institute, Maastricht University, Maastricht, the Netherlands
| | - Bart L Haagmans
- Viroscience Department, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Wei Wu
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), and Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center, Utrecht, the Netherlands; Oncode Institute, Hubrecht Institute, Utrecht, the Netherlands.
| |
Collapse
|
12
|
Billiot CE, Novak L, McDaniel MS, Lindgren NR, Swords WE. Pathogenesis of Achromobacter xylosoxidans respiratory infections: colonization, persistence, and transcriptome profiling in synthetic cystic fibrosis sputum medium. Infect Immun 2023; 91:e0041623. [PMID: 37909751 PMCID: PMC10715085 DOI: 10.1128/iai.00416-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 11/03/2023] Open
Abstract
Cystic fibrosis (CF) is a genetic disease affecting epithelial ion transport, resulting in thickened mucus and impaired mucociliary clearance. Persons with CF (pwCF) experience life-long infections of the respiratory mucosa caused by a diverse array of opportunists, which are leading causes of morbidity and mortality. In recent years, there has been increased appreciation for the range and diversity of microbes causing CF-related respiratory infections. The introduction of new therapeutics and improved detection methodology has revealed CF-related opportunists such as Achromobacter xylosoxidans (Ax). Ax is a Gram-negative bacterial species which is widely distributed in environmental sources and has been increasingly observed in sputa and other samples from pwCF, typically in patients in later stages of CF disease. In this study, we characterized CF clinical isolates of Ax and tested colonization and persistence of Ax in respiratory infection using immortalized human CF respiratory epithelial cells and BALB/c mice. Genomic analyses of clinical Ax isolates showed homologs for factors including flagellar synthesis, antibiotic resistance, and toxin secretion systems. Ax isolates adhered to polarized cultures of CFBE41o- human immortalized CF bronchial epithelial cells and caused significant cytotoxicity and depolarization of cell layers. Ax colonized and persisted in mouse lungs for up to 72 h post infection, with inflammatory consequences that include increased neutrophil influx in the lung, lung damage, cytokine production, and mortality. We also identified genes that are differentially expressed in synthetic CF sputum media. Based on these results, we conclude that Ax is an opportunistic pathogen of significance in CF.
Collapse
Affiliation(s)
- Caitlin E. Billiot
- Department of Medicine, Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Gregory Fleming James Center for Cystic Fibrosis Research, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lea Novak
- Department of Pathology, Division of Anatomic Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Melissa S. McDaniel
- Department of Medicine, Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Gregory Fleming James Center for Cystic Fibrosis Research, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Natalie R. Lindgren
- Department of Medicine, Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Gregory Fleming James Center for Cystic Fibrosis Research, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - W. Edward Swords
- Department of Medicine, Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Gregory Fleming James Center for Cystic Fibrosis Research, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
13
|
Chepelova N, Antoshin A, Voloshin S, Usanova A, Efremov Y, Makeeva M, Evlashin S, Stepanov M, Turkina A, Timashev P. Oral Galvanism Side Effects: Comparing Alloy Ions and Galvanic Current Effects on the Mucosa-like Model. J Funct Biomater 2023; 14:564. [PMID: 38132818 PMCID: PMC10744021 DOI: 10.3390/jfb14120564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
The interaction of different dental alloys with the oral environment may cause severe side effects (e.g., burning sensation, inflammatory reactions, carcinogenesis) as a result of oral galvanism. However, the pathogenesis of side effects associated with oral galvanism is still unclear, and the effects of direct current and alloy corrosion ions are considered potentially contributing factors. Therefore, the aim of this study was to systemically compare the damaging effects of (1) galvanism as a synergistic process (direct current + corrosion ions), (2) direct current separately, and (3) corrosion ions separately on an in vitro mucosa-like model based on a cell line of immortalized human keratinocytes (HaCaTs) to reveal the factors playing a pivotal role in dental alloys side effects. For this, we chose and compared the dental alloys with the highest risk of oral galvanism: Ti64-AgPd and NiCr-AgPd. We showed that galvanic current may be the leading damaging factor in the cytotoxic processes associated with galvanic coupling of metallic intraoral appliances in the oral cavity, especially in the short-term period (28 days). However, the contribution of corrosion ions (Ni2+) to the synergistic toxicity was also shown, and quite possibly, in the long term, it could be no less dangerous.
Collapse
Affiliation(s)
- Natalia Chepelova
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 8-2 Trubetskaya St., Moscow 119048, Russia; (N.C.); (S.V.); (A.U.); (Y.E.); (P.T.)
| | - Artem Antoshin
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 8-2 Trubetskaya St., Moscow 119048, Russia; (N.C.); (S.V.); (A.U.); (Y.E.); (P.T.)
| | - Sergei Voloshin
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 8-2 Trubetskaya St., Moscow 119048, Russia; (N.C.); (S.V.); (A.U.); (Y.E.); (P.T.)
| | - Anna Usanova
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 8-2 Trubetskaya St., Moscow 119048, Russia; (N.C.); (S.V.); (A.U.); (Y.E.); (P.T.)
| | - Yuri Efremov
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 8-2 Trubetskaya St., Moscow 119048, Russia; (N.C.); (S.V.); (A.U.); (Y.E.); (P.T.)
| | - Maria Makeeva
- Therapeutic Dentistry Department, Institute for Dentistry, Sechenov First Moscow State Medical University, 8-2 Trubetskaya Str., Moscow 119048, Russia; (M.M.); (A.T.)
- Conservative Dentistry Department, RUDN University, 6 Miklukho-Maklaya Street, Moscow 117198, Russia
| | - Stanislav Evlashin
- Center for Materials Technologies, Skolkovo Institute of Science and Technology, Moscow 121205, Russia;
| | - Mikhail Stepanov
- Department of Dental Surgery, Sechenov First Moscow State Medical University, 8-2 Trubetskaya Str., Moscow 119048, Russia;
| | - Anna Turkina
- Therapeutic Dentistry Department, Institute for Dentistry, Sechenov First Moscow State Medical University, 8-2 Trubetskaya Str., Moscow 119048, Russia; (M.M.); (A.T.)
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 8-2 Trubetskaya St., Moscow 119048, Russia; (N.C.); (S.V.); (A.U.); (Y.E.); (P.T.)
| |
Collapse
|
14
|
Czyz CM, Kunth PW, Gruber F, Kremslehner C, Hammers CM, Hundt JE. Requisite instruments for the establishment of three-dimensional epidermal human skin equivalents-A methods review. Exp Dermatol 2023; 32:1870-1883. [PMID: 37605856 DOI: 10.1111/exd.14911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 07/30/2023] [Accepted: 08/02/2023] [Indexed: 08/23/2023]
Abstract
Human skin equivalents (HSEs) are three-dimensional skin organ culture models raised in vitro. This review gives an overview of common techniques for setting up HSEs. The HSE consists of an artificial dermis and epidermis. 3T3-J2 murine fibroblasts, purchased human fibroblasts or freshly isolated and cultured fibroblasts, together with other components, for example, collagen type I, are used to build the scaffold. Freshly isolated and cultured keratinocytes are seeded on top. It is possible to add other cell types, for example, melanocytes, to the HSE-depending on the research question. After several days and further steps, the 3D skin can be harvested. Additionally, we show possible markers and techniques for evaluation of artificial skin. Furthermore, we provide a comparison of HSEs to human skin organ culture, a model which employs human donor skin. We outline advantages and limitations of both models and discuss future perspectives in using HSEs.
Collapse
Affiliation(s)
- Christianna Marie Czyz
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Paul Werner Kunth
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Florian Gruber
- Christian Doppler Laboratory for Skin Multimodal Analytical Imaging of Aging and Senescence - SKINMAGINE, Medical University of Vienna, Vienna, Austria
| | - Christopher Kremslehner
- Christian Doppler Laboratory for Skin Multimodal Analytical Imaging of Aging and Senescence - SKINMAGINE, Medical University of Vienna, Vienna, Austria
| | - Christoph Matthias Hammers
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
- Department of Dermatology, Venereology and Allergology, University of Kiel, Kiel, Germany
| | | |
Collapse
|
15
|
Tian J, Yang J, Chen T, Yin Y, Li N, Li Y, Luo X, Dong E, Tan H, Ma Y, Li T. Generation of Human Endometrial Assembloids with a Luminal Epithelium using Air-Liquid Interface Culture Methods. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301868. [PMID: 37635169 PMCID: PMC10602567 DOI: 10.1002/advs.202301868] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/30/2023] [Indexed: 08/29/2023]
Abstract
The endometrial lining of the uterus is essential for women's reproductive health and consists of several different types of epithelial and stromal cells. Although models such as gland-like structures (GLSs) and endometrial assembloids (EnAos) are successfully established, they lack an intact luminal epithelium, which makes it difficult to recapitulate endometrial receptivity. Here, a novel EnAo model (ALI-EnAo) is developed by combining endometrial epithelial cells (EnECs) and stromal cells (EnSCs) and using an improved matrix and air-liquid interface (ALI) culture method. ALI-EnAos exhibit intact EnSCs and glandular and luminal epithelia, which recapitulates human endometrium anatomy, cell composition, hormone-induced menstrual cycle changes, gene expression profiles, and dynamic ciliogenesis. The model suggests that EnSCs, together with the extracellular matrix and ALI culture conditions, contribute to EnAo phenotypes and characteristics reflective of the endometrial menstrual cycle. This enables to transcriptionally define endometrial cell subpopulations. It anticipates that ALI-EnAos will facilitate studies on embryo implantation, and endometrial growth, differentiation, and disease.
Collapse
Affiliation(s)
- Jiwen Tian
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnan650032China
- Medical SchoolKunming University of Science and TechnologyKunmingYunnan650032China
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnan650021China
| | - Jie Yang
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnan650032China
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnan650500China
| | - Tingwei Chen
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnan650032China
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnan650500China
| | - Yu Yin
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnan650032China
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnan650500China
| | - Nan Li
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnan650032China
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnan650500China
| | - Yunxiu Li
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnan650021China
| | - Xingyu Luo
- Medical SchoolKunming University of Science and TechnologyKunmingYunnan650032China
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnan650021China
| | - E Dong
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnan650032China
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnan650500China
| | - Haoyang Tan
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnan650032China
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnan650021China
| | - Yanping Ma
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnan650021China
| | - Tianqing Li
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnan650032China
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnan650500China
| |
Collapse
|
16
|
Kühl L, Graichen P, von Daacke N, Mende A, Wygrecka M, Potaczek DP, Miethe S, Garn H. Human Lung Organoids-A Novel Experimental and Precision Medicine Approach. Cells 2023; 12:2067. [PMID: 37626876 PMCID: PMC10453737 DOI: 10.3390/cells12162067] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/31/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
The global burden of respiratory diseases is very high and still on the rise, prompting the need for accurate models for basic and translational research. Several model systems are currently available ranging from simple airway cell cultures to complex tissue-engineered lungs. In recent years, human lung organoids have been established as highly transferrable three-dimensional in vitro model systems for lung research. For acute infectious and chronic inflammatory diseases as well as lung cancer, human lung organoids have opened possibilities for precise in vitro research and a deeper understanding of mechanisms underlying lung injury and regeneration. Human lung organoids from induced pluripotent stem cells or from adult stem cells of patients' samples introduce tools for understanding developmental processes and personalized medicine approaches. When further state-of-the-art technologies and protocols come into use, the full potential of human lung organoids can be harnessed. High-throughput assays in drug development, gene therapy, and organoid transplantation are current applications of organoids in translational research. In this review, we emphasize novel approaches in translational and personalized medicine in lung research focusing on the use of human lung organoids.
Collapse
Affiliation(s)
- Laura Kühl
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (L.K.); (P.G.); (N.v.D.); (A.M.); (D.P.P.)
| | - Pauline Graichen
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (L.K.); (P.G.); (N.v.D.); (A.M.); (D.P.P.)
| | - Nele von Daacke
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (L.K.); (P.G.); (N.v.D.); (A.M.); (D.P.P.)
| | - Anne Mende
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (L.K.); (P.G.); (N.v.D.); (A.M.); (D.P.P.)
| | - Malgorzata Wygrecka
- Center for Infection and Genomics of the Lung (CIGL), Universities of Giessen and Marburg Lung Center (UGMLC), 35392 Giessen, Germany;
- Institute of Lung Health, German Center for Lung Research (DZL), 35392 Giessen, Germany
- CSL Behring Innovation GmbH, 35041 Marburg, Germany
| | - Daniel P. Potaczek
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (L.K.); (P.G.); (N.v.D.); (A.M.); (D.P.P.)
- Center for Infection and Genomics of the Lung (CIGL), Universities of Giessen and Marburg Lung Center (UGMLC), 35392 Giessen, Germany;
- Bioscientia MVZ Labor Mittelhessen GmbH, 35394 Giessen, Germany
| | - Sarah Miethe
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (L.K.); (P.G.); (N.v.D.); (A.M.); (D.P.P.)
| | - Holger Garn
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (L.K.); (P.G.); (N.v.D.); (A.M.); (D.P.P.)
| |
Collapse
|
17
|
Del Valle JS, Chuva de Sousa Lopes SM. Bioengineered 3D Ovarian Models as Paramount Technology for Female Health Management and Reproduction. Bioengineering (Basel) 2023; 10:832. [PMID: 37508859 PMCID: PMC10376580 DOI: 10.3390/bioengineering10070832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/30/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Ovarian dysfunction poses significant threats to the health of female individuals. Ovarian failure can lead to infertility due to the lack or inefficient production of fertilizable eggs. In addition, the ovary produces hormones, such as estrogen and progesterone, that play crucial roles not only during pregnancy, but also in maintaining cardiovascular, bone, and cognitive health. Decline in estrogen and progesterone production due to ovarian dysfunction can result in menopausal-associated syndromes and lead to conditions, such as osteoporosis, cardiovascular disease, and Alzheimer's disease. Recent advances in the design of bioengineered three-dimensional (3D) ovarian models, such as ovarian organoids or artificial ovaries, have made it possible to mimic aspects of the cellular heterogeneity and functional characteristics of the ovary in vitro. These novel technologies are emerging as valuable tools for studying ovarian physiology and pathology and may provide alternatives for fertility preservation. Moreover, they may have the potential to restore aspects of ovarian function, improving the quality of life of the (aging) female population. This review focuses on the state of the art of 3D ovarian platforms, including the latest advances modeling female reproduction, female physiology, ovarian cancer, and drug screening.
Collapse
Affiliation(s)
- Julieta S Del Valle
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Susana M Chuva de Sousa Lopes
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
- Department for Reproductive Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| |
Collapse
|
18
|
Gandhi NN, Inzana TJ, Rajagopalan P. Bovine Airway Models: Approaches for Investigating Bovine Respiratory Disease. ACS Infect Dis 2023; 9:1168-1179. [PMID: 37257116 DOI: 10.1021/acsinfecdis.2c00618] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Bovine respiratory disease (BRD) is a multifactorial condition where different genera of bacteria, such as Mannheimia haemolytica, Histophilus somni, Pasteurella multocida, and Mycoplasma bovis, and viruses, like bovine respiratory syncytial virus, bovine viral diarrhea virus, and bovine herpes virus-1, infect the lower respiratory tract of cattle. These pathogens can co-infect cells in the respiratory system, thereby making specific treatment very difficult. Currently, the most common models for studying BRD include a submerged tissue culture (STC), where monolayers of epithelial cells are typically covered either in cellular or spent biofilm culture medium. Another model is an air-liquid interface (ALI), where epithelial cells are exposed on their apical side and allowed to differentiate. However, limited work has been reported on the study of three-dimensional (3D) bovine models that incorporate multiple cell types to represent the architecture of the respiratory tract. The roles of different defense mechanisms in an infected bovine respiratory system, such as mucin production, tight junction barriers, and the production of antimicrobial peptides in in vitro cultures require further investigation in order to provide a comprehensive understanding of the disease pathogenesis. In this report, we describe the different aspects of BRD, including the most implicated pathogens and the respiratory tract, which are important to incorporate in disease models assembled in vitro. Although current advancements of bovine respiratory cultures have led to knowledge of the disease, 3D multicellular organoids that better recapitulate the in vivo environment exhibit potential for future investigations.
Collapse
Affiliation(s)
- Neeti N Gandhi
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Thomas J Inzana
- College of Veterinary Medicine, Long Island University, Brookville, New York 11548, United States
| | - Padmavathy Rajagopalan
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
19
|
Billiot CE, McDaniel MS, Lindgren NR, Swords WE. Pathogenesis of Achromobacter xylosoxidans respiratory infections: colonization and persistence of airway epithelia and differential gene expression in synthetic cystic fibrosis sputum medium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.04.535650. [PMID: 37066231 PMCID: PMC10104045 DOI: 10.1101/2023.04.04.535650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Cystic fibrosis (CF) is a genetic disease affecting epithelial ion transport, resulting in thickened mucus and impaired mucociliary clearance. Persons with CF (pwCF) experience life-long respiratory mucosal infections caused by a diverse array of opportunists, and these infections are a leading cause of morbidity and mortality for pwCF. In recent years, there has been increased appreciation for the range and diversity of microbes in CF-related respiratory infections. Introduction of new therapeutics and improved detection methodology has revealed CF related opportunists such as Achromobacter xylosoxidans (Ax). Ax is a Gram-negative bacterial species that is widely distributed in the environment and has been increasingly observed in sputa and other samples from pwCF; typically Ax infections occur in patients in later stages of CF disease. In this study, we characterized CF clinical isolates of Ax and tested colonization and persistence of Ax in respiratory infection using immortalized human CF respiratory epithelial cells and BALB/c mice. Genomic analyses of clinical Ax isolates showed homologs for factors involved in flagellar synthesis, antibiotic resistance, and toxin secretion systems. Ax isolates adhered to polarized CFBE14o- human immortalized CF bronchial epithelial cells and caused significant cytotoxicity and depolarization. Ax colonized and persisted in mouse lung for up to 72 hours post infection, with inflammatory consequences that include increased neutrophilia, lung damage, cytokine production, and mortality. Transcript profiling reveled differential expression of Ax genes during growth in SCFM2 synthetic CF sputum media. Based on these results, we conclude that Ax is an opportunistic pathogen of significance in CF.
Collapse
Affiliation(s)
- Caitlin E. Billiot
- Department of Medicine, Pulmonary, Allergy, and Critical Care Medicine
- Gregory Fleming James Center for Cystic Fibrosis Research University of Alabama at Birmingham
| | - Melissa S. McDaniel
- Department of Medicine, Pulmonary, Allergy, and Critical Care Medicine
- Gregory Fleming James Center for Cystic Fibrosis Research University of Alabama at Birmingham
| | - Natalie R. Lindgren
- Department of Medicine, Pulmonary, Allergy, and Critical Care Medicine
- Gregory Fleming James Center for Cystic Fibrosis Research University of Alabama at Birmingham
| | - W. Edward Swords
- Department of Medicine, Pulmonary, Allergy, and Critical Care Medicine
- Gregory Fleming James Center for Cystic Fibrosis Research University of Alabama at Birmingham
| |
Collapse
|
20
|
Marchal-Chaud H, Rieger R, Mai VT, Courtial EJ, Ottenio M, Bonnefont-Rebeix C, Bruyère K, Boulocher C. Contactless mechanical stimulation of tissue engineered constructs: Development and validation of an air-pulse device. BIOMATERIALS ADVANCES 2023; 149:213401. [PMID: 37018914 DOI: 10.1016/j.bioadv.2023.213401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/08/2023] [Accepted: 03/22/2023] [Indexed: 03/28/2023]
Abstract
OBJECTIVE Tissue engineering (TE) is the study and development of biological substitutes to restore, maintain or improve tissue function. Tissue engineered constructs (TECs) still present differences in mechanical and biological properties compared to native tissue. Mechanotransduction is the process through which mechanical stimulation triggers proliferation, apoptosis, and extracellular matrix synthesis, among other cell activities. Regarding that aspect, the effect of in vitro stimulations such as compression, stretching, bending or fluid shear stress loading modalities have been extensively studied. A fluid flow used to produce contactless mechanical stimulation induced by an air pulse could be easily achieved in vivo without altering the tissue integrity. METHODS A new air-pulse device for contactless and controlled mechanical simulation of a TECs was developed and validated in this study conducted in the following three phases: 1) conception of the controlled air-pulse device combined with a 3D printed bioreactor; 2) experimental and numerical mechanical characterization of the air-pulse impact by digital image correlation; and 3) achieving sterility and noncytotoxicity of the air-pulse and of the 3D printed bioreactor using a novel dedicated sterilization process. RESULTS We demonstrated that the treated PLA (polylactic acid) was noncytotoxic and did not influence cell proliferation. An ethanol/autoclaved sterilization protocol for 3D printed objects in PLA has been developed in this study, enabling the use of 3D printing in cell culture. A numerical twin of the device was developed and experimentally characterized by digital image correlation. It showed a coefficient of determination R2 = 0.98 between the numerical and averaged experimental surface displacement profiles of the TEC substitute. CONCLUSION The results of the study assessed the noncytotoxicity of PLA for prototyping by 3D printing the homemade bioreactor. A novel sterilization process for PLA was developed in this study based on a thermochemical process. A numerical twin using fluid-structure interaction method has been developed to investigate the micromechanical effects of air pulses inside the TEC, which cannot all be measured experimentally, for instance, wave propagation generated during the air-pulse impact. The device could be used to study the cell response to contactless cyclic mechanical stimulation, particularly in TEC with fibroblasts, stromal cells and mesenchymal stem cells, which have been shown to be sensitive to the frequency and strain level at the air-liquid interface.
Collapse
|
21
|
Smirnov A, Candi E. Take a breath: oxygen sensing of epidermal differentiation. FEBS J 2023; 290:2029-2031. [PMID: 36811892 DOI: 10.1111/febs.16752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/24/2023]
Abstract
Skin serves as a barrier to protect our body from injury, pathogens and trans-epidermal water loss. It is the only tissue directly exposed to oxygen besides lungs. Air exposure is an essential step of in vitro generation skin graft. However, the role of oxygen in this process remains hitherto unclear. Teshima et al. unveiled the impact of the hypoxia-inducible factor (HIF) pathway on epidermal differentiation in three-dimensional skin models. The authors of this work describe how air-lifting of organotypic epidermal cultures impairs HIFs activity, leading to a proper terminal differentiation of keratinocytes and stratification.
Collapse
Affiliation(s)
- Artem Smirnov
- Department of Experimental Medicine, Torvergata Oncoscience Research Centre of Excellence, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Eleonora Candi
- Department of Experimental Medicine, Torvergata Oncoscience Research Centre of Excellence, TOR, University of Rome "Tor Vergata", Rome, Italy.,Biochemistry Laboratory, Istituto Dermopatico Immacolata (IDI-IRCCS), Rome, Italy
| |
Collapse
|
22
|
Liu Y, Li P, Jiang T, Li Y, Wang Y, Cheng Z. Epidermal growth factor receptor in asthma: A promising therapeutic target? Respir Med 2023; 207:107117. [PMID: 36626942 DOI: 10.1016/j.rmed.2023.107117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/09/2023]
Abstract
Activation of the epidermal growth factor receptor (EGFR) pathway is involved in the pathogenesis of asthma. Although decades of intensive research have focused on the role of EGFR in asthma, the specific mechanisms and pathways of EGFR signaling remain unclear. Various reports have indicated that inhibition of EGFR improves the pathological features in asthma models. However, extending these experimental findings to clinical applications is difficult. Several measures can be adopted to promote clinical application of EGFR inhibitors. This review focuses on the role of EGFR in the pathogenesis of asthma and the development of a potentially novel therapeutic target for asthma.
Collapse
Affiliation(s)
- Ye Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Pengfei Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Tianci Jiang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yue Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yu Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Zhe Cheng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
23
|
Rothen-Rutishauser B, Gibb M, He R, Petri-Fink A, Sayes CM. Human lung cell models to study aerosol delivery - considerations for model design and development. Eur J Pharm Sci 2023; 180:106337. [PMID: 36410570 DOI: 10.1016/j.ejps.2022.106337] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Human lung tissue models range from simple monolayer cultures to more advanced three-dimensional co-cultures. Each model system can address the interactions of different types of aerosols and the choice of the model and the mode of aerosol exposure depends on the relevant scenario, such as adverse outcomes and endpoints of interest. This review focuses on the functional, as well as structural, aspects of lung tissue from the upper airway to the distal alveolar compartments as this information is relevant for the design of a model as well as how the aerosol properties determine the interfacial properties with the respiratory wall. The most important aspects on how to design lung models are summarized with a focus on (i) choice of appropriate scaffold, (ii) selection of cell types for healthy and diseased lung models, (iii) use of culture condition and assembly, (iv) aerosol exposure methods, and (v) endpoints and verification process. Finally, remaining challenges and future directions in this field are discussed.
Collapse
Affiliation(s)
- Barbara Rothen-Rutishauser
- BioNanomaterials, Adolphe Merkle Institute, University Fribourg, Chemin des Verdiers 4 CH-1700, Fribourg, Switzerland.
| | - Matthew Gibb
- Department of Environmental Science, Baylor University, One Bear Place #97266, Waco, TX 76798-7266, USA
| | - Ruiwen He
- BioNanomaterials, Adolphe Merkle Institute, University Fribourg, Chemin des Verdiers 4 CH-1700, Fribourg, Switzerland
| | - Alke Petri-Fink
- BioNanomaterials, Adolphe Merkle Institute, University Fribourg, Chemin des Verdiers 4 CH-1700, Fribourg, Switzerland
| | - Christie M Sayes
- Department of Environmental Science, Baylor University, One Bear Place #97266, Waco, TX 76798-7266, USA.
| |
Collapse
|
24
|
Di Cristo L, Sabella S. Cell Cultures at the Air-Liquid Interface and Their Application in Cancer Research. Methods Mol Biol 2023; 2645:41-64. [PMID: 37202611 DOI: 10.1007/978-1-0716-3056-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Air-liquid interface (ALI) cell cultures are considered a valid tool for the replacement of animals in biomedical research. By mimicking crucial features of the human in vivo epithelial barriers (e.g., lung, intestine, and skin), ALI cell cultures enable proper structural architectures and differentiated functions of normal and diseased tissue barriers. Thereby, ALI models realistically resemble tissue conditions and provide in vivo-like responses. Since their implementation, they are routinely used in several applications, from toxicity testing to cancer research, receiving an appreciable level of acceptance (in some cases a regulatory acceptance) as attractive testing alternatives to animals. In this chapter, an overview of the ALI cell cultures will be presented together with their application in cancer cell culture, highlighting the potential advantages and disadvantages of the model.
Collapse
Affiliation(s)
- Luisana Di Cristo
- D3 PharmaChemistry, Nanoregulatory Group, Italian Institute of Technology, Genoa, Italy.
| | - Stefania Sabella
- D3 PharmaChemistry, Nanoregulatory Group, Italian Institute of Technology, Genoa, Italy
| |
Collapse
|
25
|
Long-term culture of feline oviduct epithelial cells on permeable filter supports. Cytotechnology 2022; 74:531-538. [PMID: 36238264 PMCID: PMC9525501 DOI: 10.1007/s10616-022-00542-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 08/09/2022] [Indexed: 11/30/2022] Open
Abstract
Basic knowledge about cellular and molecular mechanisms underlying feline reproduction is required to improve reproductive biotechnologies in endangered felids. Commonly, the domestic cat (Felis catus) is used as a model species, but many of the fine-tuned, dynamic reproductive processes can hardly be observed in vivo. This necessitates the development of in vitro models. The oviduct is a central reproductive organ hosting fertilization in the ampulla and early embryonic development in the isthmus part, which also functions as a sperm reservoir before fertilization. In other species, culturing oviduct epithelial cells in compartmentalized culture systems has proven useful to maintain oviduct epithelium polarization and functionality. Therefore, we made the first attempt to establish a compartmentalized long-term culture system of feline oviduct epithelial cells from both ampulla and isthmus. Cells were isolated from tissue samples (n = 33 animals) after routine gonadectomy, seeded on permeable filter supports and cultured at the liquid–liquid or air–liquid interface. Cultures were harvested after 21 days and microscopically evaluated for epithelial differentiation (monolayer formation with basal–apical polarization) and protein expression of marker genes (oviduct-specific glycoprotein, acetylated tubulin). Due to the heterogeneous and undefined native tissue material available for this study, the applied cell culture approach was only successful in a limited number of cases (five differentiated cultures). Even though the protocol needs optimization, our study showed that the compartmentalized culture approach is suitable for maintaining differentiated epithelial cells from both isthmus and ampulla of the feline oviduct.
Collapse
|
26
|
Corsini NS, Knoblich JA. Human organoids: New strategies and methods for analyzing human development and disease. Cell 2022; 185:2756-2769. [PMID: 35868278 DOI: 10.1016/j.cell.2022.06.051] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 01/06/2023]
Abstract
For decades, insight into fundamental principles of human biology and disease has been obtained primarily by experiments in animal models. While this has allowed researchers to understand many human biological processes in great detail, some developmental and disease mechanisms have proven difficult to study due to inherent species differences. The advent of organoid technology more than 10 years ago has established laboratory-grown organ tissues as an additional model system to recapitulate human-specific aspects of biology. The use of human 3D organoids, as well as other advances in single-cell technologies, has revealed unprecedented insights into human biology and disease mechanisms, especially those that distinguish humans from other species. This review highlights novel advances in organoid biology with a focus on how organoid technology has generated a better understanding of human-specific processes in development and disease.
Collapse
Affiliation(s)
- Nina S Corsini
- IMBA - Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter (VBC), Vienna, Austria
| | - Juergen A Knoblich
- IMBA - Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter (VBC), Vienna, Austria; Medical University of Vienna, Department of Neurology, Vienna, Austria.
| |
Collapse
|
27
|
Luminal and Glandular Epithelial Cells from the Porcine Endometrium maintain Cell Type-Specific Marker Gene Expression in Air-Liquid Interface Culture. Stem Cell Rev Rep 2022; 18:2928-2938. [PMID: 35849251 PMCID: PMC9622560 DOI: 10.1007/s12015-022-10410-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2022] [Indexed: 11/17/2022]
Abstract
Two different types of epithelial cells constitute the inner surface of the endometrium. While luminal epithelial cells line the uterine cavity and build the embryo-maternal contact zone, glandular epithelial cells form tubular glands reaching deeply into the endometrial stroma. To facilitate investigations considering the functional and molecular differences between the two populations of epithelial cells and their contribution to reproductive processes, we aimed at establishing differentiated in vitro models of both the luminal and the glandular epithelium of the porcine endometrium using an air–liquid interface (ALI) approach. We first tested if porcine luminal endometrium epithelial cells (PEEC-L) reproducibly form differentiated epithelial monolayers under ALI conditions by monitoring the morphology and the trans-epithelial electrical resistance (TEER). Subsequently, luminal (PEEC-L) and glandular epithelial cells (PEEC-G) were consecutively isolated from the endometrium of the uterine horn. Both cell types were characterized by marker gene expression analysis immediately after isolation. Cells were separately grown at the ALI and assessed by means of histomorphometry, TEER, and marker gene expression after 3 weeks of culture. PEEC-L and PEEC-G formed polarized monolayers of differentiated epithelial cells with a moderate TEER and in vivo-like morphology at the ALI. They exhibited distinct patterns of functional and cell type-specific marker gene expression after isolation and largely maintained these patterns during the culture period. The here presented cell culture procedure for PEEC-L and -G offers new opportunities to study the impact of embryonic signals, endocrine effectors, and reproductive toxins on both porcine endometrial epithelial cell types under standardized in vitro conditions.
Collapse
|
28
|
Abo KM, Sainz de Aja J, Lindstrom-Vautrin J, Alysandratos KD, Richards A, Garcia-de-Alba C, Huang J, Hix OT, Werder RB, Bullitt E, Hinds A, Falconer I, Villacorta-Martin C, Jaenisch R, Kim CF, Kotton DN, Wilson AA. Air-liquid interface culture promotes maturation and allows environmental exposure of pluripotent stem cell-derived alveolar epithelium. JCI Insight 2022; 7:155589. [PMID: 35315362 PMCID: PMC8986076 DOI: 10.1172/jci.insight.155589] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Type 2 alveolar epithelial cells (AT2s), facultative progenitor cells of the lung alveolus, play a vital role in the biology of the distal lung. In vitro model systems that incorporate human cells, recapitulate the biology of primary AT2s, and interface with the outside environment could serve as useful tools to elucidate functional characteristics of AT2s in homeostasis and disease. We and others recently adapted human induced pluripotent stem cell–derived AT2s (iAT2s) for air-liquid interface (ALI) culture. Here, we comprehensively characterize the effects of ALI culture on iAT2s and benchmark their transcriptional profile relative to both freshly sorted and cultured primary human fetal and adult AT2s. We find that iAT2s cultured at ALI maintain an AT2 phenotype while upregulating expression of transcripts associated with AT2 maturation. We then leverage this platform to assay the effects of exposure to clinically significant, inhaled toxicants including cigarette smoke and electronic cigarette vapor.
Collapse
Affiliation(s)
- Kristine M Abo
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA.,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Julio Sainz de Aja
- Stem Cell Program and Divisions of Hematology/Oncology and Pulmonary & Respiratory Diseases, Boston Children's Hospital, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan Lindstrom-Vautrin
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
| | - Konstantinos-Dionysios Alysandratos
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA.,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Alexsia Richards
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
| | - Carolina Garcia-de-Alba
- Stem Cell Program and Divisions of Hematology/Oncology and Pulmonary & Respiratory Diseases, Boston Children's Hospital, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Jessie Huang
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA.,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Olivia T Hix
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA.,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Rhiannon B Werder
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA.,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Esther Bullitt
- Department of Physiology & Biophysics, Boston University, Boston, Massachusetts, USA
| | - Anne Hinds
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Isaac Falconer
- Boston University School of Medicine, Boston, Massachusetts, USA
| | - Carlos Villacorta-Martin
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Carla F Kim
- Stem Cell Program and Divisions of Hematology/Oncology and Pulmonary & Respiratory Diseases, Boston Children's Hospital, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Darrell N Kotton
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA.,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Andrew A Wilson
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA.,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
29
|
Chen Q, Langenbach S, Li M, Xia YC, Gao X, Gartner MJ, Pharo EA, Williams SM, Todd S, Clarke N, Ranganathan S, Baker ML, Subbarao K, Stewart AG. ACE2 Expression in Organotypic Human Airway Epithelial Cultures and Airway Biopsies. Front Pharmacol 2022; 13:813087. [PMID: 35359837 PMCID: PMC8963460 DOI: 10.3389/fphar.2022.813087] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/31/2022] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an acute respiratory disease with systemic complications. Therapeutic strategies for COVID-19, including repurposing (partially) developed drugs are urgently needed, regardless of the increasingly successful vaccination outcomes. We characterized two-dimensional (2D) and three-dimensional models (3D) to establish a physiologically relevant airway epithelial model with potential for investigating SARS-CoV-2 therapeutics. Human airway basal epithelial cells maintained in submerged 2D culture were used at low passage to retain the capacity to differentiate into ciliated, club, and goblet cells in both air-liquid interface culture (ALI) and airway organoid cultures, which were then analyzed for cell phenotype makers. Airway biopsies from non-asthmatic and asthmatic donors enabled comparative evaluation of the level and distribution of immunoreactive angiotensin-converting enzyme 2 (ACE2). ACE2 and transmembrane serine proteinase 2 (TMPRSS2) mRNA were expressed in ALI and airway organoids at levels similar to those of native (i.e., non-cultured) human bronchial epithelial cells, whereas furin expression was more faithfully represented in ALI. ACE2 was mainly localized to ciliated and basal epithelial cells in human airway biopsies, ALI, and airway organoids. Cystic fibrosis appeared to have no influence on ACE2 gene expression. Neither asthma nor smoking status had consistent marked influence on the expression or distribution of ACE2 in airway biopsies. SARS-CoV-2 infection of ALI cultures did not increase the levels of selected cytokines. Organotypic, and particularly ALI airway cultures are useful and practical tools for investigation of SARS-CoV-2 infection and evaluating the clinical potential of therapeutics for COVID-19.
Collapse
Affiliation(s)
- Qianyu Chen
- Department of Biochemistry and Pharmacology, School of Biomedical Science, University of Melbourne, Parkville, VIC, Australia
- ARC Centre for Personalized Therapeutics Technologies, University of Melbourne, Parkville, VIC, Australia
| | - Shenna Langenbach
- Department of Biochemistry and Pharmacology, School of Biomedical Science, University of Melbourne, Parkville, VIC, Australia
- ARC Centre for Personalized Therapeutics Technologies, University of Melbourne, Parkville, VIC, Australia
| | - Meina Li
- Department of Biochemistry and Pharmacology, School of Biomedical Science, University of Melbourne, Parkville, VIC, Australia
- ARC Centre for Personalized Therapeutics Technologies, University of Melbourne, Parkville, VIC, Australia
| | - Yuxiu C. Xia
- Department of Biochemistry and Pharmacology, School of Biomedical Science, University of Melbourne, Parkville, VIC, Australia
| | - Xumei Gao
- Department of Biochemistry and Pharmacology, School of Biomedical Science, University of Melbourne, Parkville, VIC, Australia
- ARC Centre for Personalized Therapeutics Technologies, University of Melbourne, Parkville, VIC, Australia
| | - Matthew J. Gartner
- Department of Microbiology and Immunology, University of Melbourne, Parkville, VIC, Australia
| | - Elizabeth A. Pharo
- CSIRO, Health and Biosecurity Business Unit, Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Sinéad M. Williams
- CSIRO, Health and Biosecurity Business Unit, Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Shawn Todd
- CSIRO, Health and Biosecurity Business Unit, Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Nadeene Clarke
- Murdoch Children’s Research Institute, The Royal Children’s Hospital, Parkville, VIC, Australia
| | - Sarath Ranganathan
- Murdoch Children’s Research Institute, The Royal Children’s Hospital, Parkville, VIC, Australia
- Department of Pediatrics, Melbourne Medical School, University of Melbourne, Parkville, VIC, Australia
| | - Michelle L. Baker
- CSIRO, Health and Biosecurity Business Unit, Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, University of Melbourne, Parkville, VIC, Australia
- WHO Collaborating Centre for Reference and Research on Influenza at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Alastair G. Stewart
- Department of Biochemistry and Pharmacology, School of Biomedical Science, University of Melbourne, Parkville, VIC, Australia
- ARC Centre for Personalized Therapeutics Technologies, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
30
|
Histological changes and transglutaminase 2 expression in the oviduct of advanced pregnant cows. Reprod Biol 2022; 22:100616. [DOI: 10.1016/j.repbio.2022.100616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/04/2022] [Accepted: 02/06/2022] [Indexed: 11/30/2022]
|
31
|
Moreira A, Müller M, Costa PF, Kohl Y. Advanced In Vitro Lung Models for Drug and Toxicity Screening: The Promising Role of Induced Pluripotent Stem Cells. Adv Biol (Weinh) 2021; 6:e2101139. [PMID: 34962104 DOI: 10.1002/adbi.202101139] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/25/2021] [Indexed: 12/24/2022]
Abstract
The substantial socioeconomic burden of lung diseases, recently highlighted by the disastrous impact of the coronavirus disease 2019 (COVID-19) pandemic, accentuates the need for interventive treatments capable of decelerating disease progression, limiting organ damage, and contributing to a functional tissue recovery. However, this is hampered by the lack of accurate human lung research models, which currently fail to reproduce the human pulmonary architecture and biochemical environment. Induced pluripotent stem cells (iPSCs) and organ-on-chip (OOC) technologies possess suitable characteristics for the generation of physiologically relevant in vitro lung models, allowing for developmental studies, disease modeling, and toxicological screening. Importantly, these platforms represent potential alternatives for animal testing, according to the 3Rs (replace, reduce, refine) principle, and hold promise for the identification and approval of new chemicals under the European REACH (registration, evaluation, authorization and restriction of chemicals) framework. As such, this review aims to summarize recent progress made in human iPSC- and OOC-based in vitro lung models. A general overview of the present applications of in vitro lung models is presented, followed by a summary of currently used protocols to generate different lung cell types from iPSCs. Lastly, recently developed iPSC-based lung models are discussed.
Collapse
Affiliation(s)
| | - Michelle Müller
- Department of Bioprocessing and Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280, Sulzbach, Germany
| | - Pedro F Costa
- BIOFABICS, Rua Alfredo Allen 455, Porto, 4200-135, Portugal
| | - Yvonne Kohl
- Department of Bioprocessing and Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280, Sulzbach, Germany.,Postgraduate Course for Toxicology and Environmental Toxicology, Medical Faculty, University of Leipzig, Johannisallee 28, 04103, Leipzig, Germany
| |
Collapse
|
32
|
Lusk H, Burdette JE, Sanchez LM. Models for measuring metabolic chemical changes in the metastasis of high grade serous ovarian cancer: fallopian tube, ovary, and omentum. Mol Omics 2021; 17:819-832. [PMID: 34338690 PMCID: PMC8649074 DOI: 10.1039/d1mo00074h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Ovarian cancer (OC) is the most lethal gynecologic malignancy and high grade serous ovarian cancer (HGSOC) is the most common and deadly subtype, accounting for 70-80% of OC deaths. HGSOC has a distinct pattern of metastasis as many believe it originates in the fallopian tube and then it metastasizes first to the ovary, and later to the adipose-rich omentum. Metabolomics has been heavily utilized to investigate metabolite changes in HGSOC tumors and metastasis. Generally, metabolomics studies have traditionally been applied to biospecimens from patients or animal models; a number of recent studies have combined metabolomics with innovative cell-culture techniques to model the HGSOC metastatic microenvironment for the investigation of cell-to-cell communication. The purpose of this review is to serve as a tool for researchers aiming to model the metastasis of HGSOC for metabolomics analyses. It will provide a comprehensive overview of current knowledge on the origin and pattern of metastasis of HGSOC and discuss the advantages and limitations of different model systems to help investigators choose the best model for their research goals, with a special emphasis on compatibility with different metabolomics modalities. It will also examine what is presently known about the role of small molecules in the origin and metastasis of HGSOC.
Collapse
Affiliation(s)
- Hannah Lusk
- Department of Chemistry and Biochemistry, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA, 95064, USA.
| | - Joanna E Burdette
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 900 S Ashland Ave., Chicago, IL, 60607, USA
| | - Laura M Sanchez
- Department of Chemistry and Biochemistry, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA, 95064, USA.
| |
Collapse
|
33
|
Lagowala DA, Kwon S, Sidhaye VK, Kim DH. Human microphysiological models of airway and alveolar epithelia. Am J Physiol Lung Cell Mol Physiol 2021; 321:L1072-L1088. [PMID: 34612064 PMCID: PMC8715018 DOI: 10.1152/ajplung.00103.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 09/21/2021] [Accepted: 09/25/2021] [Indexed: 11/22/2022] Open
Abstract
Human organ-on-a-chip models are powerful tools for preclinical research that can be used to study the mechanisms of disease and evaluate new targets for therapeutic intervention. Lung-on-a-chip models have been one of the most well-characterized designs in this field and can be altered to evaluate various types of respiratory disease and to assess treatment candidates prior to clinical testing. These systems are capable of overcoming the flaws of conventional two-dimensional (2-D) cell culture and in vivo animal testing due to their ability to accurately recapitulate the in vivo microenvironment of human tissue with tunable material properties, microfluidic integration, delivery of precise mechanical and biochemical cues, and designs with organ-specific architecture. In this review, we first describe an overview of currently available lung-on-a-chip designs. We then present how recent innovations in human stem cell biology, tissue engineering, and microfabrication can be used to create more predictive human lung-on-a-chip models for studying respiratory disease. Finally, we discuss the current challenges and future directions of lung-on-a-chip designs for in vitro disease modeling with a particular focus on immune and multiorgan interactions.
Collapse
Affiliation(s)
- Dave Anuj Lagowala
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Seoyoung Kwon
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Venkataramana K Sidhaye
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
34
|
Abstract
Many studies have focused on the optimization of the composition of embryo culture medium; however, there are few studies involving the effect of a culture medium changing procedure on the preimplantation development of embryos. In this study, three groups were designed: a non-renewal group, a renewal group and a half-renewal group. The levels of reactive oxygen species (ROS), apoptotic index, blastocyst ratio and blastocyst total cell number were analyzed in each group. The results showed that the ROS level and the apoptotic index of blastocyst in the non-renewal group were significantly higher than in the renewal group and the half-renewal group (P < 0.05). The blastocyst ratio and blastocyst total cell number were significantly higher in the half-renewal group than that in non-renewal group and the renewal group (P < 0.05). These results demonstrated that the procedure of changing the culture medium influenced ROS level, apoptotic index, blastocyst ratio and total cell number of blastocysts. In addition, the result suggested that changing the culture medium may lead to a loss of important regulatory factors for embryos, while not changing the culture medium may lead to the accumulation of toxic substances. Half-renewal can alleviate the defects of both no renewal and renewal, and benefit embryo development. This study will be of high value as a reference for the optimization of embryo culture in vitro, and is very significant for assisted reproduction.
Collapse
|
35
|
Stejskalová A, Vankelecom H, Sourouni M, Ho MY, Götte M, Almquist BD. In vitro modelling of the physiological and diseased female reproductive system. Acta Biomater 2021; 132:288-312. [PMID: 33915315 DOI: 10.1016/j.actbio.2021.04.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023]
Abstract
The maladies affecting the female reproductive tract (FRT) range from infections to endometriosis to carcinomas. In vitro models of the FRT play an increasingly important role in both basic and translational research, since the anatomy and physiology of the FRT of humans and other primates differ significantly from most of the commonly used animal models, including rodents. Using organoid culture to study the FRT has overcome the longstanding hurdle of maintaining epithelial phenotype in culture. Both ECM-derived and engineered materials have proved critical for maintaining a physiological phenotype of FRT cells in vitro by providing the requisite 3D environment, ligands, and architecture. Advanced materials have also enabled the systematic study of factors contributing to the invasive metastatic processes. Meanwhile, microphysiological devices make it possible to incorporate physical signals such as flow and cyclic exposure to hormones. Going forward, advanced materials compatible with hormones and optimised to support FRT-derived cells' long-term growth, will play a key role in addressing the diverse array of FRT pathologies and lead to impactful new treatments that support the improvement of women's health. STATEMENT OF SIGNIFICANCE: The female reproductive system is a crucial component of the female anatomy. In addition to enabling reproduction, it has wide ranging influence on tissues throughout the body via endocrine signalling. This intrinsic role in regulating normal female biology makes it susceptible to a variety of female-specific diseases. However, the complexity and human-specific features of the reproductive system make it challenging to study. This has spurred the development of human-relevant in vitro models for helping to decipher the complex issues that can affect the reproductive system, including endometriosis, infection, and cancer. In this Review, we cover the current state of in vitro models for studying the female reproductive system, and the key role biomaterials play in enabling their development.
Collapse
|
36
|
Yaqub N, Wayne G, Birchall M, Song W. Recent advances in human respiratory epithelium models for drug discovery. Biotechnol Adv 2021; 54:107832. [PMID: 34481894 DOI: 10.1016/j.biotechadv.2021.107832] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/08/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022]
Abstract
The respiratory epithelium is intimately associated with the pathophysiologies of highly infectious viral contagions and chronic illnesses such as chronic obstructive pulmonary disorder, presently the third leading cause of death worldwide with a projected economic burden of £1.7 trillion by 2030. Preclinical studies of respiratory physiology have almost exclusively utilised non-humanised animal models, alongside reductionistic cell line-based models, and primary epithelial cell models cultured at an air-liquid interface (ALI). Despite their utility, these model systems have been limited by their poor correlation to the human condition. This has undermined the ability to identify novel therapeutics, evidenced by a 15% chance of success for medicinal respiratory compounds entering clinical trials in 2018. Consequently, preclinical studies require new translational efficacy models to address the problem of respiratory drug attrition. This review describes the utility of the current in vivo (rodent), ex vivo (isolated perfused lungs and precision cut lung slices), two-dimensional in vitro cell-line (A549, BEAS-2B, Calu-3) and three-dimensional in vitro ALI (gold-standard and co-culture) and organoid respiratory epithelium models. The limitations to the application of these model systems in drug discovery research are discussed, in addition to perspectives of the future innovations required to facilitate the next generation of human-relevant respiratory models.
Collapse
Affiliation(s)
- Naheem Yaqub
- UCL Centre for Biomaterials in Surgical Reconstruction and Regeneration, Department of Surgical Biotechnology, Division of Surgery & Interventional Science, University College London, London NW3 2PF, UK
| | - Gareth Wayne
- Novel Human Genetics, GlaxoSmithKline, Stevenage SG1 2NY, UK
| | - Martin Birchall
- The Ear Institute, Faculty of Brain Sciences, University College London, London WC1X 8EE, UK.
| | - Wenhui Song
- UCL Centre for Biomaterials in Surgical Reconstruction and Regeneration, Department of Surgical Biotechnology, Division of Surgery & Interventional Science, University College London, London NW3 2PF, UK.
| |
Collapse
|
37
|
Guindolet D, Crouzet E, He Z, Herbepin P, Perrache C, Garcin T, Gauthier AS, Forest F, Peoc'h M, Gain P, Gabison E, Thuret G. Epithelial Regeneration in Human Corneas Preserved in an Active Storage Machine. Transl Vis Sci Technol 2021; 10:31. [PMID: 34003916 PMCID: PMC7900847 DOI: 10.1167/tvst.10.2.31] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose To characterize the corneal epithelium (CE) and limbal epithelium (LE) of human corneas stored in an innovative active storage machine (ASM) after a period of organ culture (OC). Methods Corneas unsuitable for graft and stored in a standard commercial OC medium for 2 to 5 weeks were transferred into our ASM for 14 days. The ASM actively maintained an overpressure on the endothelial side (20 mm Hg) while ensuring medium renewal. We compared three modalities of storage in the ASM's epithelial chamber: (1) alternating exposure to a supplemental hormonal epithelial medium (SHEM) and air (air-lifting), (2) continuous immersion in SHEM, and (3) continuous immersion in OC medium. Passive immersion of the whole cornea in OC medium or of the CE in SHEM with or without airlifting served as controls. Paired corneas were used for better comparability. Histology, differentiation (by immunolabeling), and ultrastructure were analyzed at the end. Results The ASM with air-lifting was most effective in regenerating a pluristratified and differentiated CE (apical ZO-1 and MUC16 staining and regeneration of the glycocalyx). In addition, the LE was stratified with preserved expression of ABCB5. The ASM with immersion in SHEM or OC medium gave a less stratified and differentiated CE. In the three control groups, the epithelia, when present, were paucistratified and less differentiated. Conclusions In human corneas previously stored in OC, the ASM regenerates a CE with differentiation characteristics close to normal. Translational Relevance Regeneration of the epithelium of human corneas discarded by eye banks will increase tissue availability for research.
Collapse
Affiliation(s)
- Damien Guindolet
- Corneal Graft Biology, Engineering and Imaging Laboratory, BiiGC, EA2521, SFR143, Federative Institute of Research in Sciences and Health Engineering, Faculty of Medicine, Jean Monnet University, Saint-Etienne, France.,Cornea and External Disorders Department, Rothschild Foundation Hospital, Paris, France
| | - Emmanuel Crouzet
- Corneal Graft Biology, Engineering and Imaging Laboratory, BiiGC, EA2521, SFR143, Federative Institute of Research in Sciences and Health Engineering, Faculty of Medicine, Jean Monnet University, Saint-Etienne, France
| | - Zhiguo He
- Corneal Graft Biology, Engineering and Imaging Laboratory, BiiGC, EA2521, SFR143, Federative Institute of Research in Sciences and Health Engineering, Faculty of Medicine, Jean Monnet University, Saint-Etienne, France
| | - Pascal Herbepin
- Corneal Graft Biology, Engineering and Imaging Laboratory, BiiGC, EA2521, SFR143, Federative Institute of Research in Sciences and Health Engineering, Faculty of Medicine, Jean Monnet University, Saint-Etienne, France
| | - Chantal Perrache
- Corneal Graft Biology, Engineering and Imaging Laboratory, BiiGC, EA2521, SFR143, Federative Institute of Research in Sciences and Health Engineering, Faculty of Medicine, Jean Monnet University, Saint-Etienne, France
| | - Thibaud Garcin
- Corneal Graft Biology, Engineering and Imaging Laboratory, BiiGC, EA2521, SFR143, Federative Institute of Research in Sciences and Health Engineering, Faculty of Medicine, Jean Monnet University, Saint-Etienne, France.,Ophthalmology Department, University Hospital, Saint-Etienne, France
| | - Anne-Sophie Gauthier
- Corneal Graft Biology, Engineering and Imaging Laboratory, BiiGC, EA2521, SFR143, Federative Institute of Research in Sciences and Health Engineering, Faculty of Medicine, Jean Monnet University, Saint-Etienne, France
| | - Fabien Forest
- Corneal Graft Biology, Engineering and Imaging Laboratory, BiiGC, EA2521, SFR143, Federative Institute of Research in Sciences and Health Engineering, Faculty of Medicine, Jean Monnet University, Saint-Etienne, France.,Pathology Department, University Hospital, Saint-Etienne, France
| | - Michel Peoc'h
- Corneal Graft Biology, Engineering and Imaging Laboratory, BiiGC, EA2521, SFR143, Federative Institute of Research in Sciences and Health Engineering, Faculty of Medicine, Jean Monnet University, Saint-Etienne, France.,Pathology Department, University Hospital, Saint-Etienne, France
| | - Philippe Gain
- Corneal Graft Biology, Engineering and Imaging Laboratory, BiiGC, EA2521, SFR143, Federative Institute of Research in Sciences and Health Engineering, Faculty of Medicine, Jean Monnet University, Saint-Etienne, France.,Ophthalmology Department, University Hospital, Saint-Etienne, France
| | - Eric Gabison
- Cornea and External Disorders Department, Rothschild Foundation Hospital, Paris, France.,Ophthalmology Department, Bichat-Claude Bernard Hospital, Paris, France.,Université de Paris, Paris, France
| | - Gilles Thuret
- Corneal Graft Biology, Engineering and Imaging Laboratory, BiiGC, EA2521, SFR143, Federative Institute of Research in Sciences and Health Engineering, Faculty of Medicine, Jean Monnet University, Saint-Etienne, France.,Ophthalmology Department, University Hospital, Saint-Etienne, France.,Institut Universitaire de France, Paris, France
| |
Collapse
|
38
|
Chen KY, Tseng CH, Feng PH, Sun WL, Ho SC, Lin CW, Van Hiep N, Luo CS, Tseng YH, Chen TT, Liu WT, Lee KY, Wu SM. 3-Nitrobenzanthrone promotes malignant transformation in human lung epithelial cells through the epiregulin-signaling pathway. Cell Biol Toxicol 2021; 38:865-887. [PMID: 34036453 DOI: 10.1007/s10565-021-09612-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023]
Abstract
Exposure to environmental and occupational contaminants leads to lung cancer. 3-Nitrobenzanthrone (3-nitro-7H-benz[de]anthracen-7-one, 3-NBA) is a potential carcinogen in ambient air or diesel particulate matter. Studies have revealed that short-term exposure to 3-NBA induces cell death, reactive oxygen species activation, and DNA adduct formation and damage. However, details of the mechanism by which chronic exposure to 3-NBA influences lung carcinogenesis remain largely unknown. In this study, human lung epithelial BEAS-2B cells were continuously exposed to 0-10-μM 3-NBA for 6 months. NanoString analysis was conducted to evaluate gene expression in the cells, revealing that 3-NBA-mediated transformation results in a distinct gene expression signature including carbon cancer metabolism, metastasis, and angiogenesis. Alterations in tumor-promoting genes such as EREG (epiregulin), SOX9, E-cadherin, TWIST, and IL-6 were involved in epithelial cell aggressiveness. Kaplan-Meier plotter analyses indicated that increased EREG and IL-6 expressions in early-stage lung cancer cells are correlated with poor survival. In vivo xenografts on 3-NBA-transformed cells exhibited prominent tumor formation and metastasis. EREG knockout cells exposed to 3-NBA for a short period exhibited high apoptosis and low colony formation. By contrast, overexpression of EREG in 3-NBA-transformed cells markedly activated the PI3K/AKT and MEK/ERK signaling pathways, resulting in tumorigenicity. Furthermore, elevated IL-6 and EREG expressions synergistically led to STAT3 signaling activation, resulting in clonogenic cell survival and migration. Taken together, chronic exposure of human lung epithelial cells to 3-NBA leads to malignant transformation, in which the EREG signaling pathway plays a pivotal mediating role. • Short-term exposure of lung epithelial cells to 3-NBA can lead to ROS production and cell apoptosis. • Long-term chronic exposure to 3-NBA upregulates the levels of tumor-promoting genes such as EREG and IL-6. • Increased EREG expression in 3-NBA-transformed cells markedly contributes to tumorigenesis through PI3K/AKT and MEK/ERK activation and synergistically enhances the IL-6/STAT3 signaling pathway, which promotes tumorigenicity.
Collapse
Affiliation(s)
- Kuan-Yuan Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chien-Hua Tseng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Lun Sun
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shu-Chuan Ho
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Wei Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Nguyen Van Hiep
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,International PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ching-Shan Luo
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yen-Han Tseng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Tao Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wen-Te Liu
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kang-Yun Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
39
|
Ziegler P, Tian Y, Bai Y, Abrahamsson S, Bäckerholm A, Reznik AS, Green A, Moore JA, Lee SE, Myerburg MM, Park HJ, Tang KW, Shair KHY. A primary nasopharyngeal three-dimensional air-liquid interface cell culture model of the pseudostratified epithelium reveals differential donor- and cell type-specific susceptibility to Epstein-Barr virus infection. PLoS Pathog 2021; 17:e1009041. [PMID: 33914843 PMCID: PMC8112674 DOI: 10.1371/journal.ppat.1009041] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 05/11/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023] Open
Abstract
Epstein-Barr virus (EBV) is a ubiquitous γ-herpesvirus with latent and lytic cycles. EBV replicates in the stratified epithelium but the nasopharynx is also composed of pseudostratified epithelium with distinct cell types. Latent infection is associated with nasopharyngeal carcinoma (NPC). Here, we show with nasopharyngeal conditionally reprogrammed cells cultured at the air-liquid interface that pseudostratified epithelial cells are susceptible to EBV infection. Donors varied in susceptibility to de novo EBV infection, but susceptible cultures also displayed differences with respect to pathogenesis. The cultures from one donor yielded lytic infection but cells from two other donors were positive for EBV-encoded EBERs and negative for other lytic infection markers. All cultures stained positive for the pseudostratified markers CK7, MUC5AC, α-tubulin in cilia, and the EBV epithelial cell receptor Ephrin receptor A2. To define EBV transcriptional programs by cell type and to elucidate latent/lytic infection-differential changes, we performed single cell RNA-sequencing on one EBV-infected culture that resulted in alignment with many EBV transcripts. EBV transcripts represented a small portion of the total transcriptome (~0.17%). All cell types in the pseudostratified epithelium had detectable EBV transcripts with suprabasal cells showing the highest number of reads aligning to many EBV genes. Several restriction factors (IRF1, MX1, STAT1, C18orf25) known to limit lytic infection were expressed at lower levels in the lytic subcluster. A third of the differentially-expressed genes in NPC tumors compared to an uninfected pseudostratified ALI culture overlapped with the differentially-expressed genes in the latent subcluster. A third of these commonly perturbed genes were specific to EBV infection and changed in the same direction. Collectively, these findings suggest that the pseudostratified epithelium could harbor EBV infection and that the pseudostratified infection model mirrors many of the transcriptional changes imposed by EBV infection in NPC. It has been known for over 50 years that EBV infection is associated with NPC. Despite many advances from studies in 2-dimensional cell culture, many aspects of EBV molecular pathogenesis in the nasopharynx remain undefined because the cell types and the biology of the nasopharyngeal epithelium can only be faithfully captured in 3-dimensional cell culture. In the stratified epithelium, cellular differentiation triggers lytic infection but it is not clear to what degree the pseudostratified epithelium is involved. The pseudostratified epithelium is abundant in the lateral wall where the lymphoid-rich fossa of Rosenmüller is located and is a site where NPC tumors most often arises. While the oral epithelium is a site of EBV replication, whether the nasopharyngeal epithelium is a major source of EBV shedding in the nasopharynx is not well defined. Here, we present a 3-dimensional organoid model of the nasopharyngeal pseudostratified epithelium showing that such cells can be infected with EBV in some donor cultures, with examples of both latent and lytic infection. We propose that the cell types of the pseudostratified epithelium should be considered a component of EBV pathogenesis in the nasopharynx and that the difference in donor susceptibility and latent/lytic infection could influence EBV’s fitness in the nasopharynx.
Collapse
Affiliation(s)
- Phillip Ziegler
- Cancer Virology Program, University of Pittsburgh Medical Center (UPMC), University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Yarong Tian
- Wallenberg Centre for Molecular and Translational Medicine, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Yulong Bai
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Sanna Abrahamsson
- Wallenberg Centre for Molecular and Translational Medicine, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alan Bäckerholm
- Wallenberg Centre for Molecular and Translational Medicine, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alex S. Reznik
- Cancer Virology Program, University of Pittsburgh Medical Center (UPMC), University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Anthony Green
- University of Pittsburgh Research Histology Services, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - John A. Moore
- UPMC Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Stella E. Lee
- UPMC Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Michael M. Myerburg
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Hyun Jung Park
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Ka-Wei Tang
- Wallenberg Centre for Molecular and Translational Medicine, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Microbiology, Gothenburg, Sweden
| | - Kathy Ho Yen Shair
- Cancer Virology Program, University of Pittsburgh Medical Center (UPMC), University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
40
|
Simintiras CA, Dhakal P, Ranjit C, Fitzgerald HC, Balboula AZ, Spencer TE. Capture and metabolomic analysis of the human endometrial epithelial organoid secretome. Proc Natl Acad Sci U S A 2021; 118:e2026804118. [PMID: 33876774 PMCID: PMC8053979 DOI: 10.1073/pnas.2026804118] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Suboptimal uterine fluid (UF) composition can lead to pregnancy loss and likely contributes to offspring susceptibility to chronic adult-onset disorders. However, our understanding of the biochemical composition and mechanisms underpinning UF formation and regulation remain elusive, particularly in humans. To address this challenge, we developed a high-throughput method for intraorganoid fluid (IOF) isolation from human endometrial epithelial organoids. The IOF is biochemically distinct to the extraorganoid fluid (EOF) and cell culture medium as evidenced by the exclusive presence of 17 metabolites in IOF. Similarly, 69 metabolites were unique to EOF, showing asymmetrical apical and basolateral secretion by the in vitro endometrial epithelium, in a manner resembling that observed in vivo. Contrasting the quantitative metabolomic profiles of IOF and EOF revealed donor-specific biochemical signatures of organoids. Subsequent RNA sequencing of these organoids from which IOF and EOF were derived established the capacity to readily perform organoid multiomics in tandem, and suggests that transcriptomic regulation underpins the observed secretory asymmetry. In summary, these data provided by modeling uterine luminal and basolateral fluid formation in vitro offer scope to better understand UF composition and regulation with potential impacts on female fertility and offspring well-being.
Collapse
Affiliation(s)
| | - Pramod Dhakal
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | - Chaman Ranjit
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | | | - Ahmed Z Balboula
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211
| | - Thomas E Spencer
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211;
- Department of Obstetrics, Gynecology, and Women's Health, University of Missouri, Columbia, MO 65201
| |
Collapse
|
41
|
Using the Air-Liquid Interface Approach to Foster Apical-Basal Polarization of Mammalian Female Reproductive Tract Epithelia In Vitro. Methods Mol Biol 2021. [PMID: 33604859 DOI: 10.1007/978-1-0716-1246-0_18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
Oviduct and uterus are key female reproductive organs lined by ciliated simple columnar epithelia, which are the first line of maternal contact with gametes and the developing embryo during reproduction and which warrant the optimal developmental environment for the conceptus. A major challenge for modeling these epithelia in vitro is the preservation of apical-basal polarization and cilia formation. The air-liquid interface (ALI) culture approach is a technology originally invented for modeling epidermal and airway epithelia. It has recently been shown that it also allows the establishment of highly differentiated in vitro models of epithelia that do not have access to ambient air in vivo. In this chapter, we present a comprehensive ALI procedure to model female reproductive tract (FRT) epithelia of different mammalian species in vitro over extended time periods. As a working example, the protocol focuses on primary oviductal epithelial cells (OEC) isolated from domestic pig. Hints on protocol variations for the culture of OEC from other species are provided in the Subheading 4.
Collapse
|
42
|
Hayden PJ, Harbell JW. Special review series on 3D organotypic culture models: Introduction and historical perspective. In Vitro Cell Dev Biol Anim 2021; 57:95-103. [PMID: 33237402 PMCID: PMC7687207 DOI: 10.1007/s11626-020-00500-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 08/23/2020] [Indexed: 01/23/2023]
Abstract
Three dimensional (3D) organ-like (organotypic) culture models are a rapidly advancing area of in vitro biological science. In contrast to monolayer cell culture methods which were developed to achieve proliferation of animal cells in the beginning of in vitro biology, the advancements in 3D culture methods are designed to promote cellular differentiation, and to achieve in vivo-like 3D structure and organotypic functions. This project was conceived through the Society for In Vitro Biology to draw on the expertise of individual scientists with special expertise in organotypic cultures of selected tissues or associated interrogation methods to prepare individual-focused reviews in this series. This introductory manuscript will review the early achievements of animal cell culture in monolayer culture and the limitations of that approach to reproduce functioning organ systems. Among these are the nature and 3D architecture of the substrate on which or in which the cells are grown, physical and mechanical clues from the substrate, cell-cell interactions, and defined biochemical factors that trigger the induction of the 3D organotypic differentiation. The organoid culture requires a source of cells with proliferative capacity (ranging from tissue-derived stem or immortalized cells to the iPSC cultures), a suitable substrate or matrix with the mechanical and stimulatory properties appropriate for the organotypic construct and the necessary stimulation of the culture to drive differentiation of the cell population to form the functioning organotypic construct. Details for each type of organotypic construct will be provided in the following papers.
Collapse
Affiliation(s)
| | - John W. Harbell
- JHarbell Consulting LLC, 16334 Sunset Valley Drive, Dallas, TX 75248 USA
| |
Collapse
|
43
|
Peña-Zanoni M, García DC, Roldán-Olarte M, Valdecantos PA. A long-term in vitro culture of bovine epithelial cells on collagen rafts. Reprod Domest Anim 2021; 56:684-687. [PMID: 33458879 DOI: 10.1111/rda.13896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In the present work, we established and characterized a 3D functional polarized primary bovine oviduct epithelial cells (BOECs) culture on free-floating type I collagen hydrogels (rafts) at an air-liquid interface (ALI). Intercellular junctions, ultrastructural cellular morphology and the expression of the OVGP1 closely recapitulated those of the in vivo epithelium lining. These morphological and physiological epithelial cell features were maintained under standard DMEM/F12 with 10% foetal bovine serum culture medium for at least 28 days of ALI culture. The versatility of the BOECs raft cultures should allow testing of toxicity compounds, in vitro evaluation of physiological or pathological oviductal states, and the study of epithelial-mesenchymal interactions that are critical for the maintenance of oviductal homeostasis.
Collapse
Affiliation(s)
- Milagros Peña-Zanoni
- Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, UNT, Chacabuco 461, San Miguel de Tucumán, Tucumán, T4000ILI, Argentina
| | - Daniela Celeste García
- Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, UNT, Chacabuco 461, San Miguel de Tucumán, Tucumán, T4000ILI, Argentina.,Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, Chacabuco 461, San Miguel de Tucumán, Tucumán, T4000ILI, Argentina
| | - Mariela Roldán-Olarte
- Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, UNT, Chacabuco 461, San Miguel de Tucumán, Tucumán, T4000ILI, Argentina.,Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, Chacabuco 461, San Miguel de Tucumán, Tucumán, T4000ILI, Argentina
| | - Pablo Alberto Valdecantos
- Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, UNT, Chacabuco 461, San Miguel de Tucumán, Tucumán, T4000ILI, Argentina
| |
Collapse
|
44
|
Replication of Severe Acute Respiratory Syndrome Coronavirus 2 in Human Respiratory Epithelium. J Virol 2020; 94:JVI.00957-20. [PMID: 32434888 PMCID: PMC7375387 DOI: 10.1128/jvi.00957-20] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged by the end of 2019 and rapidly spread in 2020. At present, it is of utmost importance to understand the biology of the virus, rapidly assess the treatment potential of existing drugs, and develop new active compounds. While some animal models for such studies are under development, most of the research is carried out in Vero E6 cells. Here, we propose fully differentiated human airway epithelium cultures as a model for studies on SARS-CoV-2. Currently, there are four seasonal coronaviruses associated with relatively mild respiratory tract disease in humans. However, there is also a plethora of animal coronaviruses which have the potential to cross the species border. This regularly results in the emergence of new viruses in humans. In 2002, severe acute respiratory syndrome coronavirus (SARS-CoV) emerged and rapidly disappeared in May 2003. In 2012, Middle East respiratory syndrome coronavirus (MERS-CoV) was identified as a possible threat to humans, but its pandemic potential so far is minimal, as human-to-human transmission is ineffective. The end of 2019 brought us information about severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emergence, and the virus rapidly spread in 2020, causing an unprecedented pandemic. At present, studies on the virus are carried out using a surrogate system based on the immortalized simian Vero E6 cell line. This model is convenient for diagnostics, but it has serious limitations and does not allow for understanding of the biology and evolution of the virus. Here, we show that fully differentiated human airway epithelium cultures constitute an excellent model to study infection with the novel human coronavirus SARS-CoV-2. We observed efficient replication of the virus in the tissue, with maximal replication at 2 days postinfection. The virus replicated in ciliated cells and was released apically. IMPORTANCE Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged by the end of 2019 and rapidly spread in 2020. At present, it is of utmost importance to understand the biology of the virus, rapidly assess the treatment potential of existing drugs, and develop new active compounds. While some animal models for such studies are under development, most of the research is carried out in Vero E6 cells. Here, we propose fully differentiated human airway epithelium cultures as a model for studies on SARS-CoV-2.
Collapse
|
45
|
Interaction of particles with mucosae and cell membranes. Colloids Surf B Biointerfaces 2020; 186:110657. [DOI: 10.1016/j.colsurfb.2019.110657] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 01/15/2023]
|
46
|
Challenges in studying preimplantation embryo-maternal interaction in cattle. Theriogenology 2020; 150:139-149. [PMID: 31973965 DOI: 10.1016/j.theriogenology.2020.01.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 01/11/2020] [Indexed: 01/10/2023]
Abstract
A comprehensive understanding of the complex embryo-maternal interactions during the preimplantation period requires the analysis of the very early stages of pregnancy encompassing early embryonic development, maternal recognition and the events leading to implantation. Despite the fact that embryo development until blastocyst stage is somewhat autonomous (i.e., does not require contact with the maternal reproductive tract and can be successfully recapitulated in vitro), many studies on ruminant embryo production have focused on the fundamental question of why: (i) only 30%-40% of immature oocytes develop to the blastocyst stage and (ii) the quality of such blastocysts continually lags behind that of blastocysts produced in vivo. Clear evidence indicates that in vitro culture conditions are far from optimal with deficiencies being manifested in short- and long-term effects on the embryo. Thus, enhanced knowledge of mechanisms controlling embryo-maternal interactions would allow the design of novel strategies to improve in vitro embryo conditions and reproductive outcomes in cattle.
Collapse
|