1
|
Gordhan BG, Liebenberg D, Scarlatti G, Herrera C, Chiodi F, Martinson N, Fox J, Kana BD. Ex vivo challenge models for infectious diseases. Crit Rev Microbiol 2024; 50:785-804. [PMID: 37909097 DOI: 10.1080/1040841x.2023.2274855] [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: 02/03/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 11/02/2023]
Abstract
Traditionally, molecular mechanisms of pathogenesis for infectious agents were studied in cell culture or animal models but have limitations on the extent to which the resulting data reflect natural infection in humans. The COVID-19 pandemic has highlighted the urgent need to rapidly develop laboratory models that enable the study of host-pathogen interactions, particularly the relative efficacy of preventive measures. Recently, human and animal ex vivo tissue challenge models have emerged as a promising avenue to study immune responses, screen potential therapies and triage vaccine candidates. This approach offers the opportunity to closely approximate human disease from the perspective of pathology and immune response. It has advantages compared to animal models which are expensive, lengthy and often require containment facilities. Herein, we summarize some recent advances in the development of ex vivo tissue challenge models for COVID-19, HIV-1 and other pathogens. We focus on the contribution of these models to enhancing knowledge of host-pathogen interactions, immune modulation, and their value in testing therapeutic agents. We further highlight the advantages and limitations of using ex vivo challenge models and briefly summarize how the use of organoids provides a useful advancement over current approaches. Collectively, these developments have enormous potential for the study of infectious diseases.
Collapse
Affiliation(s)
- Bhavna Gowan Gordhan
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, University of the Witwatersrand, National Health Laboratory Service, Johannesburg, South Africa
| | - Dale Liebenberg
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, University of the Witwatersrand, National Health Laboratory Service, Johannesburg, South Africa
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Carolina Herrera
- Department of Infectious Disease, Imperial College London, London, UK
| | - Francesca Chiodi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Neil Martinson
- Perinatal HIV Research Unit (PHRU), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Julie Fox
- Guys and St. Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Bavesh Davandra Kana
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, University of the Witwatersrand, National Health Laboratory Service, Johannesburg, South Africa
| |
Collapse
|
2
|
Zhu W, Obara H. Flow structure of okra mucilage in rotating wall vessel system. Heliyon 2024; 10:e36149. [PMID: 39262968 PMCID: PMC11388502 DOI: 10.1016/j.heliyon.2024.e36149] [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: 09/06/2023] [Revised: 08/07/2024] [Accepted: 08/10/2024] [Indexed: 09/13/2024] Open
Abstract
The rotating-wall vessel (RWV) bioreactor, a 3D suspension culture system, faces challenges related to non-uniform tissue growth during the incubation of bone and heart tissues. Okra mucilage, an extract from okra pods with non-Newtonian rheological properties, has shown potential as a plasma replacement agent and has no induced cytotoxic effects. In this study, we investigated the flow structure of okra mucilage in rotating wall vessel system. By modifying the RWV and adding okra mucilage, we analyzed the flow structure using a high-speed camera and particle image velocimetry (PIV). Our results showed that okra mucilage creates a concentric circle-shaped rigid-like rotation at all rotation speeds (1-50 rpm). The high viscosity of okra mucilage resulted in a low terminal velocity for microparticles and quick response to rotational movements. These findings suggest that okra mucilage has the potential to enhance the uniformity of tissue growth in RWV systems by stabilizing the flow structure and reducing microparticle sedimentation.
Collapse
Affiliation(s)
- Weijun Zhu
- Tokyo Metropolitan University, Minamiosaw 1-1, Hachioji 192-0397, Tokyo, Japan
| | - Hiromichi Obara
- Tokyo Metropolitan University, Minamiosaw 1-1, Hachioji 192-0397, Tokyo, Japan
| |
Collapse
|
3
|
Raos D, Vučemilo Paripović N, Ozretić P, Sabol M. Current status of in vitro models for rare gynaecological cancer research. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024:108549. [PMID: 39048342 DOI: 10.1016/j.ejso.2024.108549] [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: 06/24/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024]
Abstract
Gynaecological cancers originate within the female reproductive system and are classified according to the site in the reproductive system where they arise. However, over 50 % of these malignancies are categorized as rare, encompassing 30 distinct histological subtypes, which complicates their diagnosis and treatment. The focus of this review is to give an overview of established in vitro models for the investigation of rare gynaecological cancers, as well as an overview of available online databases that contain detailed descriptions of cell line characteristics. Cell lines represent the main models for the research of carcinogenesis, drug resistance, pharmacodynamics and novel therapy treatment options. Nowadays, classic 2D cell models are increasingly being replaced with 3D cell models, such as spheroids, organoids, and tumoroids because they provide a more accurate representation of numerous tumour characteristics, and their response to therapy differs from the response of adherent cell lines. It is crucial to use the correct cell line model, as rare tumour types can show characteristics that differ from the most common tumour types and can therefore respond unexpectedly to classic treatment. Additionally, some cell lines have been misclassified or misidentified, which could lead to false results. Even though rare gynaecological cancers are rare, this review will demonstrate that there are available options for investigation of such cancers in vitro on biologically relevant models.
Collapse
Affiliation(s)
- Dora Raos
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička Cesta 54, 10 000, Zagreb, Croatia.
| | | | - Petar Ozretić
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička Cesta 54, 10 000, Zagreb, Croatia.
| | - Maja Sabol
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička Cesta 54, 10 000, Zagreb, Croatia.
| |
Collapse
|
4
|
Silva B, Marques EF, Gomes AC. Recent advances in in vitro models simulating the female genital tract toward more effective intravaginal therapeutic delivery. Expert Opin Drug Deliv 2024; 21:1007-1027. [PMID: 39001669 DOI: 10.1080/17425247.2024.2380338] [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: 02/21/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024]
Abstract
INTRODUCTION Intravaginal drug delivery has emerged as a promising avenue for treating a spectrum of systemic and local female genital tract (FGT) conditions, using biomaterials as carriers or scaffolds for targeted and efficient administration. Much effort has been made to understand the natural barriers of this route and improve the delivery system to achieve an efficient therapeutic response. AREAS COVERED In this review, we conducted a comprehensive literature search using multiple databases (PubMed Scopus Web of Science Google Scholar), to discuss the potential of intravaginal therapeutic delivery, as well as the obstacles unique to this route. The in vitro cell models of the FGT and how they can be applied to probing intravaginal drug delivery are then analyzed. We further explore the limitations of the existing models and the possibilities to make them more promising for delivery studies or biomaterial validation. Complementary information is provided by in vitro acellular techniques that may shed light on mucus-drug interaction. EXPERT OPINION Advances in 3D models and cell cultures have enhanced our understanding of the FGT, but they still fail to replicate all variables. Future research should aim to use complementary methods, ensure stability, and develop consistent protocols to improve therapy evaluation and create better predictive in vitro models for women's health.
Collapse
Affiliation(s)
- Bruna Silva
- CBMA (Centre of Molecular and Environmental Biology), Department of Biology, Campus of Gualtar, University of Minho, Braga, Portugal
- CIQUP, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Eduardo F Marques
- CIQUP, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Andreia C Gomes
- CBMA (Centre of Molecular and Environmental Biology), Department of Biology, Campus of Gualtar, University of Minho, Braga, Portugal
| |
Collapse
|
5
|
Ayehunie S, Landry T, Armento A. Vaginal irritation testing-prospects of human organotypic vaginal tissue culture models. In Vitro Cell Dev Biol Anim 2024; 60:569-582. [PMID: 38995526 DOI: 10.1007/s11626-024-00907-1] [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: 09/05/2023] [Accepted: 04/01/2024] [Indexed: 07/13/2024]
Abstract
Personal lubricants intended for local or systemic delivery via the vaginal route can induce vaginal irritation, damage the vaginal epithelial barrier which can enhance microbial entry, induce inflammation, and alter the microbiome of the vaginal ecosystem. Therefore, manufacturers of personal lubricants and medical devices are required to show biocompatibility and safety assessment data to support regulatory decision-making within a specified context of use. Furthermore, due to ethical concerns and the introduction of the 7th amendment of the European Council Directive which bans animal testing for cosmetic ingredients and products coupled with the Food and Drug Administration modernization Act 2.0 guidelines, there is a wave of drive to develop alternative test methods to predict human responses to chemical or formulation exposure. In this framework, there is a potential to use three-dimensional organotypic human vaginal-ectocervical tissue models as a screening tool to predict the vaginal irritation potential of personal lubricants and medicaments. To be physiologically relevant, the in vitro tissue models need to be reconstructed using primary epithelial cells of the specific organ or tissue and produce organ-like structure and functionality that recapitulate the in vivo-like responses. Through the years, progress has been made and vaginal tissue models are manufactured under controlled conditions with a specified performance criterion, which leads to a high level of reproducibility and reliability. The utility of vaginal tissue models has been accelerated in the last 20 years with an expanded portfolio of applications ranging from toxicity, inflammation, infection to drug safety, and efficacy studies. This article provides an overview of the state of the art of diversified applications of reconstructed vaginal tissue models and highlights their utility as a tool to predict vaginal irritation potential of feminine care products.
Collapse
Affiliation(s)
- Seyoum Ayehunie
- MatTek Corporation, 200 Homer Avenue, Ashland, MA, 01721, USA.
| | - Timothy Landry
- MatTek Corporation, 200 Homer Avenue, Ashland, MA, 01721, USA
| | - Alex Armento
- MatTek Corporation, 200 Homer Avenue, Ashland, MA, 01721, USA
| |
Collapse
|
6
|
Jackson R, Rajadhyaksha EV, Loeffler RS, Flores CE, Van Doorslaer K. Characterization of 3D organotypic epithelial tissues reveals tonsil-specific differences in tonic interferon signaling. PLoS One 2023; 18:e0292368. [PMID: 37792852 PMCID: PMC10550192 DOI: 10.1371/journal.pone.0292368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/18/2023] [Indexed: 10/06/2023] Open
Abstract
Three-dimensional (3D) culturing techniques can recapitulate the stratified nature of multicellular epithelial tissues. Organotypic 3D epithelial tissue culture methods have several applications, including the study of tissue development and function, drug discovery and toxicity testing, host-pathogen interactions, and the development of tissue-engineered constructs for use in regenerative medicine. We grew 3D organotypic epithelial tissues from foreskin, cervix, and tonsil-derived primary cells and characterized the transcriptome of these in vitro tissue equivalents. Using the same 3D culturing method, all three tissues yielded stratified squamous epithelium, validated histologically using basal and superficial epithelial cell markers. The goal of this study was to use RNA-seq to compare gene expression patterns in these three types of epithelial tissues to gain a better understanding of the molecular mechanisms underlying their function and identify potential therapeutic targets for various diseases. Functional profiling by over-representation and gene set enrichment analysis revealed tissue-specific differences: i.e., cutaneous homeostasis and lipid metabolism in foreskin, extracellular matrix remodeling in cervix, and baseline innate immune differences in tonsil. Specifically, tonsillar epithelia may play an active role in shaping the immune microenvironment of the tonsil balancing inflammation and immune responses in the face of constant exposure to microbial insults. Overall, these data serve as a resource, with gene sets made available for the research community to explore, and as a foundation for understanding the epithelial heterogeneity and how it may impact their in vitro use. An online resource is available to investigate these data (https://viz.datascience.arizona.edu/3DEpiEx/).
Collapse
Affiliation(s)
- Robert Jackson
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, Arizona, United States of America
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
| | - Esha V. Rajadhyaksha
- College of Medicine and College of Science, University of Arizona, Tucson, Arizona, United States of America
| | - Reid S. Loeffler
- Biosystems Engineering, College of Agriculture and Life Sciences, College of Engineering, University of Arizona, Tucson, Arizona, United States of America
| | - Caitlyn E. Flores
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, Arizona, United States of America
| | - Koenraad Van Doorslaer
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, Arizona, United States of America
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
- Department of Immunobiology, Cancer Biology Graduate Interdisciplinary Program, Genetics Graduate Interdisciplinary Program, and University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
7
|
Ren Z, Harriot AD, Mair DB, Chung MK, Lee PHU, Kim DH. Biomanufacturing of 3D Tissue Constructs in Microgravity and their Applications in Human Pathophysiological Studies. Adv Healthc Mater 2023; 12:e2300157. [PMID: 37483106 DOI: 10.1002/adhm.202300157] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/27/2023] [Indexed: 07/25/2023]
Abstract
The growing interest in bioengineering in-vivo-like 3D functional tissues has led to novel approaches to the biomanufacturing process as well as expanded applications for these unique tissue constructs. Microgravity, as seen in spaceflight, is a unique environment that may be beneficial to the tissue-engineering process but cannot be completely replicated on Earth. Additionally, the expense and practical challenges of conducting human and animal research in space make bioengineered microphysiological systems an attractive research model. In this review, published research that exploits real and simulated microgravity to improve the biomanufacturing of a wide range of tissue types as well as those studies that use microphysiological systems, such as organ/tissue chips and multicellular organoids, for modeling human diseases in space are summarized. This review discusses real and simulated microgravity platforms and applications in tissue-engineered microphysiological systems across three topics: 1) application of microgravity to improve the biomanufacturing of tissue constructs, 2) use of tissue constructs fabricated in microgravity as models for human diseases on Earth, and 3) investigating the effects of microgravity on human tissues using biofabricated in vitro models. These current achievements represent important progress in understanding the physiological effects of microgravity and exploiting their advantages for tissue biomanufacturing.
Collapse
Affiliation(s)
- Zhanping Ren
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Anicca D Harriot
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Devin B Mair
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | | | - Peter H U Lee
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
- Department of Cardiothoracic Surgery, Southcoast Health, Fall River, MA, 02720, USA
| | - Deok-Ho Kim
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, MD, 21205, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, 21218, USA
| |
Collapse
|
8
|
Yang Q, Li M, Yang X, Xiao Z, Tong X, Tuerdi A, Li S, Lei L. Flourishing tumor organoids: History, emerging technology, and application. Bioeng Transl Med 2023; 8:e10559. [PMID: 37693042 PMCID: PMC10487342 DOI: 10.1002/btm2.10559] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/16/2023] [Accepted: 05/25/2023] [Indexed: 09/12/2023] Open
Abstract
Malignant tumors are one of the leading causes of death which impose an increasingly heavy burden on all countries. Therefore, the establishment of research models that closely resemble original tumor characteristics is crucial to further understanding the mechanisms of malignant tumor development, developing safer and more effective drugs, and formulating personalized treatment plans. Recently, organoids have been widely used in tumor research owing to their advantages including preserving the structure, heterogeneity, and cellular functions of the original tumor, together with the ease of manipulation. This review describes the history and characteristics of tumor organoids and the synergistic combination of three-dimensional (3D) culture approaches for tumor organoids with emerging technologies, including tissue-engineered cell scaffolds, microfluidic devices, 3D bioprinting, rotating wall vessels, and clustered regularly interspaced short palindromic repeats-CRISPR-associated protein 9 (CRISPR-Cas9). Additionally, the progress in research and the applications in basic and clinical research of tumor organoid models are summarized. This includes studies of the mechanism of tumor development, drug development and screening, precision medicine, immunotherapy, and simulation of the tumor microenvironment. Finally, the existing shortcomings of tumor organoids and possible future directions are discussed.
Collapse
Affiliation(s)
- Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Mengmeng Li
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xinming Yang
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Zian Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xinying Tong
- Department of Hemodialysis, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Ayinuer Tuerdi
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Lanjie Lei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| |
Collapse
|
9
|
Pontecorvi P, Ceccarelli S, Cece F, Camero S, Lotti LV, Niccolai E, Nannini G, Gerini G, Anastasiadou E, Scialis ES, Romano E, Venneri MA, Amedei A, Angeloni A, Megiorni F, Marchese C. Assessing the Impact of Polyethylene Nano/Microplastic Exposure on Human Vaginal Keratinocytes. Int J Mol Sci 2023; 24:11379. [PMID: 37511139 PMCID: PMC10380279 DOI: 10.3390/ijms241411379] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/24/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
The global rise of single-use throw-away plastic products has elicited a massive increase in the nano/microplastics (N/MPLs) exposure burden in humans. Recently, it has been demonstrated that disposable period products may release N/MPLs with usage, which represents a potential threat to women's health which has not been scientifically addressed yet. By using polyethyl ene (PE) particles (200 nm to 9 μm), we showed that acute exposure to a high concentration of N/MPLs induced cell toxicity in vaginal keratinocytes after effective cellular uptake, as viability and apoptosis data suggest, along with transmission electron microscopy (TEM) observations. The internalised N/MPLs altered the expression of junctional and adherence proteins and the organisation of the actin cortex, influencing the level of genes involved in oxidative stress signalling pathways and that of miRNAs related to epithelial barrier function. When the exposure to PE N/MPLs was discontinued or became chronic, cells were able to recover from the negative effects on viability and differentiation/proliferation gene expression in a few days. However, in all cases, PE N/MPL exposure prompted a sustained alteration of DNA methyltransferase and DNA demethylase expression, which might impact epigenetic regulation processes, leading to accelerated cell ageing and inflammation, or the occurrence of malignant transformation.
Collapse
Affiliation(s)
- Paola Pontecorvi
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (F.C.); (S.C.); (L.V.L.); (G.G.); (M.A.V.); (A.A.); (F.M.)
| | - Simona Ceccarelli
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (F.C.); (S.C.); (L.V.L.); (G.G.); (M.A.V.); (A.A.); (F.M.)
| | - Fabrizio Cece
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (F.C.); (S.C.); (L.V.L.); (G.G.); (M.A.V.); (A.A.); (F.M.)
| | - Simona Camero
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (F.C.); (S.C.); (L.V.L.); (G.G.); (M.A.V.); (A.A.); (F.M.)
| | - Lavinia Vittoria Lotti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (F.C.); (S.C.); (L.V.L.); (G.G.); (M.A.V.); (A.A.); (F.M.)
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (E.N.); (G.N.); (A.A.)
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (E.N.); (G.N.); (A.A.)
| | - Giulia Gerini
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (F.C.); (S.C.); (L.V.L.); (G.G.); (M.A.V.); (A.A.); (F.M.)
| | - Eleni Anastasiadou
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035, 00189 Rome, Italy;
| | - Elena Sofia Scialis
- Department of Innovative Technologies in Medicine and Dentistry, University “G. D’Annunzio” Chieti—Pescara, Via dei Vestini 31, 66100 Chieti, Italy;
| | - Enrico Romano
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy;
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (F.C.); (S.C.); (L.V.L.); (G.G.); (M.A.V.); (A.A.); (F.M.)
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (E.N.); (G.N.); (A.A.)
| | - Antonio Angeloni
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (F.C.); (S.C.); (L.V.L.); (G.G.); (M.A.V.); (A.A.); (F.M.)
| | - Francesca Megiorni
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (F.C.); (S.C.); (L.V.L.); (G.G.); (M.A.V.); (A.A.); (F.M.)
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (F.C.); (S.C.); (L.V.L.); (G.G.); (M.A.V.); (A.A.); (F.M.)
| |
Collapse
|
10
|
Jimenez NR, Maarsingh JD, Łaniewski P, Herbst-Kralovetz MM. Commensal Lactobacilli Metabolically Contribute to Cervical Epithelial Homeostasis in a Species-Specific Manner. mSphere 2023; 8:e0045222. [PMID: 36629413 PMCID: PMC9942568 DOI: 10.1128/msphere.00452-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/09/2022] [Indexed: 01/12/2023] Open
Abstract
In reproductive-age women, the vaginal microbiome is typically dominated by one or a few Lactobacillus species, including Lactobacillus crispatus, Lactobacillus iners, Lactobacillus paragasseri, Lactobacillus mulieris, and Lactobaccillus crispatus, has been associated with optimal cervicovaginal health; however, much is still unknown about how other lactobacilli metabolically contribute to cervicovaginal health. We hypothesized that metabolites of each Lactobacillus species differ and uniquely contribute to health and homeostasis. To address this hypothesis, we utilized a human three-dimensional (3D) cervical epithelial cell model in conjunction with genomics analyses and untargeted metabolomics to determine the metabolic contributions of less-studied vaginal lactobacilli-L. iners, L. paragasseri, and L. mulieris. Our study validated that vaginal lactobacilli exhibit a close phylogenetic relationship. Genomic findings from publicly available strains and those used in our study indicated that L. iners is metabolically distinct from other species of lactobacilli, likely due to a reduced genome size. Lactobacilli and mock controls were distinguishable based on global metabolic profiles. We identified 95 significantly altered metabolites (P < 0.05) between individual lactobacilli and mock controls. Metabolites related to amino acid metabolism were shared among the lactobacilli. N-Acetylated amino acids with potential antimicrobial properties were significantly elevated in a species-specific manner. L. paragasseri and L. iners shared aromatic, but not carbohydrate-derived, lactic acid metabolites with potential antimicrobial properties that may contribute to homeostasis of the cervicovaginal environment. Additionally, L. iners uniquely altered lipid metabolism, which may be a sign of adaptation to the cervicovaginal niche. Overall, these findings further elucidate the metabolic contributions of three key vaginal Lactobacillus species in gynecological health. IMPORTANCE Lactobacillus species contribute to cervicovaginal health by their production of lactic acid and other antimicrobial compounds. Yet, much is still unknown regarding the metabolic potential of lesser-studied but common vaginal lactobacilli. Here, we used untargeted metabolomics coupled with our 3D cervical epithelial cell model to identify metabolic differences among vaginal Lactobacillus species (Lactobacillus iners, Lactobacillus paragasseri, and Lactobacillus mulieris) and how those differences related to maintaining homeostasis of the cervical epithelium. Human 3D cell models are essential tools for studying host-bacteria interactions and reducing confounding factors inherent in clinical studies. Therefore, these unique models allowed us to decipher the putative lactobacilli mechanisms that contribute to their roles in health or disease. Metabolic analyses revealed distinct profiles of each Lactobacillus species but also shared metabolic contributions associated with antimicrobial activity: amino acid metabolism, N-acetylated amino acids, and aromatic lactic acids. These patterns provided validation of metabolites associated with health in clinical studies and provided novel targets, including immunomodulatory and antimicrobial metabolites, for postbiotic therapies.
Collapse
Affiliation(s)
- Nicole R. Jimenez
- Department of Obstetrics and Gynecology, College of Medicine—Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Jason D. Maarsingh
- Department of Obstetrics and Gynecology, College of Medicine—Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Paweł Łaniewski
- Department of Basic Medical Sciences, College of Medicine—Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Melissa M. Herbst-Kralovetz
- Department of Obstetrics and Gynecology, College of Medicine—Phoenix, University of Arizona, Phoenix, Arizona, USA
- Department of Basic Medical Sciences, College of Medicine—Phoenix, University of Arizona, Phoenix, Arizona, USA
| |
Collapse
|
11
|
Jackson R, Rajadhyaksha EV, Loeffler RS, Flores CE, Van Doorslaer K. Characterization of 3D organotypic epithelial tissues reveals tonsil-specific differences in tonic interferon signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.19.524743. [PMID: 36711548 PMCID: PMC9882319 DOI: 10.1101/2023.01.19.524743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Three-dimensional (3D) culturing techniques can recapitulate the stratified nature of multicellular epithelial tissues. Organotypic 3D epithelial tissue culture methods have several applications, including the study of tissue development and function, drug discovery and toxicity testing, host-pathogen interactions, and the development of tissue-engineered constructs for use in regenerative medicine. We grew 3D organotypic epithelial tissues from foreskin, cervix, and tonsil-derived primary cells and characterized the transcriptome of these in vitro tissue equivalents. Using the same 3D culturing method, all three tissues yielded stratified squamous epithelium, validated histologically using basal and superficial epithelial cell markers. The goal of this study was to use RNA-seq to compare gene expression patterns in these three types of epithelial tissues to gain a better understanding of the molecular mechanisms underlying their function and identify potential therapeutic targets for various diseases. Functional profiling by over-representation and gene set enrichment analysis revealed tissue-specific differences: i.e. , cutaneous homeostasis and lipid metabolism in foreskin, extracellular matrix remodeling in cervix, and baseline innate immune differences in tonsil. Specifically, tonsillar epithelia may play an active role in shaping the immune microenvironment of the tonsil balancing inflammation and immune responses in the face of constant exposure to microbial insults. Overall, these data serve as a resource, with gene sets made available for the research community to explore, and as a foundation for understanding the epithelial heterogeneity and how it may impact their in vitro use. An online resource is available to investigate these data ( https://viz.datascience.arizona.edu/3DEpiEx/ ).
Collapse
Affiliation(s)
- Robert Jackson
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
| | - Esha V Rajadhyaksha
- College of Medicine and College of Science, University of Arizona, Tucson, AZ, USA
| | - Reid S Loeffler
- Biosystems Engineering, College of Agriculture and Life Sciences; College of Engineering, University of Arizona, Tucson, AZ, USA
| | - Caitlyn E Flores
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ, USA
| | - Koenraad Van Doorslaer
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- Department of Immunobiology; Cancer Biology Graduate Interdisciplinary Program; Genetics Graduate Interdisciplinary Program; and University of Arizona Cancer Center, University of Arizona, Tucson, AZ USA
| |
Collapse
|
12
|
Shapiro RL, DeLong K, Zulfiqar F, Carter D, Better M, Ensign LM. In vitro and ex vivo models for evaluating vaginal drug delivery systems. Adv Drug Deliv Rev 2022; 191:114543. [PMID: 36208729 PMCID: PMC9940824 DOI: 10.1016/j.addr.2022.114543] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/26/2022] [Accepted: 09/13/2022] [Indexed: 01/24/2023]
Abstract
Vaginal drug delivery systems are often preferred for treating a variety of diseases and conditions of the female reproductive tract (FRT), as delivery can be more targeted with less systemic side effects. However, there are many anatomical and biological barriers to effective treatment via the vaginal route. Further, biocompatibility with the local tissue and microbial microenvironment is desired. A variety of in vitro and ex vivo models are described herein for evaluating the physicochemical properties and toxicity profile of vaginal drug delivery systems. Deciding whether to utilize organoids in vitro or fresh human cervicovaginal mucus ex vivo requires careful consideration of the intended use and the formulation characteristics. Optimally, in vitro and ex vivo experimentation will inform or predict in vivo performance, and examples are given that describe utilization of a range of methods from in vitro to in vivo. Lastly, we highlight more advanced model systems for other mucosa as inspiration for the future in model development for the FRT.
Collapse
Affiliation(s)
- Rachel L Shapiro
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Chemical & Biomolecular Engineering, Johns Hopkins University, 3400 N Charles St., Baltimore, MD 21218, USA.
| | - Kevin DeLong
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, 1800 Orleans St., Baltimore, MD 21287, USA.
| | - Fareeha Zulfiqar
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, 1800 Orleans St., Baltimore, MD 21287, USA.
| | - Davell Carter
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 725 N Wolfe St., Baltimore, MD 21287, USA.
| | - Marina Better
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 725 N Wolfe St., Baltimore, MD 21287, USA.
| | - Laura M Ensign
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21231, USA; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, 1800 Orleans St., Baltimore, MD 21287, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 725 N Wolfe St., Baltimore, MD 21287, USA; Department of Chemical & Biomolecular Engineering, Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA; Departments of Gynecology and Obstetrics, Infectious Diseases, and Oncology, Johns Hopkins University School of Medicine, 1800 Orleans St., Baltimore, MD 21287, USA; Department of Biomedical Engineering, Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA.
| |
Collapse
|
13
|
Edwards VL, McComb E, Gleghorn JP, Forney L, Bavoil PM, Ravel J. Three-dimensional models of the cervicovaginal epithelia to study host-microbiome interactions and sexually transmitted infections. Pathog Dis 2022; 80:6655985. [PMID: 35927516 PMCID: PMC9419571 DOI: 10.1093/femspd/ftac026] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/14/2022] [Accepted: 08/02/2022] [Indexed: 02/03/2023] Open
Abstract
2D cell culture systems have historically provided controlled, reproducible means to analyze host-pathogen interactions observed in the human reproductive tract. Although inexpensive, straightforward, and requiring a very short time commitment, these models recapitulate neither the functionality of multilayered cell types nor the associated microbiome that occurs in a human. Animal models have commonly been used to recreate the complexity of human infections. However, extensive modifications of animal models are required to recreate interactions that resemble those in the human reproductive tract. 3D cell culture models have emerged as alternative means of reproducing vital elements of human infections at a fraction of the cost of animal models and on a scale that allows for replicative experiments. Here, we describe a new 3D model that utilizes transwells with epithelial cells seeded apically and a basolateral extracellular matrix (ECM)-like layer. The model produced tissues with morphologic and physiological resemblance to human cervical and vaginal epithelia, including mucus levels produced by cervical cells. Infection by Chlamydia trachomatis and Neisseria gonorrhoeae was demonstrated, as well as the growth of bacterial species observed in the human vaginal microbiota. This enabled controlled mechanistic analyses of the interactions between host cells, the vaginal microbiota, and STI pathogens. Affordable and semi high-throughput 3D models of the cervicovaginal epithelia that are physiologically relevant by sustaining vaginal bacterial colonization, and facilitate studies of chlamydial and gonococcal infections.
Collapse
Affiliation(s)
- Vonetta L Edwards
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | | | - Jason P Gleghorn
- Department of Biomedical Engineering, University of Delaware, Newark, DE, United States
| | - Larry Forney
- Department of Biological Sciences, University of Idaho, Moscow, ID, United States
| | - Patrik M Bavoil
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States,Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD, United States
| | - Jacques Ravel
- Corresponding author: Institute for Genome Sciences and Department of Microbiology and Immunology, University of Maryland School of Medicine, Health Science Research Facility (HSRDF), 670 W. Baltimore Street, Baltimore, MD 21201, United States. Tel: +1 410-706-5674; E-mail:
| |
Collapse
|
14
|
Towards a deeper understanding of the vaginal microbiota. Nat Microbiol 2022; 7:367-378. [PMID: 35246662 DOI: 10.1038/s41564-022-01083-2] [Citation(s) in RCA: 143] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 02/03/2022] [Indexed: 12/30/2022]
Abstract
The human vaginal microbiota is a critical determinant of vaginal health. These communities live in close association with the vaginal epithelium and rely on host tissues for resources. Although often dominated by lactobacilli, the vaginal microbiota is also frequently composed of a collection of facultative and obligate anaerobes. The prevalence of these communities with a paucity of Lactobacillus species varies among women, and epidemiological studies have associated them with an increased risk of adverse health outcomes. The mechanisms that drive these associations have yet to be described in detail, with few studies establishing causative relationships. Here, we review our current understanding of the vaginal microbiota and its connection with host health. We centre our discussion around the biology of the vaginal microbiota when Lactobacillus species are dominant versus when they are not, including host factors that are implicated in shaping these microbial communities and the resulting adverse health outcomes. We discuss current approaches to modulate the vaginal microbiota, including probiotics and vaginal microbiome transplants, and argue that new model systems of the cervicovaginal environment that incorporate the vaginal microbiota are needed to progress from association to mechanism and this will prove invaluable for future research.
Collapse
|
15
|
McKenzie R, Maarsingh JD, Łaniewski P, Herbst-Kralovetz MM. Immunometabolic Analysis of Mobiluncus mulieris and Eggerthella sp. Reveals Novel Insights Into Their Pathogenic Contributions to the Hallmarks of Bacterial Vaginosis. Front Cell Infect Microbiol 2022; 11:759697. [PMID: 35004344 PMCID: PMC8733642 DOI: 10.3389/fcimb.2021.759697] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/06/2021] [Indexed: 01/11/2023] Open
Abstract
The cervicovaginal microbiome plays an important role in protecting women from dysbiosis and infection caused by pathogenic microorganisms. In healthy reproductive-age women the cervicovaginal microbiome is predominantly colonized by protective Lactobacillus spp. The loss of these protective bacteria leads to colonization of the cervicovaginal microenvironment by pathogenic microorganisms resulting in dysbiosis and bacterial vaginosis (BV). Mobiluncus mulieris and Eggerthella sp. are two of the many anaerobes that can contribute to BV, a condition associated with multiple adverse obstetric and gynecological outcomes. M. mulieris has been linked to high Nugent scores (relating to BV morphotypes) and preterm birth (PTB), whilst some bacterial members of the Eggerthellaceae family are highly prevalent in BV, and identified in ~85-95% of cases. The functional impact of M. mulieris and Eggerthella sp. in BV is still poorly understood. To determine the individual immunometabolic contributions of Eggerthella sp. and M. mulieris within the cervicovaginal microenvironment, we utilized our well-characterized human three-dimensional (3-D) cervical epithelial cell model in combination with multiplex immunoassays and global untargeted metabolomics approaches to identify key immune mediators and metabolites related to M. mulieris and Eggerthella sp. infections. We found that infection with M. mulieris significantly elevated multiple proinflammatory markers (IL-6, IL-8, TNF-α and MCP-1) and altered metabolites related to energy metabolism (nicotinamide and succinate) and oxidative stress (cysteinylglycine, cysteinylglycine disulfide and 2-hydroxygluatrate). Eggerthella sp. infection significantly elevated multiple sphingolipids and glycerolipids related to epithelial barrier function, and biogenic amines (putrescine and cadaverine) associated with elevated vaginal pH, vaginal amine odor and vaginal discharge. Our study elucidated that M. mulieris elevated multiple proinflammatory markers relating to PTB and STI acquisition, as well as altered energy metabolism and oxidative stress, whilst Eggerthella sp. upregulated multiple biogenic amines associated with the clinical diagnostic criteria of BV. Future studies are needed to evaluate how these bacteria interact with other BV-associated bacteria within the cervicovaginal microenvironment.
Collapse
Affiliation(s)
- Ross McKenzie
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, United States.,Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Jason D Maarsingh
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, United States
| | - Paweł Łaniewski
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, United States
| | - Melissa M Herbst-Kralovetz
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, United States.,Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, United States
| |
Collapse
|
16
|
Plesniarski A, Siddik AB, Su RC. The Microbiome as a Key Regulator of Female Genital Tract Barrier Function. Front Cell Infect Microbiol 2022; 11:790627. [PMID: 34976864 PMCID: PMC8719631 DOI: 10.3389/fcimb.2021.790627] [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] [Received: 10/07/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
The microbiome, the collection of microbial species at a site or compartment, has been an underappreciated realm of human health up until the last decade. Mounting evidence suggests the microbiome has a critical role in regulating the female genital tract (FGT) mucosa's function as a barrier against sexually transmitted infections (STIs) and pathogens. In this review, we provide the most recent experimental systems and studies for analyzing the interplay between the microbiome and host cells and soluble factors with an influence on barrier function. Key components, such as microbial diversity, soluble factors secreted by host and microbe, as well as host immune system, all contribute to both the physical and immunologic aspects of the FGT mucosal barrier. Current gaps in what is known about the effects of the microbiome on FGT mucosal barrier function are compared and contrasted with the literature of the gut and respiratory mucosa. This review article presents evidence supporting that the vaginal microbiome, directly and indirectly, contributes to how well the FGT protects against infection.
Collapse
Affiliation(s)
- Andrew Plesniarski
- JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Abu Bakar Siddik
- JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Ruey-Chyi Su
- JC Wilt Infectious Diseases Research Centre, National Microbiology Laboratories, Public Health Agency of Canada, Winnipeg, MB, Canada.,Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
17
|
Francés-Herrero E, Lopez R, Hellström M, de Miguel-Gómez L, Herraiz S, Brännström M, Pellicer A, Cervelló I. OUP accepted manuscript. Hum Reprod Update 2022; 28:798-837. [PMID: 35652272 PMCID: PMC9629485 DOI: 10.1093/humupd/dmac025] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 04/13/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND To provide the optimal milieu for implantation and fetal development, the female reproductive system must orchestrate uterine dynamics with the appropriate hormones produced by the ovaries. Mature oocytes may be fertilized in the fallopian tubes, and the resulting zygote is transported toward the uterus, where it can implant and continue developing. The cervix acts as a physical barrier to protect the fetus throughout pregnancy, and the vagina acts as a birth canal (involving uterine and cervix mechanisms) and facilitates copulation. Fertility can be compromised by pathologies that affect any of these organs or processes, and therefore, being able to accurately model them or restore their function is of paramount importance in applied and translational research. However, innate differences in human and animal model reproductive tracts, and the static nature of 2D cell/tissue culture techniques, necessitate continued research and development of dynamic and more complex in vitro platforms, ex vivo approaches and in vivo therapies to study and support reproductive biology. To meet this need, bioengineering is propelling the research on female reproduction into a new dimension through a wide range of potential applications and preclinical models, and the burgeoning number and variety of studies makes for a rapidly changing state of the field. OBJECTIVE AND RATIONALE This review aims to summarize the mounting evidence on bioengineering strategies, platforms and therapies currently available and under development in the context of female reproductive medicine, in order to further understand female reproductive biology and provide new options for fertility restoration. Specifically, techniques used in, or for, the uterus (endometrium and myometrium), ovary, fallopian tubes, cervix and vagina will be discussed. SEARCH METHODS A systematic search of full-text articles available in PubMed and Embase databases was conducted to identify relevant studies published between January 2000 and September 2021. The search terms included: bioengineering, reproduction, artificial, biomaterial, microfluidic, bioprinting, organoid, hydrogel, scaffold, uterus, endometrium, ovary, fallopian tubes, oviduct, cervix, vagina, endometriosis, adenomyosis, uterine fibroids, chlamydia, Asherman’s syndrome, intrauterine adhesions, uterine polyps, polycystic ovary syndrome and primary ovarian insufficiency. Additional studies were identified by manually searching the references of the selected articles and of complementary reviews. Eligibility criteria included original, rigorous and accessible peer-reviewed work, published in English, on female reproductive bioengineering techniques in preclinical (in vitro/in vivo/ex vivo) and/or clinical testing phases. OUTCOMES Out of the 10 390 records identified, 312 studies were included for systematic review. Owing to inconsistencies in the study measurements and designs, the findings were assessed qualitatively rather than by meta-analysis. Hydrogels and scaffolds were commonly applied in various bioengineering-related studies of the female reproductive tract. Emerging technologies, such as organoids and bioprinting, offered personalized diagnoses and alternative treatment options, respectively. Promising microfluidic systems combining various bioengineering approaches have also shown translational value. WIDER IMPLICATIONS The complexity of the molecular, endocrine and tissue-level interactions regulating female reproduction present challenges for bioengineering approaches to replace female reproductive organs. However, interdisciplinary work is providing valuable insight into the physicochemical properties necessary for reproductive biological processes to occur. Defining the landscape of reproductive bioengineering technologies currently available and under development for women can provide alternative models for toxicology/drug testing, ex vivo fertility options, clinical therapies and a basis for future organ regeneration studies.
Collapse
Affiliation(s)
| | | | - Mats Hellström
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lucía de Miguel-Gómez
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain
- Fundación IVI, IVI-RMA Global, Valencia, Spain
| | - Sonia Herraiz
- Fundación IVI, IVI-RMA Global, Valencia, Spain
- Reproductive Medicine Research Group, IIS La Fe, Valencia, Spain
| | - Mats Brännström
- Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Stockholm IVF-EUGIN, Stockholm, Sweden
| | - Antonio Pellicer
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain
- IVI Roma Parioli, IVI-RMA Global, Rome, Italy
| | | |
Collapse
|
18
|
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
|
19
|
Xue Y, Seiler MJ, Tang WC, Wang JY, Delgado J, McLelland BT, Nistor G, Keirstead HS, Browne AW. Retinal organoids on-a-chip: a micro-millifluidic bioreactor for long-term organoid maintenance. LAB ON A CHIP 2021; 21:3361-3377. [PMID: 34236056 PMCID: PMC8387452 DOI: 10.1039/d1lc00011j] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Retinal degeneration is a leading cause of vision impairment and blindness worldwide and medical care for advanced disease does not exist. Stem cell-derived retinal organoids (RtOgs) became an emerging tool for tissue replacement therapy. However, existing RtOg production methods are highly heterogeneous. Controlled and predictable methodology and tools are needed to standardize RtOg production and maintenance. In this study, we designed a shear stress-free micro-millifluidic bioreactor for nearly labor-free retinal organoid maintenance. We used a stereolithography (SLA) 3D printer to fabricate a mold from which Polydimethylsiloxane (PDMS) was cast. We optimized the chip design using in silico simulations and in vitro evaluation to optimize mass transfer efficiency and concentration uniformity in each culture chamber. We successfully cultured RtOgs at three different differentiation stages (day 41, 88, and 128) on an optimized bioreactor chip for more than 1 month. We used different quantitative and qualitative techniques to fully characterize the RtOgs produced by static dish culture and bioreactor culture methods. By analyzing the results from phase contrast microscopy, single-cell RNA sequencing (scRNA seq), quantitative polymerase chain reaction (qPCR), immunohistology, and electron microscopy, we found that bioreactor-cultured RtOgs developed cell types and morphology comparable to static cultured ones and exhibited similar retinal genes expression levels. We also evaluated the metabolic activity of RtOgs in both groups using fluorescence lifetime imaging (FLIM), and found that the outer surface region of bioreactor cultured RtOgs had a comparable free/bound NADH ratio and overall lower long lifetime species (LLS) ratio than static cultured RtOgs during imaging. To summarize, we validated an automated micro-millifluidic device with significantly reduced shear stress to produce RtOgs of comparable quality to those maintained in conventional static culture.
Collapse
Affiliation(s)
- Yuntian Xue
- Biomedical Engineering, University of California, Irvine, Irvine, CA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Son GH, Lee JJ, Kim Y, Lee KY. The Role of Antimicrobial Peptides in Preterm Birth. Int J Mol Sci 2021; 22:ijms22168905. [PMID: 34445608 PMCID: PMC8396209 DOI: 10.3390/ijms22168905] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
Antimicrobial peptides (AMPs) are short cationic amphipathic peptides with a wide range of antimicrobial properties and play an important role in the maintenance of immune homeostasis by modulating immune responses in the reproductive tract. As intra-amniotic infection and microbial dysbiosis emerge as common causes of preterm births (PTBs), a better understanding of the AMPs involved in the development of PTB is essential. The altered expression of AMPs has been reported in PTB-related clinical presentations, such as preterm labor, intra-amniotic infection/inflammation, premature rupture of membranes, and cervical insufficiency. Moreover, it was previously reported that dysregulation of AMPs may affect the pregnancy prognosis. This review aims to describe the expression of AMPs associated with PTBs and to provide new perspectives on the role of AMPs in PTB.
Collapse
Affiliation(s)
- Ga-Hyun Son
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Hallym University College of Medicine, Kangnam Sacred Heart Hospital, Seoul 07441, Korea;
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon 24523, Korea; (J.-J.L.); (Y.K.)
- Correspondence: ; Tel.: +82-2-6960-1205
| | - Jae-Jun Lee
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon 24523, Korea; (J.-J.L.); (Y.K.)
- Departments of Anesthesiology and Pain Medicine, College of Medicine, Hallym University, Chuncheon 24523, Korea
| | - Youngmi Kim
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon 24523, Korea; (J.-J.L.); (Y.K.)
| | - Keun-Young Lee
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Hallym University College of Medicine, Kangnam Sacred Heart Hospital, Seoul 07441, Korea;
| |
Collapse
|
21
|
Salliss ME, Maarsingh JD, Garza C, Łaniewski P, Herbst-Kralovetz MM. Veillonellaceae family members uniquely alter the cervical metabolic microenvironment in a human three-dimensional epithelial model. NPJ Biofilms Microbiomes 2021; 7:57. [PMID: 34230496 PMCID: PMC8260719 DOI: 10.1038/s41522-021-00229-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
Bacterial vaginosis (BV) is a gynecologic disorder characterized by a shift in cervicovaginal microbiota from Lactobacillus spp. dominance to a polymicrobial biofilm composed of diverse anaerobes. We utilized a well-characterized human three-dimensional cervical epithelial cell model in conjunction with untargeted metabolomics and immunoproteomics analyses to determine the immunometabolic contribution of three members of the Veillonellaceae family: Veillonella atypica, Veillonella montpellierensis and Megasphaera micronuciformis at this site. We found that Veillonella spp. infections induced significant elevation of polyamines. M. micronuciformis infections significantly increased soluble inflammatory mediators, induced moderate levels of cell cytotoxicity, and accumulation of cell membrane lipids relative to Veillonella spp. Notably, both V. atypica and V. montpellierensis infections resulted in consumption of lactate, a key metabolite linked to gynecologic and reproductive health. Collectively our approach and data provide unique insights into the specific contributions of Veillonellaceae members to the pathogenesis of BV and women's health.
Collapse
Affiliation(s)
- Mary E Salliss
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- Department of Biology and Biochemistry, University of Bath, Bath, UK
| | - Jason D Maarsingh
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Camryn Garza
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- Arizona State University, Tempe, AZ, USA
| | - Paweł Łaniewski
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Melissa M Herbst-Kralovetz
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA.
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA.
| |
Collapse
|
22
|
Origoni M, Fedele F, Parma M, Di Fatta S, Bergamini V, Candiani M, Fedele L. The Peritoneal Neovagina after Davydov's Laparoscopic Procedure in Mayer-Rokitansky-Küster-Hauser Syndrome: Morphology and Ultrastructure Investigation of the New Epithelium. J Minim Invasive Gynecol 2021; 28:1795-1799. [PMID: 33852967 DOI: 10.1016/j.jmig.2021.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 02/01/2021] [Accepted: 04/06/2021] [Indexed: 11/17/2022]
Abstract
STUDY OBJECTIVE To investigate the clinical appearance and morphologic and ultrastructural aspects of the mucosa of the peritoneal neovagina after laparoscopic Davydov neovaginoplasty in patients with Mayer-Rokitansky-Küster-Hauser syndrome. DESIGN The study group was a prospective, observational, experimental cohort of cases treated in the same institution between 2015 and 2019. Patients were followed up at 3, 6, and 12 months after surgery and then every 12 months. SETTING Single-center academic institution and teaching hospital in Milan, Italy. PATIENTS Fifty-one consecutive subjects with clinical and imaging diagnosis of Mayer-Rokitansky-Küster-Hauser syndrome surgically treated by the same team and postoperatively followed. INTERVENTIONS All the subjects underwent the same standardized surgical procedure and thereafter were followed up at 3, 6, and 12 months after surgery and then every 12 months; a minimum follow-up of 12 months was achieved in all cases. Vaginoscopy and Schiller test were performed at each follow-up visit, and a biopsy specimen of the neovagina was obtained in a limited number of patients (6 out of 51) for light microscopy (LM) and scanning electron microscopy (SEM) analysis of the tissue. MEASUREMENTS AND MAIN RESULTS In vaginoscopy, the neovaginal mucosa appeared homogeneous, smooth, and pink all along the neovaginal tract; the Schiller test detected iodine positivity at different degrees of extension upward from the hymenal ring, starting at 3 months postoperatively with almost complete positivity between 6 to 12 months in all cases. LM demonstrated adequate thickness and differentiation of the new mucosa along with the presence of glycogen storage; SEM revealed an ultrastructural surface appearance very close to normality. The main difference compared with a normal vagina was the reduced presence of vaginal mucosal folds. CONCLUSION Under different techniques (vaginoscopy, Schiller test, LM, and SEM), a minimum of 6 months after surgery, the peritoneal neovagina epithelium showed aspects comparable to the natal mucosa of the vagina.
Collapse
Affiliation(s)
- Massimo Origoni
- Department of Gynecology & Obstetrics, Ospedale San Raffaele Scientific Institute, Vita-Salute San Raffaele University School of Medicine, Milan (Drs. Origoni, Fedele, Parma, Di Fatta, Candiani, and Fedele).
| | - Francesco Fedele
- Department of Gynecology & Obstetrics, Ospedale San Raffaele Scientific Institute, Vita-Salute San Raffaele University School of Medicine, Milan (Drs. Origoni, Fedele, Parma, Di Fatta, Candiani, and Fedele)
| | - Marta Parma
- Department of Gynecology & Obstetrics, Ospedale San Raffaele Scientific Institute, Vita-Salute San Raffaele University School of Medicine, Milan (Drs. Origoni, Fedele, Parma, Di Fatta, Candiani, and Fedele)
| | - Simona Di Fatta
- Department of Gynecology & Obstetrics, Ospedale San Raffaele Scientific Institute, Vita-Salute San Raffaele University School of Medicine, Milan (Drs. Origoni, Fedele, Parma, Di Fatta, Candiani, and Fedele)
| | - Valentino Bergamini
- Department of Gynecology & Obstetrics B, Azienda Ospedaliera Universitaria Integrata Verona, Verona (Dr. Bergamini), Italy
| | - Massimo Candiani
- Department of Gynecology & Obstetrics, Ospedale San Raffaele Scientific Institute, Vita-Salute San Raffaele University School of Medicine, Milan (Drs. Origoni, Fedele, Parma, Di Fatta, Candiani, and Fedele)
| | - Luigi Fedele
- Department of Gynecology & Obstetrics, Ospedale San Raffaele Scientific Institute, Vita-Salute San Raffaele University School of Medicine, Milan (Drs. Origoni, Fedele, Parma, Di Fatta, Candiani, and Fedele)
| |
Collapse
|
23
|
Saba I, Barat C, Chabaud S, Reyjon N, Leclerc M, Jakubowska W, Orabi H, Lachhab A, Pelletier M, Tremblay MJ, Bolduc S. Immunocompetent Human 3D Organ-Specific Hormone-Responding Vaginal Mucosa Model of HIV-1 Infection. Tissue Eng Part C Methods 2021; 27:152-166. [PMID: 33573474 DOI: 10.1089/ten.tec.2020.0333] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The lack of appropriate experimental models often limits our ability to investigate the establishment of infections in specific tissues. To reproduce the structural and spatial organization of vaginal mucosae to study human immunodeficiency virus type-1 (HIV-1) infection, we used the self-assembly technique to bioengineer tridimensional vaginal mucosae using human cells extracted from HIV-1-negative healthy pre- and postmenopausal donors. We produced a stroma, free of exogenous material, that can be adapted to generate near-to-native vaginal tissue with the best complexity obtained with seeded epithelial cells on the organ-specific stroma. The autologous engineered tissues had mechanical properties close to native mucosa and shared similar glycogen production, which declined in reconstructed tissues of the postmenopausal donor. The in vitro-engineered tissues were also rendered immune competent by adding human monocyte-derived macrophages (MDMs) on the epithelium or in the stroma layers. The model was infected with HIV-1, and viral replication and transcytosis were observed when immunocompetent reconstructed vaginal mucosa tissue has incorporated MDMs into the stroma and infected with free HIV-1 green fluorescent protein (GFP) viral particles. These data illustrate a natural permissiveness of immunocompetent untransformed human vaginal mucosae to HIV-1 infection. This model offers a physiological tool to explore viral load, HIV-1 transmission in an environment that may contribute to the virus propagation, and new antiviral treatments in vitro. Impact statement This study introduces an innovative immunocompetent three-dimensional human organ-specific vaginal mucosa free of exogenous material for in vitro modeling of human immunodeficiency virus type-1 (HIV-1) infection. The proposed model is histologically close to native tissue, especially by presenting glycogen accumulation in the epithelium's superficial cells, responsive to estrogen, and able to sustain a monocyte-derived macrophage population infected or not by HIV-1 during ∼2 months.
Collapse
Affiliation(s)
- Ingrid Saba
- Centre de Recherche en Organogenèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec City, Canada
| | - Corinne Barat
- Infectious and Immune Diseases, Centre de Recherche du CHU de Québec-Université Laval, Québec City, Canada
| | - Stéphane Chabaud
- Centre de Recherche en Organogenèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec City, Canada
| | - Nolan Reyjon
- Centre de Recherche en Organogenèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec City, Canada
| | - Maude Leclerc
- Centre de Recherche en Organogenèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec City, Canada
| | - Weronika Jakubowska
- Centre de Recherche en Organogenèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec City, Canada
| | - Hazem Orabi
- Centre de Recherche en Organogenèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec City, Canada
| | - Asmaa Lachhab
- Infectious and Immune Diseases, Centre de Recherche du CHU de Québec-Université Laval, Québec City, Canada
| | - Martin Pelletier
- Infectious and Immune Diseases, Centre de Recherche du CHU de Québec-Université Laval, Québec City, Canada
| | - Michel J Tremblay
- Infectious and Immune Diseases, Centre de Recherche du CHU de Québec-Université Laval, Québec City, Canada
| | - Stéphane Bolduc
- Centre de Recherche en Organogenèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec City, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec City, Canada
| |
Collapse
|
24
|
Łaniewski P, Owen KA, Khnanisho M, Brotman RM, Herbst-Kralovetz MM. Clinical and Personal Lubricants Impact the Growth of Vaginal Lactobacillus Species and Colonization of Vaginal Epithelial Cells: An in Vitro Study. Sex Transm Dis 2021; 48:63-70. [PMID: 32842049 PMCID: PMC8793461 DOI: 10.1097/olq.0000000000001272] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Vaginal lubricants are commonly used during gynecological examinations, during sexual activities, or to alleviate vaginal dryness. Many lubricants contain potentially bacteriostatic or bactericidal agents (parabens, chlorhexidine gluconate, nonoxynol-9). Our objective was to evaluate the impact of lubricants that vary in formulation on the growth and viability of vaginal Lactobacillus species and vaginal epithelial cell (VEC) colonization in an in vitro model. METHODS Growth curve, disk diffusion, and minimal inhibitory assays were used to determine the impact of lubricants or excipients on the growth of Lactobacillus crispatus, Lactobacillus gasseri, Lactobacillus jensenii, and Lactobacillus iners. L. crispatus strain was used in VEC colonization assays. Statistical differences were determined by analysis of variance. RESULTS Lubricants containing chlorhexidine gluconate or nonoxynol-9 (N-9; Conceptrol, K-Y Jelly, and Surgilube) significantly inhibited Lactobacillus species growth (P < 0.05). In contrast, other clinical lubricants (E-Z Lubricating Jelly, McKesson Lubricating) and personal lubricants (Astroglide Liquid, Good Clean Love Almost Naked, K-Y Warming Jelly) did not exhibit this effect. Chlorhexidine gluconate had a detrimental effect on Lactobacillus growth and exhibited stronger antimicrobial activity compared with methylparaben and propylparaben (P < 0.0001). There were lubricants that did not induce cytotoxicity in VEC (Good Clean Love Almost Naked, E-Z Lubricating Jelly, McKesson Lubricating Jelly), but these products did substantially decrease the attachment of L. crispatus to VEC, particularly when VEC were preexposed to lubricants before inoculation with bacteria (P < 0.0001). CONCLUSIONS This in vitro model indicates that select vaginal lubricants, particularly those with chlorhexidine gluconate, have potentially adverse effects on women's health by reducing growth and recolonization of vaginal Lactobacillus species.
Collapse
Affiliation(s)
- Paweł Łaniewski
- College of Medicine-Phoenix, University of Arizona, Phoenix, AZ
| | - Kimberley A. Owen
- College of Medicine-Phoenix, University of Arizona, Phoenix, AZ
- University of Bath, Bath, UK
| | - Michael Khnanisho
- College of Medicine-Phoenix, University of Arizona, Phoenix, AZ
- Arizona State University, Tempe, AZ
| | | | | |
Collapse
|
25
|
Advanced 3D Cell Culture Techniques in Micro-Bioreactors, Part II: Systems and Applications. Processes (Basel) 2020. [DOI: 10.3390/pr9010021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this second part of our systematic review on the research area of 3D cell culture in micro-bioreactors we give a detailed description of the published work with regard to the existing micro-bioreactor types and their applications, and highlight important results gathered with the respective systems. As an interesting detail, we found that micro-bioreactors have already been used in SARS-CoV research prior to the SARS-CoV2 pandemic. As our literature research revealed a variety of 3D cell culture configurations in the examined bioreactor systems, we defined in review part one “complexity levels” by means of the corresponding 3D cell culture techniques applied in the systems. The definition of the complexity is thereby based on the knowledge that the spatial distribution of cell-extracellular matrix interactions and the spatial distribution of homologous and heterologous cell–cell contacts play an important role in modulating cell functions. Because at least one of these parameters can be assigned to the 3D cell culture techniques discussed in the present review, we structured the studies according to the complexity levels applied in the MBR systems.
Collapse
|
26
|
Advanced 3D Cell Culture Techniques in Micro-Bioreactors, Part I: A Systematic Analysis of the Literature Published between 2000 and 2020. Processes (Basel) 2020. [DOI: 10.3390/pr8121656] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Bioreactors have proven useful for a vast amount of applications. Besides classical large-scale bioreactors and fermenters for prokaryotic and eukaryotic organisms, micro-bioreactors, as specialized bioreactor systems, have become an invaluable tool for mammalian 3D cell cultures. In this systematic review we analyze the literature in the field of eukaryotic 3D cell culture in micro-bioreactors within the last 20 years. For this, we define complexity levels with regard to the cellular 3D microenvironment concerning cell–matrix-contact, cell–cell-contact and the number of different cell types present at the same time. Moreover, we examine the data with regard to the micro-bioreactor design including mode of cell stimulation/nutrient supply and materials used for the micro-bioreactors, the corresponding 3D cell culture techniques and the related cellular microenvironment, the cell types and in vitro models used. As a data source we used the National Library of Medicine and analyzed the studies published from 2000 to 2020.
Collapse
|
27
|
Jackson R, Maarsingh J, Herbst-Kralovetz MM, Van Doorslaer K. 3D Oral and Cervical Tissue Models for Studying Papillomavirus Host-Pathogen Interactions. CURRENT PROTOCOLS IN MICROBIOLOGY 2020; 59:e129. [PMID: 33232584 PMCID: PMC11088941 DOI: 10.1002/cpmc.129] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human papillomavirus (HPV) infection occurs in differentiating epithelial tissues. Cancers caused by high-risk types (e.g., HPV16 and HPV18) typically occur at oropharyngeal and anogenital anatomical sites. The HPV life cycle is differentiation-dependent, requiring tissue culture methodology that is able to recapitulate the three-dimensional (3D) stratified epithelium. Here we report two distinct and complementary methods for growing differentiating epithelial tissues that mimic many critical morphological and biochemical aspects of in vivo tissue. The first approach involves growing primary human epithelial cells on top of a dermal equivalent consisting of collagen fibers and living fibroblast cells. When these cells are grown at the liquid-air interface, differentiation occurs and allows for epithelial stratification. The second approach uses a rotating wall vessel bioreactor. The low-fluid-shear microgravity environment inside the bioreactor allows the cells to use collagen-coated microbeads as a growth scaffold and self-assemble into 3D cellular aggregates. These approaches are applied to epithelial cells derived from HPV-positive and HPV-negative oral and cervical tissues. The second part of the article introduces potential downstream applications for these 3D tissue models. We describe methods that will allow readers to start successfully culturing 3D tissues from oral and cervical cells. These tissues have been used for microscopic visualization, scanning electron microscopy, and large omics-based studies to gain insights into epithelial biology, the HPV life cycle, and host-pathogen interactions. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Establishing human primary cell-derived 3D organotypic raft cultures Support Protocol 1: Isolation of epithelial cells from patient-derived tissues Support Protocol 2: Growth and maintenance of primary human epithelial cells in monolayer culture Support Protocol 3: PCR-based HPV screening of primary cell cultures Basic Protocol 2: Establishing human 3D cervical tissues using the rotating wall vessel bioreactor Support Protocol 4: Growth and maintenance of human A2EN cells in monolayer culture Support Protocol 5: Preparation of the slow-turning lateral vessel bioreactor Support Protocol 6: Preparation of Cytodex-3 microcarrier beads Basic Protocol 3: Histological assessment of 3D organotypic raft tissues Basic Protocol 4: Spatial analysis of protein expression in 3D organotypic raft cultures Basic Protocol 5: Immunofluorescence imaging of RWV-derived 3D tissues Basic Protocol 6: Ultrastructural visualization and imaging of RWV-derived 3D tissues Basic Protocol 7: Characterization of gene expression by RT-qPCR.
Collapse
Affiliation(s)
- Robert Jackson
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA 85721
| | - Jason Maarsingh
- Department of Obstetrics and Gynecology, University of Arizona, College of Medicine-Phoenix, Phoenix, AZ, USA 85004
| | - Melissa M. Herbst-Kralovetz
- Department of Obstetrics and Gynecology, University of Arizona, College of Medicine-Phoenix, Phoenix, AZ, USA 85004
- Department of Basic Medical Sciences; BIO5 Institute; Clinical Translational Sciences Graduate Program; University of Arizona Cancer Center, University of Arizona, College of Medicine-Phoenix, Phoenix, AZ, USA 85004
| | - Koenraad Van Doorslaer
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA 85721
- Department of Immunobiology; BIO5 Institute; Cancer Biology Graduate Interdisciplinary Program; Genetics Graduate Interdisciplinary Program; and University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA 85721
| |
Collapse
|
28
|
Ilhan ZE, Łaniewski P, Tonachio A, Herbst-Kralovetz MM. Members of Prevotella Genus Distinctively Modulate Innate Immune and Barrier Functions in a Human Three-Dimensional Endometrial Epithelial Cell Model. J Infect Dis 2020; 222:2082-2092. [PMID: 32515473 PMCID: PMC7661762 DOI: 10.1093/infdis/jiaa324] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/05/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Prevotella species are commonly isolated from the reproductive tract of women with obstetric/gynecologic health complications. However, contributions of this genus to changes in local microenvironment are not well characterized. Our objective was to evaluate species-specific effects of Prevotella on the human endometrial epithelium. METHODS Thirteen Prevotella strains, originally isolated from the human oral cavity, amniotic fluid, endometrium, or vagina (including women with bacterial vaginosis), were obtained from BEI and ATCC resources. Bacteria were evaluated in silico and in vitro using human endometrial epithelial cells (EEC) grown as monolayers or a 3-dimensional (3D) model. RESULTS Genomic characterization illustrated metabolic and phylogenetic diversity of Prevotella genus. Among tested species, P. disiens exhibited cytotoxicity. Scanning electron microscopy analysis of the 3D EEC model revealed species-specific colonization patterns and alterations of ultracellular structures. Infection with sialidase-producing P. timonensis resulted in elongated microvilli, and increased MUC3 and MUC4 expression. Infections with Prevotella species, including P. bivia, did not result in significant proinflammatory activation of EEC. CONCLUSIONS Collectively, findings indicate that Prevotella species are metabolically diverse and overall not cytotoxic or overtly inflammatory in EEC; however, these bacteria can form biofilms, alter barrier properties of the endometrial epithelium, and ultimately impact colonization of secondary colonizers.
Collapse
Affiliation(s)
- Zehra Esra Ilhan
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Paweł Łaniewski
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Adriana Tonachio
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Melissa M Herbst-Kralovetz
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| |
Collapse
|
29
|
Heidari-Khoei H, Esfandiari F, Hajari MA, Ghorbaninejad Z, Piryaei A, Baharvand H. Organoid technology in female reproductive biomedicine. Reprod Biol Endocrinol 2020; 18:64. [PMID: 32552764 PMCID: PMC7301968 DOI: 10.1186/s12958-020-00621-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Recent developments in organoid technology are revolutionizing our knowledge about the biology, physiology, and function of various organs. Female reproductive biology and medicine also benefit from this technology. Organoids recapitulate features of different reproductive organs including the uterus, fallopian tubes, and ovaries, as well as trophoblasts. The genetic stability of organoids and long-lasting commitment to their tissue of origin during long-term culture makes them attractive substitutes for animal and in vitro models. Despite current limitations, organoids offer a promising platform to address fundamental questions regarding the reproductive system's physiology and pathology. They provide a human source to harness stem cells for regenerative medicine, heal damaged epithelia in specific diseases, and study biological processes in healthy and pathological conditions. The combination of male and female reproductive organoids with other technologies, such as microfluidics technology, would enable scientists to create a multi-organoid-on-a-chip platform for the next step to human-on-a-chip platforms for clinical applications, drug discovery, and toxicology studies. The present review discusses recent advances in producing organoid models of reproductive organs and highlights their applications, as well as technical challenges and future directions.
Collapse
Affiliation(s)
- Heidar Heidari-Khoei
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box: 16635-148, Tehran, 1665659911, Iran
| | - Fereshteh Esfandiari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box: 16635-148, Tehran, 1665659911, Iran
| | - Mohammad Amin Hajari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box: 16635-148, Tehran, 1665659911, Iran
| | - Zeynab Ghorbaninejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box: 16635-148, Tehran, 1665659911, Iran
| | - Abbas Piryaei
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box: 19395-4719, Tehran, Iran.
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box: 16635-148, Tehran, 1665659911, Iran.
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
30
|
Wilkinson EM, Łaniewski P, Herbst-Kralovetz MM, Brotman RM. Personal and Clinical Vaginal Lubricants: Impact on Local Vaginal Microenvironment and Implications for Epithelial Cell Host Response and Barrier Function. J Infect Dis 2020; 220:2009-2018. [PMID: 31539059 DOI: 10.1093/infdis/jiz412] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/08/2019] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND A majority of US women report past use of vaginal lubricants to enhance the ease and comfort of intimate sexual activities. Lubricants are also administered frequently in clinical practice. We sought to investigate if hyperosmolar lubricants are toxic to the vaginal mucosal epithelia. METHODS We tested a panel of commercially available lubricants across a range of osmolalities in human monolayer vaginal epithelial cell (VEC) culture and a robust 3-dimensional (3-D) VEC model. The impact of each lubricant on cellular morphology, cytotoxicity, barrier targets, and the induction of inflammatory mediators was examined. Conceptrol, containing nonoxynol-9, was used as a cytotoxicity control. RESULTS We observed a loss of intercellular connections, and condensation of chromatin, with increasing lubricant osmolality. EZ Jelly, K-Y Jelly, Astroglide, and Conceptrol induced cytotoxicity in both models at 24 hours. There was a strong positive correlation (r = 0.7326) between lubricant osmolality and cytotoxicity in monolayer VECs, and cell viability was reduced in VECs exposed to all the lubricants tested for 24 hours, except McKesson. Notably, select lubricants altered cell viability, barrier targets, and inflammatory mediators in 3-D VECs. CONCLUSIONS These findings indicate that hyperosmolar lubricants alter VEC morphology and are selectively cytotoxic, inflammatory, and barrier disrupting in the 3-D VEC model.
Collapse
Affiliation(s)
- Ellen M Wilkinson
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona.,Department of Biology and Biochemistry, University of Bath, United Kingdom
| | - Paweł Łaniewski
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Baltimore
| | - Melissa M Herbst-Kralovetz
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona.,Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Baltimore
| | - Rebecca M Brotman
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore
| |
Collapse
|
31
|
Łaniewski P, Ilhan ZE, Herbst-Kralovetz MM. The microbiome and gynaecological cancer development, prevention and therapy. Nat Rev Urol 2020; 17:232-250. [PMID: 32071434 PMCID: PMC9977514 DOI: 10.1038/s41585-020-0286-z] [Citation(s) in RCA: 217] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2020] [Indexed: 12/16/2022]
Abstract
The female reproductive tract (FRT), similar to other mucosal sites, harbours a site-specific microbiome, which has an essential role in maintaining health and homeostasis. In the majority of women of reproductive age, the microbiota of the lower FRT (vagina and cervix) microenvironment is dominated by Lactobacillus species, which benefit the host through symbiotic relationships. By contrast, the upper FRT (uterus, Fallopian tubes and ovaries) might be sterile in healthy individuals or contain a low-biomass microbiome with a diverse mixture of microorganisms. When dysbiosis occurs, altered immune and metabolic signalling can affect hallmarks of cancer, including chronic inflammation, epithelial barrier breach, changes in cellular proliferation and apoptosis, genome instability, angiogenesis and metabolic dysregulation. These pathophysiological changes might lead to gynaecological cancer. Emerging evidence shows that genital dysbiosis and/or specific bacteria might have an active role in the development and/or progression and metastasis of gynaecological malignancies, such as cervical, endometrial and ovarian cancers, through direct and indirect mechanisms, including modulation of oestrogen metabolism. Cancer therapies might also alter microbiota at sites throughout the body. Reciprocally, microbiota composition can influence the efficacy and toxic effects of cancer therapies, as well as quality of life following cancer treatment. Modulation of the microbiome via probiotics or microbiota transplant might prove useful in improving responsiveness to cancer treatment and quality of life. Elucidating these complex host-microbiome interactions, including the crosstalk between distal and local sites, will translate into interventions for prevention, therapeutic efficacy and toxic effects to enhance health outcomes for women with gynaecological cancers.
Collapse
Affiliation(s)
- Paweł Łaniewski
- Department of Basic Medical Sciences, College of Medicine – Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Zehra Esra Ilhan
- Department of Obstetrics and Gynecology, College of Medicine – Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Melissa M. Herbst-Kralovetz
- Department of Basic Medical Sciences, College of Medicine – Phoenix, University of Arizona, Phoenix, AZ, USA,Department of Obstetrics and Gynecology, College of Medicine – Phoenix, University of Arizona, Phoenix, AZ, USA,UA Cancer Center, University of Arizona, Phoenix/Tucson, AZ, USA,Correspondence:
| |
Collapse
|
32
|
Analysis of Host Responses to Neisseria gonorrhoeae Using a Human Three-Dimensional Endometrial Epithelial Cell Model. Methods Mol Biol 2020; 1997:347-361. [PMID: 31119633 DOI: 10.1007/978-1-4939-9496-0_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Neisseria gonorrhoeae infections have been associated with complications including chronic endometritis and pelvic inflammatory disease. Robust in vitro models of the female reproductive tract are urgently needed to better understand the biological mechanisms leading to these pathophysiological changes. Our human three-dimensional (3D) endometrial epithelial cell (EEC) model, which is generated using the HEC-1A cell line and rotating wall vessel (RWV) bioreactor technology, replicates several hallmarks of endometrial tissue in vivo. Studying the interactions of N. gonorrhoeae with the host using this newly characterized human 3D EEC model allows for the investigation of unique mechanisms of gonococcal pathogenesis in the upper female reproductive tract. In this chapter, we describe methodologies that can be used to investigate the interactions of N. gonorrhoeae with the human 3D endometrial epithelium. Protocols for generating the human 3D EEC model using the RWV technology and assessing the host response (including morphological/ultrastructural changes to the epithelial cells; cytokine/chemokine secretion or gene expression changes) following infection with N. gonorrhoeae are presented.
Collapse
|
33
|
Modeling Host-Pathogen Interactions in the Context of the Microenvironment: Three-Dimensional Cell Culture Comes of Age. Infect Immun 2018; 86:IAI.00282-18. [PMID: 30181350 DOI: 10.1128/iai.00282-18] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tissues and organs provide the structural and biochemical landscapes upon which microbial pathogens and commensals function to regulate health and disease. While flat two-dimensional (2-D) monolayers composed of a single cell type have provided important insight into understanding host-pathogen interactions and infectious disease mechanisms, these reductionist models lack many essential features present in the native host microenvironment that are known to regulate infection, including three-dimensional (3-D) architecture, multicellular complexity, commensal microbiota, gas exchange and nutrient gradients, and physiologically relevant biomechanical forces (e.g., fluid shear, stretch, compression). A major challenge in tissue engineering for infectious disease research is recreating this dynamic 3-D microenvironment (biological, chemical, and physical/mechanical) to more accurately model the initiation and progression of host-pathogen interactions in the laboratory. Here we review selected 3-D models of human intestinal mucosa, which represent a major portal of entry for infectious pathogens and an important niche for commensal microbiota. We highlight seminal studies that have used these models to interrogate host-pathogen interactions and infectious disease mechanisms, and we present this literature in the appropriate historical context. Models discussed include 3-D organotypic cultures engineered in the rotating wall vessel (RWV) bioreactor, extracellular matrix (ECM)-embedded/organoid models, and organ-on-a-chip (OAC) models. Collectively, these technologies provide a more physiologically relevant and predictive framework for investigating infectious disease mechanisms and antimicrobial therapies at the intersection of the host, microbe, and their local microenvironments.
Collapse
|
34
|
Gardner JK, Herbst-Kralovetz MM. IL-36γ induces a transient HSV-2 resistant environment that protects against genital disease and pathogenesis. Cytokine 2018; 111:63-71. [PMID: 30118914 DOI: 10.1016/j.cyto.2018.07.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/10/2018] [Accepted: 07/30/2018] [Indexed: 01/03/2023]
Abstract
Herpes simplex virus 2 (HSV-2) causes a persistent, lifelong infection that increases risk for sexually transmitted infection acquisition. Both the lack of a vaccine and the need for chronic suppressive therapies to control infection presents the need to further understand immune mechanisms in response to acute HSV-2 infection. The IL-36 cytokines are recently identified members of the IL-1 family and function as inflammatory mediators at epithelial sites. Here, we first used a well-characterized three-dimensional (3-D) human vaginal epithelial cell (VEC) model to understand the role of IL-36γ in the context of HSV-2 infection. In 3-D VEC, IL-36γ is induced by HSV-2 infection, and pretreatment with exogenous IL-36γ significantly reduced HSV-2 replication. To assess the impact of IL-36γ treatment on HSV-2 disease pathogenesis, we employed a lethal genital infection model. We showed that IL-36γ treatment in mice prior to lethal intravaginal challenge significantly limited vaginal viral replication, delayed disease onset, decreased disease severity, and significantly increased survival. We demonstrated that IL-36γ treatment transiently induced pro-inflammatory cytokines, chemokines, and antimicrobial peptides in murine lower female reproductive tract (FRT) tissue and vaginal lavages. Induction of the chemokines CCL20 and KC in IL-36γ treated mice also corresponded with increased polymorphonuclear (PMN) leukocyte infiltration observed in vaginal smears. Altogether, these studies demonstrate that IL-36γ drives the transient production of immune mediators and promotes PMN recruitment in the vaginal microenvironment that increases resistance to HSV-2 infection and disease. Our data indicate that IL-36γ may participate as a key player in host defense mechanisms against invading pathogens in the FRT.
Collapse
Affiliation(s)
- Jameson K Gardner
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA; Molecular and Cellular Biology Graduate Program, School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Melissa M Herbst-Kralovetz
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA; Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA.
| |
Collapse
|
35
|
Wilkinson EM, Ilhan ZE, Herbst-Kralovetz MM. Microbiota–drug interactions: Impact on metabolism and efficacy of therapeutics. Maturitas 2018; 112:53-63. [DOI: 10.1016/j.maturitas.2018.03.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 03/23/2018] [Accepted: 03/29/2018] [Indexed: 02/06/2023]
|
36
|
Baker JM, Chase DM, Herbst-Kralovetz MM. Uterine Microbiota: Residents, Tourists, or Invaders? Front Immunol 2018; 9:208. [PMID: 29552006 PMCID: PMC5840171 DOI: 10.3389/fimmu.2018.00208] [Citation(s) in RCA: 212] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 01/24/2018] [Indexed: 12/14/2022] Open
Abstract
Uterine microbiota have been reported under various conditions and populations; however, it is uncertain the level to which these bacteria are residents that maintain homeostasis, tourists that are readily eliminated or invaders that contribute to human disease. This review provides a historical timeline and summarizes the current status of this topic with the aim of promoting research priorities and discussion on this controversial topic. Discrepancies exist in current reports of uterine microbiota and are critically reviewed and examined. Established and putative routes of bacterial seeding of the human uterus and interactions with distal mucosal sites are discussed. Based upon the current literature, we highlight the need for additional robust clinical and translational studies in this area. In addition, we discuss the necessity for investigating host–microbiota interactions and the physiologic and functional impact of these microbiota on the local endometrial microenvironment as these mechanisms may influence poor reproductive, obstetric, and gynecologic health outcomes and sequelae.
Collapse
Affiliation(s)
- James M Baker
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, United States.,Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Dana M Chase
- Arizona Oncology (US Oncology Network), University of Arizona College of Medicine, Creighton University School of Medicine at St. Joseph's Hospital, Phoenix, AZ, United States
| | - Melissa M Herbst-Kralovetz
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, United States.,Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, United States
| |
Collapse
|
37
|
Mazzocchi A, Soker S, Skardal A. Biofabrication Technologies for Developing In Vitro Tumor Models. CANCER DRUG DISCOVERY AND DEVELOPMENT 2018. [DOI: 10.1007/978-3-319-60511-1_4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
38
|
Vyshenska D, Lam KC, Shulzhenko N, Morgun A. Interplay between viruses and bacterial microbiota in cancer development. Semin Immunol 2017; 32:14-24. [PMID: 28602713 DOI: 10.1016/j.smim.2017.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 05/03/2017] [Accepted: 05/30/2017] [Indexed: 12/29/2022]
Abstract
During the last few decades we have become accustomed to the idea that viruses can cause tumors. It is much less considered and discussed, however, that most people infected with oncoviruses will never develop cancer. Therefore, the genetic and environmental factors that tip the scales from clearance of viral infection to development of cancer are currently an area of active investigation. Microbiota has recently emerged as a potentially critical factor that would affect this balance by increasing or decreasing the ability of viral infection to promote carcinogenesis. In this review, we provide a model of microbiome contribution to the development of oncogenic viral infections and viral associated cancers, give examples of this process in human tumors, and describe the challenges that prevent progress in the field as well as their potential solutions.
Collapse
Affiliation(s)
- Dariia Vyshenska
- College of Pharmacy, Oregon State University, 1601 SW Jefferson Way, Corvallis, OR 97331, USA
| | - Khiem C Lam
- College of Pharmacy, Oregon State University, 1601 SW Jefferson Way, Corvallis, OR 97331, USA
| | - Natalia Shulzhenko
- College of Veterinary Medicine, Oregon State University, 208 Dryden Hall, Corvallis, OR 97331, USA.
| | - Andrey Morgun
- College of Pharmacy, Oregon State University, 1601 SW Jefferson Way, Corvallis, OR 97331, USA.
| |
Collapse
|
39
|
Devarasetty M, Wang E, Soker S, Skardal A. Mesenchymal stem cells support growth and organization of host-liver colorectal-tumor organoids and possibly resistance to chemotherapy. Biofabrication 2017; 9:021002. [PMID: 28589925 DOI: 10.1088/1758-5090/aa7484] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Despite having yielded extensive breakthroughs in cancer research, traditional 2D cell cultures have limitations in studying cancer progression and metastasis and screening therapeutic candidates. 3D systems can allow cells to grow, migrate, and interact with each other and the surrounding matrix, resulting in more realistic constructs. Furthermore, interactions between host tissue and developing tumors influence the susceptibility of tumors to drug treatments. Host-liver colorectal-tumor spheroids composed of primary human hepatocytes, mesenchymal stem cells (MSC) and colon carcinoma HCT116 cells were created in simulated microgravity rotating wall vessel (RWV) bioreactors. The cells were seeded on hyaluronic acid-based microcarriers, loaded with liver-specific growth factors and ECM components. Only in the presence of MSC, large tumor foci rapidly formed inside the spheroids and increased in size steadily over time, while not greatly impacting albumin secretion from hepatocytes. The presence of MSC appeared to drive self-organization and formation of a stroma-like tissue surrounding the tumor foci and hepatocytes. Exposure to a commonly used chemotherapeutic 5-FU showed a dose-dependent cytotoxicity. However, if tumor organoids were allowed to mature in the RWV, they were less sensitive to the drug treatment. These data demonstrate the potential utility of liver tumor organoids for cancer progression and drug response modeling.
Collapse
Affiliation(s)
- Mahesh Devarasetty
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, United States of America. Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Winston-Salem, NC, United States of America
| | | | | | | |
Collapse
|
40
|
De Gregorio V, Imparato G, Urciuolo F, Tornesello ML, Annunziata C, Buonaguro FM, Netti PA. An Engineered Cell-Instructive Stroma for the Fabrication of a Novel Full Thickness Human Cervix Equivalent In Vitro. Adv Healthc Mater 2017; 6. [PMID: 28371541 DOI: 10.1002/adhm.201601199] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/18/2017] [Indexed: 12/22/2022]
Abstract
There is a growing interest for developing organotypic cervical models by using primary cervical cells that are able to reproduce the physiological relevant stromal microenvironment and the distinctive histology of the native cervical epithelium. Here for the first time it is reported the production of an organotypic cervical model featured by a scaffold-free stromal tissue resembling the extracellular matrix (ECM) composition and organization of the native counterpart as well as a completely well-differentiated epithelium. To reach this aim, human cervical microtissue precursors have been produced, characterized, and used as functional building units to fabricate a cell-synthesized cervical stroma equivalent by means of a bottom-up approach. Immunotypization, and molecular and morphological analyses reveal the extent of fundamental epithelial biomarkers and the presence of collagen and noncollagenous molecules, demonstrating that the natural tissue architecture and biological characteristics of cervical tissues are reproduced. The results of this study suggest that the bottom-up technology used to produce these 3D human cervical equivalents provides a fully functional organotypic cervical model that may be used as a valuable tool to investigate the epithelial-stromal interactions as well as for testing new therapeutics in vitro.
Collapse
Affiliation(s)
- Vincenza De Gregorio
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125, Naples, Italy
| | - Giorgia Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125, Naples, Italy
| | - Francesco Urciuolo
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125, Naples, Italy
| | - Maria L Tornesello
- Molecular Biology and Viral Oncology Unit Istituto Nazionale, Tumori IRCCS "Fondazione Pascale", 80131, Naples, Italy
| | - Clorinda Annunziata
- Molecular Biology and Viral Oncology Unit Istituto Nazionale, Tumori IRCCS "Fondazione Pascale", 80131, Naples, Italy
| | - Franco M Buonaguro
- Molecular Biology and Viral Oncology Unit Istituto Nazionale, Tumori IRCCS "Fondazione Pascale", 80131, Naples, Italy
| | - Paolo A Netti
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125, Naples, Italy
- Molecular Biology and Viral Oncology Unit Istituto Nazionale, Tumori IRCCS "Fondazione Pascale", 80131, Naples, Italy
- Department of Chemical Materials and Industrial Production (DICMAPI), University of Naples Federico II, P.leTecchio 80, 80125, Naples, Italy
| |
Collapse
|
41
|
Hvastkovs EG, Rusling JF. Modern Approaches to Chemical Toxicity Screening. CURRENT OPINION IN ELECTROCHEMISTRY 2017; 3:18-22. [PMID: 29250606 PMCID: PMC5729768 DOI: 10.1016/j.coelec.2017.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Chemical toxicity has a serious impact on public health, and toxicity failures of drug candidates drive up drug development costs. Many in vitro bioassays exist for toxicity screening, and newer versions of these tend to be high throughput or high content assays, some of which rely on electrochemical detection. Toxicity very often results from metabolites of the chemicals we are exposed to, so it is important that assays feature metabolic conversion. Combining bioassays, computational predictions, and accurate chemical pathway elucidation presents our best chance for reliable toxicity prediction. Employing electrochemical and electrochemiluminescent approaches, cell-free microfluidic arrays can measure relative rates of formation of DNA-metabolite adduct formation (a measure of genotoxicity) as well as DNA oxidation levels resulting from enzyme-generated metabolites. Enzymes for several organ types can be studied simultaneously. These arrays can be used to identify the most reactive metabolites, and subsequent mechanistic details can then be investigated with high throughput LC-HPLC using enzyme/DNA-coated magnetic beads.
Collapse
Affiliation(s)
- Eli G Hvastkovs
- Department of Chemistry, East Carolina University, Greenville, NC 27858, USA
| | - James F Rusling
- Department of Chemistry, University of Connecticut, Storrs, CT 06269, USA
- Institute of Material Science, University of Connecticut, Storrs, CT 06269, USA
- Department of Surgery and Neag Cancer Center, University of Connecticut Health Center, Farmington, CT 06032, USA
- School of Chemistry, National University of Ireland at Galway, Ireland
| |
Collapse
|
42
|
Herbst-Kralovetz MM, Pyles RB, Ratner AJ, Sycuro LK, Mitchell C. New Systems for Studying Intercellular Interactions in Bacterial Vaginosis. J Infect Dis 2017; 214 Suppl 1:S6-S13. [PMID: 27449872 DOI: 10.1093/infdis/jiw130] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Bacterial vaginosis (BV) affects almost a quarter of US women, making it a condition of major public health relevance. Key questions remain regarding the etiology of BV, mechanisms for its association with poor reproductive health outcomes, and reasons for high rates of treatment failure. New model systems are required to answer these remaining questions, elucidate the complex host-microbe and microbe-microbe interactions, and develop new, effective interventions. In this review, we cover the strengths and limitations of in vitro and in vivo model systems to study these complex intercellular interactions. Furthermore, we discuss advancements needed to maximize the translational utility of the model systems. As no single model can recapitulate all of the complex physiological and environmental conditions of the human vaginal microenvironment, we conclude that combinatorial approaches using in vitro and in vivo model systems will be required to address the remaining fundamental questions surrounding the enigma that is BV.
Collapse
Affiliation(s)
| | - Richard B Pyles
- Department of Pediatrics, University of Texas Medical Branch, Galveston
| | - Adam J Ratner
- Departments of Pediatrics and Microbiology, New York University School of Medicine, New York
| | - Laura K Sycuro
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Caroline Mitchell
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston
| |
Collapse
|
43
|
Crabbé A, Liu Y, Matthijs N, Rigole P, De La Fuente-Nùñez C, Davis R, Ledesma MA, Sarker S, Van Houdt R, Hancock REW, Coenye T, Nickerson CA. Antimicrobial efficacy against Pseudomonas aeruginosa biofilm formation in a three-dimensional lung epithelial model and the influence of fetal bovine serum. Sci Rep 2017; 7:43321. [PMID: 28256611 PMCID: PMC5335707 DOI: 10.1038/srep43321] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 01/25/2017] [Indexed: 12/14/2022] Open
Abstract
In vitro models that mimic in vivo host-pathogen interactions are needed to evaluate candidate drugs that inhibit bacterial virulence traits. We established a new approach to study Pseudomonas aeruginosa biofilm susceptibility on biotic surfaces, using a three-dimensional (3-D) lung epithelial cell model. P. aeruginosa formed antibiotic resistant biofilms on 3-D cells without affecting cell viability. The biofilm-inhibitory activity of antibiotics and/or the anti-biofilm peptide DJK-5 were evaluated on 3-D cells compared to a plastic surface, in medium with and without fetal bovine serum (FBS). In both media, aminoglycosides were more efficacious in the 3-D cell model. In serum-free medium, most antibiotics (except polymyxins) showed enhanced efficacy when 3-D cells were present. In medium with FBS, colistin was less efficacious in the 3-D cell model. DJK-5 exerted potent inhibition of P. aeruginosa association with both substrates, only in serum-free medium. DJK-5 showed stronger inhibitory activity against P. aeruginosa associated with plastic compared to 3-D cells. The combined addition of tobramycin and DJK-5 exhibited more potent ability to inhibit P. aeruginosa association with both substrates. In conclusion, lung epithelial cells influence the efficacy of most antimicrobials against P. aeruginosa biofilm formation, which in turn depends on the presence or absence of FBS.
Collapse
Affiliation(s)
- Aurélie Crabbé
- Laboratory of Pharmaceutical Microbiology (LPM), Ghent University, Ghent, Belgium.,The Biodesign Institute, Center for Infectious Diseases and Vaccinology, Arizona State University, Tempe, Arizona, United States of America
| | - Yulong Liu
- The Biodesign Institute, Center for Infectious Diseases and Vaccinology, Arizona State University, Tempe, Arizona, United States of America
| | - Nele Matthijs
- Laboratory of Pharmaceutical Microbiology (LPM), Ghent University, Ghent, Belgium
| | - Petra Rigole
- Laboratory of Pharmaceutical Microbiology (LPM), Ghent University, Ghent, Belgium
| | - César De La Fuente-Nùñez
- University of British Columbia, Centre for Microbial Diseases and Immunity Research, Vancouver, British Columbia, Canada
| | - Richard Davis
- The Biodesign Institute, Center for Infectious Diseases and Vaccinology, Arizona State University, Tempe, Arizona, United States of America
| | - Maria A Ledesma
- The Biodesign Institute, Center for Infectious Diseases and Vaccinology, Arizona State University, Tempe, Arizona, United States of America
| | - Shameema Sarker
- The Biodesign Institute, Center for Infectious Diseases and Vaccinology, Arizona State University, Tempe, Arizona, United States of America
| | - Rob Van Houdt
- Unit of Microbiology, Belgian Nuclear Research Centre (SCK·CEN), Mol, Belgium
| | - Robert E W Hancock
- University of British Columbia, Centre for Microbial Diseases and Immunity Research, Vancouver, British Columbia, Canada
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology (LPM), Ghent University, Ghent, Belgium
| | - Cheryl A Nickerson
- The Biodesign Institute, Center for Infectious Diseases and Vaccinology, Arizona State University, Tempe, Arizona, United States of America.,School of Life Sciences, Arizona State University, Tempe, Arizona 85287, United States of America
| |
Collapse
|
44
|
Human Three-Dimensional Endometrial Epithelial Cell Model To Study Host Interactions with Vaginal Bacteria and Neisseria gonorrhoeae. Infect Immun 2017; 85:IAI.01049-16. [PMID: 28052997 DOI: 10.1128/iai.01049-16] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 12/12/2022] Open
Abstract
Colonization of the endometrium by pathogenic bacteria ascending from the lower female reproductive tract (FRT) is associated with many gynecologic and obstetric health complications. To study these host-microbe interactions in vitro, we developed a human three-dimensional (3-D) endometrial epithelial cell (EEC) model using the HEC-1A cell line and the rotating wall vessel (RWV) bioreactor technology. Our model, composed of 3-D EEC aggregates, recapitulates several functional/structural characteristics of human endometrial epithelial tissue, including cell differentiation, the presence of junctional complexes/desmosomes and microvilli, and the production of membrane-associated mucins and Toll-like receptors (TLRs). TLR function was evaluated by exposing the EEC aggregates to viral and bacterial products. Treatment with poly(I·C) and flagellin but not with synthetic lipoprotein (fibroblast-stimulating lipoprotein 1 [FSL-1]) or lipopolysaccharide (LPS) significantly induced proinflammatory mediators in a dose-dependent manner. To simulate ascending infection, we infected EEC aggregates with commensal and pathogenic bacteria: Lactobacillus crispatus, Gardnerella vaginalis, and Neisseria gonorrhoeae All vaginal microbiota and N. gonorrhoeae efficiently colonized the 3-D surface, localizing to crevices of the EEC model and interacting with multiple adjacent cells simultaneously. However, only infection with pathogenic N. gonorrhoeae and not infection with the other bacteria tested significantly induced proinflammatory mediators and significant ultrastructural changes to the host cells. The latter observation is consistent with clinical findings and illustrated the functional specificity of our system. Additionally, we highlighted the utility of the 3-D EEC model for the study of the pathogenesis of N. gonorrhoeae using a well-characterized ΔpilT mutant. Overall, this study demonstrates that the human 3-D EEC model is a robust tool for studying host-microbe interactions and bacterial pathogenesis in the upper FRT.
Collapse
|
45
|
Orabi H, Saba I, Rousseau A, Bolduc S. Novel three-dimensional autologous tissue-engineered vaginal tissues using the self-assembly technique. Transl Res 2017; 180:22-36. [PMID: 27543901 DOI: 10.1016/j.trsl.2016.07.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 07/17/2016] [Accepted: 07/23/2016] [Indexed: 02/08/2023]
Abstract
Many diseases necessitate the substitution of vaginal tissues. Current replacement therapies are associated with many complications. In this study, we aimed to create bioengineered neovaginas with the self-assembly technique using autologous vaginal epithelial (VE) and vaginal stromal (VS) cells without the use of exogenous materials and to document the survival and incorporation of these grafts into the tissues of nude female mice. Epithelial and stromal cells were isolated from vaginal biopsies. Stromal cells were driven to form collagen sheets, 3 of which were superimposed to form vaginal stromas. VE cells were seeded on top of these stromas and allowed to mature in an air-liquid interface. The vaginal equivalents were implanted subcutaneously in female nude mice, which were sacrificed after 1 and 2 weeks after surgery. The in vitro and animal-retrieved equivalents were assessed using histologic, functional, and mechanical evaluations. Vaginal equivalents could be handled easily. VE cells formed a well-differentiated epithelial layer with a continuous basement membrane. The equivalent matrix was composed of collagen I and III and elastin. The epithelium, basement membrane, and stroma were comparable to those of native vaginal tissues. The implanted equivalents formed mature vaginal epithelium and matrix that were integrated into the mice tissues. Using the self-assembly technique, in vitro vaginal tissues were created with many functional and biological similarities to native vagina without any foreign material. They formed functional vaginal tissues after in vivo animal implantation. It is appropriate for vaginal substitution and disease modeling for infectious studies, vaginal applicants, and drug testing.
Collapse
Affiliation(s)
- Hazem Orabi
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Faculté de médecine, Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada; Department of Surgery, Université Laval, Québec, Canada; Department of Urology, Assiut University, Assiut, Egypt.
| | - Ingrid Saba
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Faculté de médecine, Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada; Department of Surgery, Université Laval, Québec, Canada
| | - Alexandre Rousseau
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Faculté de médecine, Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada; Department of Surgery, Université Laval, Québec, Canada
| | - Stéphane Bolduc
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Faculté de médecine, Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada; Department of Surgery, Université Laval, Québec, Canada.
| |
Collapse
|
46
|
Abstract
Organotypic models are 3D in vitro representations of an in vivo environment. Their complexity can range from an epidermal replica to the establishment of a cancer microenvironment. These models have been used for many years, in an attempt to mimic the structure and function of cells and tissues found inside the body. Methods for developing 3D organotypic models differ according to the tissue of interest and the experimental design. For example, cultures may be grown submerged in culture medium and or at an air-liquid interface. Our group is focusing on an air-liquid interface 3D organotypic model. These cultures are grown on a nylon membrane-covered metal grid with the cells embedded in a Collagen-Matrigel gel. This allows cells to grow in an air-liquid interface to enable diffusion and nourishment from the medium below. Subsequently, the organotypic cultures can be used for immunohistochemical staining of various components of ERK signaling, which is a key player in mediating communication between cells and their microenvironment.
Collapse
|
47
|
Gardner JK, Herbst-Kralovetz MM. Three-Dimensional Rotating Wall Vessel-Derived Cell Culture Models for Studying Virus-Host Interactions. Viruses 2016; 8:v8110304. [PMID: 27834891 PMCID: PMC5127018 DOI: 10.3390/v8110304] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 10/18/2016] [Accepted: 10/31/2016] [Indexed: 12/31/2022] Open
Abstract
The key to better understanding complex virus-host interactions is the utilization of robust three-dimensional (3D) human cell cultures that effectively recapitulate native tissue architecture and model the microenvironment. A lack of physiologically-relevant animal models for many viruses has limited the elucidation of factors that influence viral pathogenesis and of complex host immune mechanisms. Conventional monolayer cell cultures may support viral infection, but are unable to form the tissue structures and complex microenvironments that mimic host physiology and, therefore, limiting their translational utility. The rotating wall vessel (RWV) bioreactor was designed by the National Aeronautics and Space Administration (NASA) to model microgravity and was later found to more accurately reproduce features of human tissue in vivo. Cells grown in RWV bioreactors develop in a low fluid-shear environment, which enables cells to form complex 3D tissue-like aggregates. A wide variety of human tissues (from neuronal to vaginal tissue) have been grown in RWV bioreactors and have been shown to support productive viral infection and physiological meaningful host responses. The in vivo-like characteristics and cellular features of the human 3D RWV-derived aggregates make them ideal model systems to effectively recapitulate pathophysiology and host responses necessary to conduct rigorous basic science, preclinical and translational studies.
Collapse
Affiliation(s)
- Jameson K Gardner
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ 85004, USA.
| | - Melissa M Herbst-Kralovetz
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ 85004, USA.
| |
Collapse
|
48
|
Three-dimensional cell culture models for investigating human viruses. Virol Sin 2016; 31:363-379. [PMID: 27822716 PMCID: PMC7090760 DOI: 10.1007/s12250-016-3889-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 10/21/2016] [Indexed: 12/15/2022] Open
Abstract
Three-dimensional (3D) culture models are physiologically relevant, as they provide reproducible results, experimental flexibility and can be adapted for high-throughput experiments. Moreover, these models bridge the gap between traditional two-dimensional (2D) monolayer cultures and animal models. 3D culture systems have significantly advanced basic cell science and tissue engineering, especially in the fields of cell biology and physiology, stem cell research, regenerative medicine, cancer research, drug discovery, and gene and protein expression studies. In addition, 3D models can provide unique insight into bacteriology, virology, parasitology and host-pathogen interactions. This review summarizes and analyzes recent progress in human virological research with 3D cell culture models. We discuss viral growth, replication, proliferation, infection, virus-host interactions and antiviral drugs in 3D culture models.
Collapse
|
49
|
Effects of Female Sex Hormones on Susceptibility to HSV-2 in Vaginal Cells Grown in Air-Liquid Interface. Viruses 2016; 8:v8090241. [PMID: 27589787 PMCID: PMC5035955 DOI: 10.3390/v8090241] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/19/2016] [Accepted: 08/22/2016] [Indexed: 01/05/2023] Open
Abstract
The lower female reproductive tract (FRT) is comprised of the cervix and vagina, surfaces that are continuously exposed to a variety of commensal and pathogenic organisms. Sexually transmitted viruses, such as herpes simplex virus type 2 (HSV-2), have to traverse the mucosal epithelial lining of the FRT to establish infection. The majority of current culture systems that model the host-pathogen interactions in the mucosal epithelium have limitations in simulating physiological conditions as they employ a liquid-liquid interface (LLI), in which both apical and basolateral surfaces are submerged in growth medium. We designed the current study to simulate in vivo conditions by growing an immortalized vaginal epithelial cell line (Vk2/E6E7) in culture with an air-liquid interface (ALI) and examined the effects of female sex hormones on their growth, differentiation, and susceptibility to HSV-2 under these conditions, in comparison to LLI cultures. ALI conditions induced Vk2/E6E7 cells to grow into multi-layered cultures compared to the monolayers present in LLI conditions. Vk2 cells in ALI showed higher production of cytokeratin in the presence of estradiol (E2), compared to cells grown in progesterone (P4). Cells grown under ALI conditions were exposed to HSV-2-green fluorescent protein (GFP) and the highest infection and replication was observed in the presence of P4. Altogether, this study suggests that ALI cultures more closely simulate the in vivo conditions of the FRT compared to the conventional LLI cultures. Furthermore, under these conditions P4 was found to confer higher susceptibility to HSV-2 infection in vaginal cells. The vaginal ALI culture system offers a better alternative to study host-pathogen interactions.
Collapse
|
50
|
Winkle SM, Throop AL, Herbst-Kralovetz MM. IL-36γ Augments Host Defense and Immune Responses in Human Female Reproductive Tract Epithelial Cells. Front Microbiol 2016; 7:955. [PMID: 27379082 PMCID: PMC4911402 DOI: 10.3389/fmicb.2016.00955] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 06/02/2016] [Indexed: 12/12/2022] Open
Abstract
IL-36γ is a proinflamatory cytokine which belongs to the IL-1 family of cytokines. It is expressed in the skin and by epithelial cells (ECs) lining lung and gut tissue. We used human 3-D organotypic cells, that recapitulate either in vivo human vaginal or cervical tissue, to explore the possible role of IL-36γ in host defense against pathogens in the human female reproductive tract (FRT). EC were exposed to compounds derived from virus or bacterial sources and induction and regulation of IL-36γ and its receptor was determined. Polyinosinic-polycytidylic acid (poly I:C), flagellin, and synthetic lipoprotein (FSL-1) significantly induced expression of IL-36γ in a dose-dependent manner, and appeared to be TLR-dependent. Recombinant IL-36γ treatment resulted in self-amplification of IL-36γ and its receptor (IL-36R) via increased gene expression, and promoted other inflammatory signaling pathways. This is the first report to demonstrate that the IL-36 receptor and IL-36γ are present in the human FRT EC and that they are differentially induced by microbial products at this site. We conclude that IL-36γ is a driver for epithelial and immune activation following microbial insult and, as such, may play a critical role in host defense in the FRT.
Collapse
Affiliation(s)
- Sean M Winkle
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix AZ, USA
| | - Andrea L Throop
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix AZ, USA
| | - Melissa M Herbst-Kralovetz
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix AZ, USA
| |
Collapse
|