1
|
Walraven T, Busch M, Wang J, Donkers JM, Duijvestein M, van de Steeg E, Kramer NI, Bouwmeester H. Elevated risk of adverse effects from foodborne contaminants and drugs in inflammatory bowel disease: a review. Arch Toxicol 2024; 98:3519-3541. [PMID: 39249550 PMCID: PMC11489187 DOI: 10.1007/s00204-024-03844-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/19/2024] [Indexed: 09/10/2024]
Abstract
The global burden of Inflammatory bowel disease (IBD) has been rising over the last decades. IBD is an intestinal disorder with a complex and largely unknown etiology. The disease is characterized by a chronically inflamed gastrointestinal tract, with intermittent phases of exacerbation and remission. This compromised intestinal barrier can contribute to, enhance, or even enable the toxicity of drugs, food-borne chemicals and particulate matter. This review discusses whether the rising prevalence of IBD in our society warrants the consideration of IBD patients as a specific population group in toxicological safety assessment. Various in vivo, ex vivo and in vitro models are discussed that can simulate hallmarks of IBD and may be used to study the effects of prevalent intestinal inflammation on the hazards of these various toxicants. In conclusion, risk assessments based on healthy individuals may not sufficiently cover IBD patient safety and it is suggested to consider this susceptible subgroup of the population in future toxicological assessments.
Collapse
Affiliation(s)
- Tom Walraven
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands.
| | - Mathias Busch
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| | - Jingxuan Wang
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| | - Joanne M Donkers
- Department of Metabolic Health Research, Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Marjolijn Duijvestein
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Evita van de Steeg
- Department of Metabolic Health Research, Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Nynke I Kramer
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
2
|
Özkan A, LoGrande NT, Feitor JF, Goyal G, Ingber DE. Intestinal organ chips for disease modelling and personalized medicine. Nat Rev Gastroenterol Hepatol 2024; 21:751-773. [PMID: 39192055 DOI: 10.1038/s41575-024-00968-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 08/29/2024]
Abstract
Alterations in intestinal structure, mechanics and physiology underlie acute and chronic intestinal conditions, many of which are influenced by dysregulation of microbiome, peristalsis, stroma or immune responses. Studying human intestinal physiology or pathophysiology is difficult in preclinical animal models because their microbiomes and immune systems differ from those of humans. Although advances in organoid culture partially overcome this challenge, intestinal organoids still lack crucial features that are necessary to study functions central to intestinal health and disease, such as digestion or fluid flow, as well as contributions from long-term effects of living microbiome, peristalsis and immune cells. Here, we review developments in organ-on-a-chip (organ chip) microfluidic culture models of the human intestine that are lined by epithelial cells and interfaced with other tissues (such as stroma or endothelium), which can experience both fluid flow and peristalsis-like motions. Organ chips offer powerful ways to model intestinal physiology and disease states for various human populations and individual patients, and can be used to gain new insight into underlying molecular and biophysical mechanisms of disease. They can also be used as preclinical tools to discover new drugs and then validate their therapeutic efficacy and safety in the same human-relevant model.
Collapse
Affiliation(s)
- Alican Özkan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Nina Teresa LoGrande
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Jessica F Feitor
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, USA.
| |
Collapse
|
3
|
Park J, Lee G, Park JK. Functional Assessment of a Bioprinted Immuno-Mimetic Peyer's Patch Recapitulating Gut-Associated Lymphoid Tissue. Adv Healthc Mater 2024:e2402722. [PMID: 39487612 DOI: 10.1002/adhm.202402722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/11/2024] [Indexed: 11/04/2024]
Abstract
Gut immune models have attracted much interest in better understanding the microbiome in the human gastrointestinal tract. The gut-associated lymphoid tissue (GALT) has complex structures that interact with microorganisms, including the intestinal monolayer as a physiological barrier and the Peyer's patch (PP) involved in the immune system. Although essential for studying GALT and microbiome interactions, current research often uses simplified models that only recapitulate some components. In this study, GALT is recapitulated to consider the morphology and function of lymphocyte-containing PP beneath the intestinal monolayer and to analyze microbiome interaction. Using the bioprinting technique, a dome-shaped structure array for the PP is fabricated, and epithelial cells are cocultured to form the intestinal monolayer. The developed GALT model shows stable cell differentiation on the hydrogel while exhibiting durability against lipopolysaccharides. It also exhibits increased responsiveness to Escherichia coli, as indicated by elevated nitric oxide levels. In addition, the model underscores the critical role of GALT in maintaining bacterial coexistence and in facilitating immune defense against foreign antigens through the secretion of immunoglobulin A by lymphocyte spheroids. The proposed GALT model is expected to provide significant insights into studying the gut-immune system complexity and microbiome.
Collapse
Affiliation(s)
- Jongho Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Gihyun Lee
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Je-Kyun Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
- KI for Health Science and Technology, KAIST Institutes (KI), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
- KI for NanoCentury, KAIST Institutes (KI), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| |
Collapse
|
4
|
Wang X, Zhu Y, Cheng Z, Zhang C, Liao Y, Liu B, Zhang D, Li Z, Fang Y. Emerging microfluidic gut-on-a-chip systems for drug development. Acta Biomater 2024; 188:48-64. [PMID: 39299625 DOI: 10.1016/j.actbio.2024.09.012] [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: 05/16/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
The gut is a vital organ that is central to the absorption and metabolic processing of orally administered drugs. While there have been many models developed with the goal of studying the absorption of drugs in the gut, these models fail to adequately recapitulate the diverse, complex gastrointestinal microenvironment. The recent emergence of microfluidic organ-on-a-chip technologies has provided a novel means of modeling the gut, yielding radical new insights into the structure of the gut and the mechanisms through which it shapes disease, with key implications for biomedical developmental efforts. Such organ-on-a-chip technologies have been demonstrated to exhibit greater cost-effectiveness, fewer ethical concerns, and a better ability to address inter-species differences in traditional animal models in the context of drug development. The present review offers an overview of recent developments in the reconstruction of gut structure and function in vitro using microfluidic gut-on-a-chip (GOC) systems, together with a discussion of the potential applications of these platforms in the context of drug development and the challenges and future prospects associated with this technology. STATEMENT OF SIGNIFICANCE: This paper outlines the characteristics of the different cell types most frequently used to construct microfluidic gut-on-a-chip models and the microfluidic devices employed for the study of drug absorption. And the applications of gut-related multichip coupling and disease modelling in the context of drug development is systematically reviewed. With the detailed summarization of microfluidic chip-based gut models and discussion of the prospective directions for practical application, this review will provide insights to the innovative design and application of microfluidic gut-on-a-chip for drug development.
Collapse
Affiliation(s)
- Xueqi Wang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, PR China
| | - Yuzhuo Zhu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, PR China
| | - Zhaoming Cheng
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, PR China
| | - Chuanjun Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, PR China
| | - Yumeng Liao
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Boshi Liu
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Di Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| | - Zheng Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| | - Yuxin Fang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin 301617, PR China; Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
5
|
Zhang Y, Wang Y, Zhang X, Wang P, Shi F, Zhang Z, Wang R, Wu D, She J. Gastrointestinal Self-Adaptive and Nutrient Self-Sufficient Akkermansia muciniphila-Gelatin Porous Microgels for Synergistic Therapy of Ulcerative Colitis. ACS NANO 2024; 18:26807-26827. [PMID: 39301762 DOI: 10.1021/acsnano.4c07658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
To realize effective and long-term synergistic therapy of ulcerative colitis (UC) with probiotics, we developed gastrointestinal self-adaptive and nutrient self-sufficient Akkermansia muciniphila (AKK)-gelatin porous microgels (AKK@GPMGs). In AKK@GPMGs, AKK was covered with sequential layers of proanthocyanidins (PAs), mucin (MUC), and phosphatidylcholine (PC) to obtain AKK@PAs-MUC-PC (AKK@PMP), and then encapsulated within the methacrylate-modified gelatin porous microgels. AKK@GPMGs provide sufficient mucus as a nutrition source for AKK and boost resistance to stomach acid by 30.49-fold, and colonization in the intestines is enhanced by 83.46 times. The microgels can be dissociated by matrix metalloproteinase at the inflammatory sites of the intestine, and release AKK@PMP, which acts as "band-aid" that adheres to the inflamed colon for a long time and offers improved synergistic therapy for UC. Compared to uncoated AKK, AKK@GPMGs increase reactive oxygen species scavenging capacity by 26.47 times, improve the intestinal mucus layer thickness by 5.63 times, increase the goblet cells abundance by 3.93 times, reduce intestinal permeability by 5.60 times and significantly enhance beneficial gut microbiota while repressing harmful microbiota. These results indicate that AKK@GPMGs can restore mucus layer and tight junction integrity, reduce inflammation and oxidative stress, and regulate gut microbiota homeostasis to effectively treat intestinal inflammation.
Collapse
Affiliation(s)
- Yujie Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, P.R. China
| | - Ya Wang
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Xiaojiang Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, P.R. China
| | - Pengqian Wang
- Department of Chemical Engineering, School of Water and Environment, Chang'an University, Xi'an 710064, P.R. China
| | - Feiyu Shi
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, P.R. China
| | - Zhe Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, P.R. China
| | - Ruochen Wang
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Daocheng Wu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
| | - Junjun She
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, P.R. China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| |
Collapse
|
6
|
Wu J, Zhang B, Liu X, Gu W, Xu F, Wang J, Liu Q, Wang R, Hu Y, Liu J, Ji X, Lv H, Li X, Peng L, Li X, Zhang Y, Wang S. An Intelligent Intestine-on-a-Chip for Rapid Screening of Probiotics with Relief-Enteritis Function. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2408485. [PMID: 39344562 DOI: 10.1002/adma.202408485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/11/2024] [Indexed: 10/01/2024]
Abstract
Screening probiotics with specific functions is essential for advancing probiotic research. Current screening methods primarily use animal studies or clinical trials, which are inefficient and costly in terms of time, money, and labor. An intelligent intestine-on-a-chip integrating machine learning (ML) is developed to screen relief-enteritis functional probiotics. A high-throughput microfluidic chip combined with environment control systems provides a standardized and scalable intestinal microenvironment for multiple probiotic cocultures. An unsupervised ML-based score analyzer is constructed to accurately, comprehensively, and efficiently evaluate interactions between 12 Bifidobacterium strains and host cells of the colitis model in the intestine-on-a-chips. The most effective contender, Bifidobacterium longum 3-14, is discovered to relieve intestinal inflammation and enhance epithelial barrier function in vitro and in vivo. A distinct advantage of this strategy is that it can intelligently differentiate small therapeutic variations in probiotic strains and prioritize their efficacies, allowing for economical, efficient, accurate functional probiotics screening.
Collapse
Affiliation(s)
- Jing Wu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Bowei Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiaoxia Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Wentao Gu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Fupei Xu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jin Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Qisijing Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Ruican Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yaozhong Hu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jingmin Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xuemeng Ji
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Huan Lv
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xinyang Li
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Lijun Peng
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiang Li
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yan Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Shuo Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| |
Collapse
|
7
|
Schaller ML, Sykes MM, Mecano J, Solanki S, Huang W, Rebernick RJ, Beydoun S, Wang E, Bugarin-Lapuz A, Shah YM, Leiser SF. Fmo5 plays a sex-specific role in goblet cell maturation and mucus barrier formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.588360. [PMID: 38645243 PMCID: PMC11030302 DOI: 10.1101/2024.04.05.588360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Background and Aims The intestine plays a key role in metabolism, nutrient and water absorption, and provides both physical and immunological defense against dietary and luminal antigens. The protective mucosal lining in the intestine is a critical component of intestinal barrier that when compromised, can lead to increased permeability, a defining characteristic of inflammatory bowel disease (IBD), among other intestinal diseases. Here, we define a new role for the flavin-containing monooxygenase (FMO) family of enzymes in maintaining a healthy intestinal epithelium. Methods Using Caenorhabditis elegans we measure intestinal barrier function, actin expression, and intestinal damage response. In mice, we utilize an intestine-specific, tamoxifen- inducible knockout model of the mammalian homolog of Cefmo-2 , Fmo5, and assess histology, mucus barrier thickness, and goblet cell physiology. We also treat mice with the ER chaperone Tauroursodeoxycholic acid (TUDCA). Results In nematodes, we find Cefmo-2 is necessary and sufficient for intestinal barrier function, intestinal actin expression, and is induced by intestinal damage. In mice, we find striking changes to the intestine within two weeks following Fmo5 disruption. Alterations include sex-dependent changes in colon epithelial histology, goblet cell localization, and mucus barrier formation. These changes are significantly more severe in female mice, mirroring differences observed in IBD patients. Furthermore, we find increased protein folding stress in Fmo5 knockout animals and successfully rescue the severe female phenotype with addition of a chemical ER chaperone. Conclusions Together, our results identify a highly conserved and novel role for Fmo5 in the mammalian intestine and support a key role for Fmo5 in maintenance of ER/protein homeostasis and proper mucus barrier formation.
Collapse
|
8
|
Zhang X, Su R, Wang H, Wu R, Fan Y, Bin Z, Gao C, Wang C. The promise of Synovial Joint-on-a-Chip in rheumatoid arthritis. Front Immunol 2024; 15:1408501. [PMID: 39324139 PMCID: PMC11422143 DOI: 10.3389/fimmu.2024.1408501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Rheumatoid arthritis (RA) affects millions of people worldwide, but there are limited drugs available to treat it, so acquiring a more comprehensive comprehension of the underlying reasons and mechanisms behind inflammation is crucial, as well as developing novel therapeutic approaches to manage it and mitigate or forestall associated harm. It is evident that current in vitro models cannot faithfully replicate all aspects of joint diseases, which makes them ineffective as tools for disease research and drug testing. Organ-on-a-chip (OoC) technology is an innovative platform that can mimic the microenvironment and physiological state of living tissues more realistically than traditional methods by simulating the spatial arrangement of cells and interorgan communication. This technology allows for the precise control of fluid flow, nutrient exchange, and the transmission of physicochemical signals, such as bioelectrical, mechanical stimulation and shear force. In addition, the integration of cutting-edge technologies like sensors, 3D printing, and artificial intelligence enhances the capabilities of these models. Here, we delve into OoC models with a particular focus on Synovial Joints-on-a-Chip, where we outline their structure and function, highlighting the potential of the model to advance our understanding of RA. We integrate the actual evidence regarding various OoC models and their possible integration for multisystem disease study in RA research for the first time and introduce the prospects and opportunities of the chip in RA etiology and pathological mechanism research, drug research, disease prevention and human precision medicine. Although many challenges remain, OoC holds great promise as an in vitro model that approaches physiology and dynamics.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Rui Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Hui Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Ruihe Wu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Yuxin Fan
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Zexuan Bin
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Chong Gao
- Pathology, Joint Program in Transfusion Medicine, Brigham and Women’s Hospital/Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Caihong Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| |
Collapse
|
9
|
Kang SJ, Lee JH, Rhee WJ. Engineered plant-derived extracellular vesicles for targeted regulation and treatment of colitis-associated inflammation. Theranostics 2024; 14:5643-5661. [PMID: 39310109 PMCID: PMC11413791 DOI: 10.7150/thno.97139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/31/2024] [Indexed: 09/25/2024] Open
Abstract
Rationale: Inflammatory bowel disease (IBD) is a chronic disorder characterized by persistent inflammation of the gastrointestinal tract. Due to the elusive causes and complex mechanisms of this disorder, the development of highly effective therapeutic drugs is crucial. Extracellular vesicles (EVs) are small membrane-bound structures released by cells into the surrounding environment. Recent research has witnessed a substantial surge in the utilization of plant-derived EVs that offer advantages such as high productivity, low production costs, diverse biological functions, and low cytotoxicity. Herein, Red cabbage-derived EVs (Rabex) were investigated and engineered as potential therapeutic agents for IBD. Methods: Rabex was engineered by surface conjugation with hyaluronic acid (t-Rabex) to simultaneously enhance the targeting of intestinal epithelial and immune cells, thereby improving their therapeutic targeting and efficacy. The properties and therapeutic potential of t-Rabex were assessed through both in vitro studies and in vivo experiments, focusing on their capacity to reach the gastrointestinal tract and exert a therapeutic effect compared to unmodified Rabex. Results: Rabex exhibited dual functions, including the suppression of inflammation in macrophages and promotion of colon epithelial cell regeneration, both of which are critical for effective IBD treatment. In vitro and in vivo studies of t-Rabex have demonstrated its superior targeting efficiency to the gastrointestinal tract and therapeutic efficacy compared to Rabex, making it a promising and more effective IBD treatment. Understanding the mechanism of action of t-Rabex in colonic tissues highlighted its anti-inflammatory, antioxidative, and tight-junction maintenance properties. Conclusions: These findings underscore the potential of t-Rabex as a precise therapeutic agent for IBD and shed light on the diverse applications of plant-derived EVs.
Collapse
Affiliation(s)
- Su Jin Kang
- Department of Bioengineering and Nano-Bioengineering, Incheon National University Incheon 22012, Republic of Korea
| | - Jeong Hyun Lee
- Department of Bioengineering and Nano-Bioengineering, Incheon National University Incheon 22012, Republic of Korea
| | - Won Jong Rhee
- Department of Bioengineering and Nano-Bioengineering, Incheon National University Incheon 22012, Republic of Korea
- Division of Bioengineering, Incheon National University Incheon 22012, Republic of Korea
- Research Center for Bio Materials & Process Development, Incheon National University, Incheon 22012, Republic of Korea
| |
Collapse
|
10
|
Xiao N, Zhang X, Xi Y, Li Z, Wei Y, Shen J, Wang L, Qin D, Xie Z, Li Z. Study on the effects of intestinal flora on gouty arthritis. Front Cell Infect Microbiol 2024; 14:1341953. [PMID: 39176260 PMCID: PMC11339034 DOI: 10.3389/fcimb.2024.1341953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 07/22/2024] [Indexed: 08/24/2024] Open
Abstract
Gouty arthritis (GA), a metabolic and immunologic disease, primarily affects joints. Dysbiosis of intestinal flora is an important cause of GA. The metabolic disorders of intestinal flora leading to GA and immune disorders might play an important role in patients with hyperuricemia and established GA. However, the exact mechanisms, through which the dysbiosis of intestinal flora causes the development of GA, are not fully understood yet. Moreover, several therapies commonly used to treat GA might alter the intestinal flora, suggesting that modulation of the intestinal flora might help prevent or treat GA. Therefore, a better understanding of the changes in the intestinal flora of GA patients might facilitate the discovery of new diagnostic and therapeutic approaches. The current review article discusses the effects of intestinal flora dysbiosis on the pathogenesis of GA and the cross-regulatory effects between gut flora and drugs for treating GA. This article also highlights the modulatory effects of gut flora by traditional Chinese medicine (TCM) to lower uric acid levels and relieve joint pain as well as provides a summary and outlook, which might help guide future research efforts.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Dongdong Qin
- Yunnan University of Chinese Medicine, Kunming, China
| | - Zhaohu Xie
- Yunnan University of Chinese Medicine, Kunming, China
| | - Zhaofu Li
- Yunnan University of Chinese Medicine, Kunming, China
| |
Collapse
|
11
|
Liu H, Wang Y, Zhang X, Zhang M, Wang P, Shang J, Li Z, Gong L, Guo J, Sun W, Pi J, Li X, Ding W, Wang D, Li Z, Zhang J, Wang L, Geng X, Yang R, Zhou P, Tang W, Zhang X, Chen C, Yang S, Qin J. Standard: human intestine-on-a-chip. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:16. [PMID: 39101982 DOI: 10.1186/s13619-024-00198-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/17/2024] [Indexed: 08/06/2024]
Abstract
Organs-on-chips are microphysiological systems that allow to replicate the key functions of human organs and accelerate the innovation in life sciences including disease modeling, drug development, and precision medicine. However, due to the lack of standards in their definition, structural design, cell source, model construction, and functional validation, a wide range of translational application of organs-on-chips remains a challenging. "Organs-on-chips: Intestine" is the first group standard on human intestine-on-a-chip in China, jointly agreed and released by the experts from the Chinese Society of Biotechnology on 29th April 2024. This standard specifies the scope, terminology, definitions, technical requirements, detection methods, and quality control in building the human intestinal model on a chip. The publication of this group standard will guide the institutional establishment, acceptance and execution of proper practical protocols and accelerate the international standardization of intestine-on-a-chip for translational applications.
Collapse
Affiliation(s)
- Haitao Liu
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yaqing Wang
- University of Science and Technology of China, Hefei, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, China
| | - Xu Zhang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Min Zhang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Peng Wang
- University of Science and Technology of China, Hefei, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, China
| | - Jing Shang
- China Pharmaceutical University, Nanjing, China
| | - Zhongqiang Li
- China National Institute of Standardization, Beijing, China
| | - Likun Gong
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jiabin Guo
- Chinese People's Liberation Army (PLA) Center for Disease Control and Prevention, Beijing, China
| | - Wei Sun
- Tsinghua University, Beijing, China
| | - Jingbo Pi
- China Medical University, Shenyang, China
| | - Xianliang Li
- Department of HBP Surgery, Beijing Chao Yang Hospital, the Capital Medical University, Beijing, China
| | - Wei Ding
- SPH KDL Health Beijing, Shanghai Pharma, Beijing, China
| | - Dianbing Wang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | | | - Jingzhong Zhang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Lan Wang
- Dalian Xin'en Medical Technology Co., LTD, Dalian, China
| | - Xingchao Geng
- National Institutes for Food and Drug Control, Beijing, China
| | - Ruifu Yang
- Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Pingkun Zhou
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Wanjin Tang
- China National Institute of Standardization, Beijing, China
| | - Xian'en Zhang
- Shenzhen University of Advanced Technology, Shenzhen, China
| | - Chunying Chen
- National Center for Nanoscience and Technology, Beijing, China
| | - Shengli Yang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.
| |
Collapse
|
12
|
Kim R, Sung JH. Recent Advances in Gut- and Gut-Organ-Axis-on-a-Chip Models. Adv Healthc Mater 2024; 13:e2302777. [PMID: 38243887 DOI: 10.1002/adhm.202302777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/21/2023] [Indexed: 01/22/2024]
Abstract
The human gut extracts nutrients from the diet while forming the largest barrier against the outer environment. In addition, the gut actively maintains homeostasis through intricate interactions with the gut microbes, the immune system, the enteric nervous system, and other organs. These interactions influence digestive health and, furthermore, play crucial roles in systemic health and disease. Given its primary role in absorbing and metabolizing orally administered drugs, there is significant interest in the development of preclinical in vitro model systems that can accurately emulate the intestine in vivo. A gut-on-a-chip system holds great potential as a testing and screening platform because of its ability to emulate the physiological aspects of in vivo tissues and expandability to incorporate and combine with other organs. This review aims to identify the key physiological features of the human gut that need to be incorporated to build more accurate preclinical models and highlights the recent progress in gut-on-a-chip systems and competing technologies toward building more physiologically relevant preclinical model systems. Furthermore, various efforts to construct multi-organ systems with the gut, called gut-organ-axis-on-a-chip models, are discussed. In vitro gut models with physiological relevance can provide valuable platforms for bridging the gap between preclinical and clinical studies.
Collapse
Affiliation(s)
- Raehyun Kim
- Department of Biological and Chemical Engineering, Hongik University, Sejong, 30016, Republic of Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea
| |
Collapse
|
13
|
Calzuola ST, Newman G, Feaugas T, Perrault CM, Blondé JB, Roy E, Porrini C, Stojanovic GM, Vidic J. Membrane-based microfluidic systems for medical and biological applications. LAB ON A CHIP 2024; 24:3579-3603. [PMID: 38954466 DOI: 10.1039/d4lc00251b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Microfluidic devices with integrated membranes that enable control of mass transport in constrained environments have shown considerable growth over the last decade. Membranes are a key component in several industrial processes such as chemical, pharmaceutical, biotechnological, food, and metallurgy separation processes as well as waste management applications, allowing for modular and compact systems. Moreover, the miniaturization of a process through microfluidic devices leads to process intensification together with reagents, waste and cost reduction, and energy and space savings. The combination of membrane technology and microfluidic devices allows therefore magnification of their respective advantages, providing more valuable solutions not only for industrial processes but also for reproducing biological processes. This review focuses on membrane-based microfluidic devices for biomedical science with an emphasis on microfluidic artificial organs and organs-on-chip. We provide the basic concepts of membrane technology and the laws governing mass transport. The role of the membrane in biomedical microfluidic devices, along with the required properties, available materials, and current challenges are summarized. We believe that the present review may be a starting point and a resource for researchers who aim to replicate a biological phenomenon on-chip by applying membrane technology, for moving forward the biomedical applications.
Collapse
Affiliation(s)
- Silvia Tea Calzuola
- UMR7646 Laboratoire d'hydrodynamique (LadHyX), Ecole Polytechnique, Palaiseau, France.
- Eden Tech, Paris, France
| | - Gwenyth Newman
- Eden Tech, Paris, France
- Department of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Thomas Feaugas
- Eden Tech, Paris, France
- Department of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Milan, Italy
| | | | | | | | | | - Goran M Stojanovic
- Faculty of Technical Sciences, University of Novi Sad, T. D. Obradovića 6, 21000 Novi Sad, Serbia
| | - Jasmina Vidic
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
14
|
Villegas-Novoa C, Wang Y, Sims CE, Allbritton NL. Creation of a spatially complex mucus bilayer on an in vitro colon model. Sci Rep 2024; 14:16849. [PMID: 39039235 PMCID: PMC11263341 DOI: 10.1038/s41598-024-67591-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/12/2024] [Indexed: 07/24/2024] Open
Abstract
The colonic epithelium is comprised of three-dimensional crypts (3D) lined with mucus secreted by a heterogeneous population of goblet cells. In this study, we report the formation of a long-lived, and self-renewing replica of human 3D crypts with a mucus layer patterned in the X-Y-Z dimensions. Primary colon cells were cultured on a shaped scaffold under an air-liquid interface to yield architecturally accurate crypts with a mucus bilayer (605 ± 180 μm thick) possessing an inner (149 ± 50 μm) and outer (435 ± 111 μm) region. Lectins with distinct carbohydrate-binding preferences demonstrated that the mucus in the intercrypt regions was chemically distinct from that above and within the crypts replicating in vivo chemical patterning. Constitutive mucus secretion ejected beads from crypt lumens in 8-10 days, while agonist-stimulated secretion increased mucus thickness by 17-fold in 8 h. The tissue was long-lived, > 50 days, the longest time assessed. In conclusion, the in vitro mucus replicated key physiology of the human mucus, including the bilayer (Z) structure and intercrypt-crypt (X-Y) zones, constitutive mucus flow, spatially complex chemical attributes, and mucus secretion response to stimulation, with the potential to reveal local and global determinants of mucus function and its breakdown in disease.
Collapse
Affiliation(s)
| | - Yuli Wang
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Christopher E Sims
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Nancy L Allbritton
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
15
|
Wang H, Li X, Shi P, You X, Zhao G. Establishment and evaluation of on-chip intestinal barrier biosystems based on microfluidic techniques. Mater Today Bio 2024; 26:101079. [PMID: 38774450 PMCID: PMC11107260 DOI: 10.1016/j.mtbio.2024.101079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/17/2024] [Accepted: 05/01/2024] [Indexed: 05/24/2024] Open
Abstract
As a booming engineering technology, the microfluidic chip has been widely applied for replicating the complexity of human intestinal micro-physiological ecosystems in vitro. Biosensors, 3D imaging, and multi-omics have been applied to engineer more sophisticated intestinal barrier-on-chip platforms, allowing the improved monitoring of physiological processes and enhancing chip performance. In this review, we report cutting-edge advances in the microfluidic techniques applied for the establishment and evaluation of intestinal barrier platforms. We discuss different design principles and microfabrication strategies for the establishment of microfluidic gut barrier models in vitro. Further, we comprehensively cover the complex cell types (e.g., epithelium, intestinal organoids, endothelium, microbes, and immune cells) and controllable extracellular microenvironment parameters (e.g., oxygen gradient, peristalsis, bioflow, and gut-organ axis) used to recapitulate the main structural and functional complexity of gut barriers. We also present the current multidisciplinary technologies and indicators used for evaluating the morphological structure and barrier integrity of established gut barrier models in vitro. Finally, we highlight the challenges and future perspectives for accelerating the broader applications of these platforms in disease simulation, drug development, and personalized medicine. Hence, this review provides a comprehensive guide for the development and evaluation of microfluidic-based gut barrier platforms.
Collapse
Affiliation(s)
- Hui Wang
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, China
| | - Xiangyang Li
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, 471023, China
- Haihe Laboratory of Synthetic Biology, Tianjin, 300308, China
| | - Pengcheng Shi
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, 471023, China
| | - Xiaoyan You
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, China
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, 471023, China
| | - Guoping Zhao
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308, China
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China
- CAS-Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
16
|
Tian Y, Wu G, Zhao X, Zhang H, Ren M, Song X, Chang H, Jing Z. Probiotics combined with atorvastatin administration in the treatment of hyperlipidemia: A randomized, double-blind, placebo-controlled clinical trial. Medicine (Baltimore) 2024; 103:e37883. [PMID: 38788020 PMCID: PMC11124713 DOI: 10.1097/md.0000000000037883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/21/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Hyperlipidemia is a common feature of chronic diseases. The aim of this work was designed to assess the role of probiotics (Lactobacillus casei Zhang, Bifidobactetium animalis subsp. lactis V9, and Lactobacillus plantarum P-8) in the treatment of hyperlipidemia. METHODS Thirty three patients with hyperlipidemia were randomly divided into a probiotic group (n = 18) and a control group (n = 15). The probiotic group was administered probiotics (2 g once daily) and atorvastatin 20 mg (once daily), and the control group was administered a placebo (2 g once daily) and atorvastatin 20 mg (once daily). Serum and fecal samples were gathered for subsequent analyses. RESULTS Time had a significant effect on the total cholesterol (TC), triglycerides (TG), and low-density lipoprotein-cholesterol (LDL-C) levels in the probiotic and control groups (P < .05). The gut microbial abundance in the probiotic group was markedly higher than that in the control group following 3-month probiotic treatment (P < .05). At the phylum level, probiotics exerted no notable effects on the relative abundance of Firmicutes, Bacteroidetes, and Actinobacteria but elevated that of Tenericutes and reduced Proteobacteria. At the genus level, probiotics increased the relative abundance of Bifidobacterium, Lactobacillus, and Akkermansia, and decreased that of Escherichia, Eggerthella, and Sutterella relative to the control group in months 1, 2, and 3 (P < .05). CONCLUSIONS Probiotics optimize the gut microbiota structure and decrease the amount of harmful bacteria in patients with hyperlipidemia. Probiotics can influence the composition of gut microorganisms and increase their diversity and abundance in vivo. It is recommended to use probiotics combined with atorvastatin to treat patients with hyperlipidemia.
Collapse
Affiliation(s)
- Yingjie Tian
- Department of Cardiology, Heart Center, Inner Mongolia People’s Hospital, Hohhot, People’s Republic of China
- Inner Mongolia Cardiovascular Disease Clinical Research Center, Hohhot, People’s Republic of China
| | - Guang Wu
- Department of Cardiology, Heart Center, Inner Mongolia People’s Hospital, Hohhot, People’s Republic of China
| | - Xingsheng Zhao
- Department of Cardiology, Heart Center, Inner Mongolia People’s Hospital, Hohhot, People’s Republic of China
- Inner Mongolia Cardiovascular Disease Clinical Research Center, Hohhot, People’s Republic of China
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| | - Maojia Ren
- Department of Cardiology, Heart Center, Inner Mongolia People’s Hospital, Hohhot, People’s Republic of China
| | - Xiaopeng Song
- Department of Cardiology, Heart Center, Inner Mongolia People’s Hospital, Hohhot, People’s Republic of China
| | - Hao Chang
- Department of Cardiology, Heart Center, Inner Mongolia People’s Hospital, Hohhot, People’s Republic of China
| | - Zelin Jing
- Department of Neurosurgery, Hohhot First Hospital, Hohhot, People’s Republic of China
| |
Collapse
|
17
|
Morrison AI, Sjoerds MJ, Vonk LA, Gibbs S, Koning JJ. In vitro immunity: an overview of immunocompetent organ-on-chip models. Front Immunol 2024; 15:1373186. [PMID: 38835750 PMCID: PMC11148285 DOI: 10.3389/fimmu.2024.1373186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
Impressive advances have been made to replicate human physiology in vitro over the last few years due to the growth of the organ-on-chip (OoC) field in both industrial and academic settings. OoCs are a type of microphysiological system (MPS) that imitates functional and dynamic aspects of native human organ biology on a microfluidic device. Organoids and organotypic models, ranging in their complexity from simple single-cell to complex multi-cell type constructs, are being incorporated into OoC microfluidic devices to better mimic human physiology. OoC technology has now progressed to the stage at which it has received official recognition by the Food and Drug Administration (FDA) for use as an alternative to standard procedures in drug development, such as animal studies and traditional in vitro assays. However, an area that is still lagging behind is the incorporation of the immune system, which is a critical element required to investigate human health and disease. In this review, we summarise the progress made to integrate human immunology into various OoC systems, specifically focusing on models related to organ barriers and lymphoid organs. These models utilise microfluidic devices that are either commercially available or custom-made. This review explores the difference between the use of innate and adaptive immune cells and their role for modelling organ-specific diseases in OoCs. Immunocompetent multi-OoC models are also highlighted and the extent to which they recapitulate systemic physiology is discussed. Together, the aim of this review is to describe the current state of immune-OoCs, the limitations and the future perspectives needed to improve the field.
Collapse
Affiliation(s)
- Andrew I. Morrison
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
| | - Mirthe J. Sjoerds
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Leander A. Vonk
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Susan Gibbs
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, Netherlands
| | - Jasper J. Koning
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
| |
Collapse
|
18
|
Liang H, Tao S, Wang Y, Zhao J, Yan C, Wu Y, Liu N, Qin Y. Astragalus polysaccharide: implication for intestinal barrier, anti-inflammation, and animal production. Front Nutr 2024; 11:1364739. [PMID: 38757131 PMCID: PMC11096541 DOI: 10.3389/fnut.2024.1364739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024] Open
Abstract
Intestine is responsible for nutrients absorption and plays a key role in defending against various dietary allergens, antigens, toxins, and pathogens. Accumulating evidence reported a critical role of intestine in maintaining animal and human health. Since the use of antibiotics as growth promoters in animal feed has been restricted in many countries, alternatives to antibiotics have been globally investigated, and polysaccharides are considered as environmentally friendly and promising alternatives to improve intestinal health, which has become a research hotspot due to its antibiotic substitution effect. Astragalus polysaccharide (APS), a biological macromolecule, is extracted from astragalus and has been reported to exhibit complex biological activities involved in intestinal barrier integrity maintenance, intestinal microbiota regulation, short-chain fatty acids (SCFAs) production, and immune response regulation, which are critical for intestine health. The biological activity of APS is related to its chemical structure. In this review, we outlined the source and structure of APS, highlighted recent findings on the regulation of APS on physical barrier, biochemical barrier, immunological barrier, and immune response as well as the latest progress of APS as an antibiotic substitute in animal production. We hope this review could provide scientific basis and new insights for the application of APS in nutrition, clinical medicine and health by understanding particular effects of APS on intestine health, anti-inflammation, and animal production.
Collapse
Affiliation(s)
- Hui Liang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Siming Tao
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Yanya Wang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Jing Zhao
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Chang Yan
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Yingjie Wu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Ning Liu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Yinghe Qin
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| |
Collapse
|
19
|
Lee J, Menon N, Lim CT. Dissecting Gut-Microbial Community Interactions using a Gut Microbiome-on-a-Chip. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302113. [PMID: 38414327 PMCID: PMC11132043 DOI: 10.1002/advs.202302113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/21/2023] [Indexed: 02/29/2024]
Abstract
While the human gut microbiota has a significant impact on gut health and disease, understanding of the roles of gut microbes, interactions, and collective impact of gut microbes on various aspects of human gut health is limited by the lack of suitable in vitro model system that can accurately replicate gut-like environment and enable the close visualization on causal and mechanistic relationships between microbial constitutents and the gut. , In this study, we present a scalable Gut Microbiome-on-a-Chip (GMoC) with great imaging capability and scalability, providing a physiologically relevant dynamic gut-microbes interfaces. This chip features a reproducible 3D stratified gut epithelium derived from Caco-2 cells (µGut), mimicking key intestinal architecture, functions, and cellular complexity, providing a physiolocially relevant gut environment for microbes residing in the gut. Incorporating tumorigenic bacteria, enterotoxigenic Bacteroides fragilis (ETBF), into the GMoC enable the observation of pathogenic behaviors of ETBF, leading to µGut disruption and pro-tumorigenic signaling activations. Pre-treating the µGut with a beneficial gut microbe Lactobacillus spp., effectively prevent ETBF-mediated gut pathogenesis, preserving the healthy state of the µGut through competition-mediated colonization resistance. The GMoC holds potential as a valuable tool for exploring unknown roles of gut microbes in microbe-induced pathogenesis and microbe-based therapeutic development.
Collapse
Affiliation(s)
- Jeeyeon Lee
- Institute for Health Innovation and Technology (iHealthtech)National University of SingaporeSingapore117599Singapore
| | - Nishanth Menon
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Chwee Teck Lim
- Institute for Health Innovation and Technology (iHealthtech)National University of SingaporeSingapore117599Singapore
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Mechanobiology InstituteNational University of SingaporeSingapore117411Singapore
| |
Collapse
|
20
|
Sampath C, Chukkapalli SS, Raju AV, Alluri LSC, Srisai D, Gangula PR. Cinnamaldehyde Protects against P. gingivalis Induced Intestinal Epithelial Barrier Dysfunction in IEC-6 Cells via the PI3K/Akt-Mediated NO/Nrf2 Signaling Pathway. Int J Mol Sci 2024; 25:4734. [PMID: 38731952 PMCID: PMC11083591 DOI: 10.3390/ijms25094734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Porphyromonas gingivalis (Pg), a Gram-negative oral pathogen, promotes and accelerates periodontitis-associated gut disorders. Intestinal epithelial barrier dysfunction is crucial in the pathogenesis of intestinal and systemic diseases. In this study, we sought to elucidate the protective role of cinnamaldehyde (CNM, an activator of Nrf2) against P. gingivalis (W83) and Pg-derived lipopolysaccharide (Pg-LPS) induced intestinal epithelial barrier dysfunction via antioxidative mechanisms in IEC-6 cells. IEC-6 (ATCC, CRL-1592) cells were pretreated with or without CNM (100 µM), in the presence or absence of P. gingivalis (strain W83, 109 MOI) or Pg-LPS (1, 10, and 100 µg/mL), respectively, between 0-72 h time points by adopting a co-culture method. Intestinal barrier function, cytokine secretion, and intestinal oxidative stress protein markers were analyzed. P. gingivalis or Pg-LPS significantly (p < 0.05) increased reactive oxygen species (ROS) and malondialdehyde (MDA) levels expressing oxidative stress damage. Pg-LPS, as well as Pg alone, induces inflammatory cytokines via TLR-4 signaling. Furthermore, infection reduced Nrf2 and NAD(P)H quinone dehydrogenase 1 (NQO1). Interestingly, inducible nitric oxide synthase (iNOS) protein expression significantly (p < 0.05) increased with Pg-LPS or Pg infection, with elevated levels of nitric oxide (NO). CNM treatment suppressed both Pg- and Pg-LPS-induced intestinal oxidative stress damage by reducing ROS, MDA, and NO production. Furthermore, CNM treatment significantly upregulated the expression of tight junction proteins via increasing the phosphorylation levels of PI3K/Akt/Nrf2 suppressing inflammatory cytokines. CNM protected against Pg infection-induced intestinal epithelial barrier dysfunction by activating the PI3K/Akt-mediated Nrf2 signaling pathway in IEC-6 cells.
Collapse
Affiliation(s)
- Chethan Sampath
- Department of Diabetes, Metabolism and Endocrinology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of ODS & Research, School of Dentistry, Meharry Medical College, Nashville, TN 37208, USA;
| | - Sasanka S. Chukkapalli
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA;
| | - Abhinav V. Raju
- College of Osteopathic Medicine, Kansas City University, Kansas City, MO 64106, USA;
| | | | - Dollada Srisai
- Department of ODS & Research, School of Dentistry, Meharry Medical College, Nashville, TN 37208, USA;
| | - Pandu R. Gangula
- Department of ODS & Research, School of Dentistry, Meharry Medical College, Nashville, TN 37208, USA;
| |
Collapse
|
21
|
Xiao N, Xie Z, He Z, Xu Y, Zhen S, Wei Y, Zhang X, Shen J, Wang J, Tian Y, Zuo J, Peng J, Li Z. Pathogenesis of gout: Exploring more therapeutic target. Int J Rheum Dis 2024; 27:e15147. [PMID: 38644732 DOI: 10.1111/1756-185x.15147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/23/2024]
Abstract
Gout is a chronic metabolic and immune disease, and its specific pathogenesis is still unclear. When the serum uric acid exceeds its saturation in the blood or tissue fluid, it is converted to monosodium urate crystals, which lead to acute arthritis of varying degrees, urinary stones, or irreversible peripheral joint damage, and in severe cases, impairment of vital organ function. Gout flare is a clinically significant state of acute inflammation in gout. The current treatment is mostly anti-inflammatory analgesics, which have numerous side effects with limited treatment methods. Gout pathogenesis involves many aspects. Therefore, exploring gout pathogenesis from multiple perspectives is conducive to identifying more therapeutic targets and providing safer and more effective alternative treatment options for patients with gout flare. Thus, this article is of great significance for further exploring the pathogenesis of gout. The author summarizes the pathogenesis of gout from four aspects: signaling pathways, inflammatory factors, intestinal flora, and programmed cell death, focusing on exploring more new therapeutic targets.
Collapse
Affiliation(s)
- Niqin Xiao
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Zhaohu Xie
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Zhiyan He
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Yundong Xu
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Shuyu Zhen
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Yuanyuan Wei
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Xiaoyu Zhang
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Jiayan Shen
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Jian Wang
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Yadan Tian
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Jinlian Zuo
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Jiangyun Peng
- The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Zhaofu Li
- Yunnan University of Chinese Medicine, Kunming, China
| |
Collapse
|
22
|
Zhang J, Huang YJ, Trapecar M, Wright C, Schneider K, Kemmitt J, Hernandez-Gordillo V, Yoon JY, Poyet M, Alm EJ, Breault DT, Trumper DL, Griffith LG. An immune-competent human gut microphysiological system enables inflammation-modulation by Faecalibacterium prausnitzii. NPJ Biofilms Microbiomes 2024; 10:31. [PMID: 38553449 PMCID: PMC10980819 DOI: 10.1038/s41522-024-00501-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 03/11/2024] [Indexed: 04/02/2024] Open
Abstract
Crosstalk of microbes with human gut epithelia and immune cells is crucial for gut health. However, there is no existing system for a long-term co-culture of human innate immune cells with epithelium and oxygen-intolerant commensal microbes, hindering the understanding of microbe-immune interactions in a controlled manner. Here, we established a gut epithelium-microbe-immune (GuMI) microphysiological system to maintain the long-term continuous co-culture of Faecalibacterium prausnitzii/Faecalibacterium duncaniae with colonic epithelium, antigen-presenting cells (APCs, herein dendritic cells and macrophages), and CD4+ naive T cells circulating underneath the colonic epithelium. In GuMI-APC condition, multiplex cytokine assays suggested that APCs contribute to the elevated level of cytokines and chemokines secreted into both apical and basolateral compartments compared to GuMI condition that lacks APC. In GuMI-APC with F. prausnitzii (GuMI-APC-FP), F. prausnitzii increased the transcription of pro-inflammatory genes such as toll-like receptor 1 (TLR1) and interferon alpha 1 (IFNA1) in the colonic epithelium, without a significant effect on cytokine secretion, compared to the GuMI-APC without bacteria (GuMI-APC-NB). In contrast, in the presence of CD4+ naive T cells (GuMI-APCT-FP), TLR1, IFNA1, and IDO1 transcription levels decreased with a simultaneous increase in F. prausnitzii-induced secretion of pro-inflammatory cytokines (e.g., IL8) compared to GuMI-APC-FP that lacks T cells. These results highlight the contribution of individual innate immune cells in regulating the immune response triggered by the gut commensal F. prausnitzii. The integration of defined populations of immune cells in the gut microphysiological system demonstrated the usefulness of GuMI physiomimetic platform to study microbe-epithelial-immune interactions in healthy and disease conditions.
Collapse
Affiliation(s)
- Jianbo Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands.
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam UMC, Location Academic Medical Center, Amsterdam, the Netherlands.
| | - Yu-Ja Huang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Martin Trapecar
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Charles Wright
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kirsten Schneider
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - John Kemmitt
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Jun Young Yoon
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Yonsei University, Seoul, South Korea
| | - Mathilde Poyet
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute of Experimental Medicine, University of Kiel, Kiel, Germany
| | - Eric J Alm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David T Breault
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - David L Trumper
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
23
|
Cho DE, Hong JP, Kim Y, Sim JY, Kim HS, Kim SR, Lee B, Cho HS, Cho IH, Shin S, Yeom M, Kwon SK, Lee IS, Park H, Kim K, Hahm DH. Role of gut-derived bacterial lipopolysaccharide and peripheral TLR4 in immobilization stress-induced itch aggravation in a mouse model of atopic dermatitis. Sci Rep 2024; 14:6263. [PMID: 38491103 PMCID: PMC10942979 DOI: 10.1038/s41598-024-56936-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/12/2024] [Indexed: 03/18/2024] Open
Abstract
Psychological stress and intestinal leakage are key factors in atopic dermatitis (AD) recurrence and exacerbation. Here, we demonstrate the mechanism underlying bacterial translocation across intestinal epithelial barrier damaged due to stress and further aggravation of trimellitic anhydride (TMA)-induced itch, which remain unclear, in AD mice. Immobilization (IMO) stress exacerbated scratching bouts and colon histological damage, and increased serum corticosterone and lipopolysaccharide (LPS). Orally administered fluorescein isothiocyanate (FITC)-dextran and surgically injected (into the colon) Cy5.5-conjugated LPS were detected in the serum and skin after IMO stress, respectively. The relative abundance of aerobic or facultative anaerobic bacteria was increased in the colon mucus layer, and Lactobacillus murinus, E. coli, Staphylococcus nepalensis, and several strains of Bacillus sp. were isolated from the spleens and mesenteric lymph nodes. Oral antibiotics or intestinal permeability blockers, such as lubiprostone (Lu), 2,4,6-triaminopyrimidine (TAP) and ML-7, inhibited IMO stress-associated itch; however, it was reinduced through intradermal or i.p. injection of LPS without IMO stress. I.p. injection of TAK-242 (resatorvid), a TLR4 inhibitor, abrogated IMO stress-associated itch, which was also confirmed in TLR4-KO mice. IMO stress alone did not cause itch in naïve mice. IMO stress-induced itch aggravation in TMA-treated AD mice might be attributed to the translocation of gut-derived bacterial cells and LPS, which activates peripheral TLR4 signaling.
Collapse
Affiliation(s)
- Da-Eun Cho
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Joon-Pyo Hong
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Yoongeun Kim
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ju Yeon Sim
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Heenam Stanley Kim
- Division of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Song-Rae Kim
- Chuncheon Center, Korea Basic Science Institute (KBSI), Chuncheon, 24341, Republic of Korea
| | - Bombi Lee
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyo-Sung Cho
- Department of Korean Medical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ik-Hyun Cho
- Department of Korean Medical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Sooan Shin
- ACCURIEBIO Co., IRIS Lab., 6th Floor, Sangwon 12-gil 34, Seongdong-gu, Seoul, 04790, Republic of Korea
| | - Mijung Yeom
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Soon-Kyeong Kwon
- Division of Applied Life Science (Brain Korea 21 PLUS), Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - In-Seon Lee
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Korean Medical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hijoon Park
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Korean Medical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Kyuseok Kim
- Department of Ophthalmology, Otorhinolaryngology and Dermatology of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Dae-Hyun Hahm
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
24
|
Nguyen OTP, Misun PM, Hierlemann A, Lohasz C. A Versatile Intestine-on-Chip System for Deciphering the Immunopathogenesis of Inflammatory Bowel Disease. Adv Healthc Mater 2024; 13:e2302454. [PMID: 38253407 PMCID: PMC11468350 DOI: 10.1002/adhm.202302454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 12/21/2023] [Indexed: 01/24/2024]
Abstract
The multifactorial nature of inflammatory bowel disease (IBD) necessitates reliable and practical experimental models to elucidate its etiology and pathogenesis. To model the intestinal microenvironment at the onset of IBD in vitro, it is important to incorporate relevant cellular and noncellular components before inducing stepwise pathogenic developments. A novel intestine-on-chip system for investigating multiple aspects of IBD's immunopathogenesis is presented. The system includes an array of tight and polarized barrier models formed from intestinal epithelial cells on an in-vivo-like subepithelial matrix within one week. The dynamic remodeling of the subepithelial matrix by cells or their secretome demonstrates the physiological relevance of the on-chip barrier models. The system design enables introduction of various immune cell types and inflammatory stimuli at specific locations in the same barrier model, which facilitates investigations of the distinct roles of each cell type in intestinal inflammation development. It is showed that inflammatory behavior manifests in an upregulated expression of inflammatory markers and cytokines (TNF-α). The neutralizing effect of the anti-inflammatory antibody Infliximab on levels of TNF-α and its inducible cytokines could be explicitly shown. Overall, an innovative approach to systematically developing a microphysiological system to comprehend immune-system-mediated disorders of IBD and to identify new therapeutic strategies is presented.
Collapse
Affiliation(s)
- Oanh T. P. Nguyen
- Bio Engineering LaboratoryDepartment of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| | - Patrick M. Misun
- Bio Engineering LaboratoryDepartment of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| | - Andreas Hierlemann
- Bio Engineering LaboratoryDepartment of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| | - Christian Lohasz
- Bio Engineering LaboratoryDepartment of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| |
Collapse
|
25
|
Ko J, Song J, Choi N, Kim HN. Patient-Derived Microphysiological Systems for Precision Medicine. Adv Healthc Mater 2024; 13:e2303161. [PMID: 38010253 PMCID: PMC11469251 DOI: 10.1002/adhm.202303161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Indexed: 11/29/2023]
Abstract
Patient-derived microphysiological systems (P-MPS) have emerged as powerful tools in precision medicine that provide valuable insight into individual patient characteristics. This review discusses the development of P-MPS as an integration of patient-derived samples, including patient-derived cells, organoids, and induced pluripotent stem cells, into well-defined MPSs. Emphasizing the necessity of P-MPS development, its significance as a nonclinical assessment approach that bridges the gap between traditional in vitro models and clinical outcomes is highlighted. Additionally, guidance is provided for engineering approaches to develop microfluidic devices and high-content analysis for P-MPSs, enabling high biological relevance and high-throughput experimentation. The practical implications of the P-MPS are further examined by exploring the clinically relevant outcomes obtained from various types of patient-derived samples. The construction and analysis of these diverse samples within the P-MPS have resulted in physiologically relevant data, paving the way for the development of personalized treatment strategies. This study describes the significance of the P-MPS in precision medicine, as well as its unique capacity to offer valuable insights into individual patient characteristics.
Collapse
Affiliation(s)
- Jihoon Ko
- Department of BioNano TechnologyGachon UniversitySeongnam‐siGyeonggi‐do13120Republic of Korea
| | - Jiyoung Song
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
| | - Nakwon Choi
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST SchoolSeoul02792Republic of Korea
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
| | - Hong Nam Kim
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST SchoolSeoul02792Republic of Korea
- School of Mechanical EngineeringYonsei UniversitySeoul03722Republic of Korea
- Yonsei‐KIST Convergence Research InstituteYonsei UniversitySeoul03722Republic of Korea
| |
Collapse
|
26
|
Wang X, Luo Y, He S, Lu Y, Gong Y, Gao L, Mao S, Liu X, Jiang N, Pu Q, Du D, Shu Y, Hai S, Li S, Chen HN, Zhao Y, Xie D, Qi S, Lei P, Hu H, Xu H, Zhou ZG, Dong B, Zhang H, Zhang Y, Dai L. Age-, sex- and proximal-distal-resolved multi-omics identifies regulators of intestinal aging in non-human primates. NATURE AGING 2024; 4:414-433. [PMID: 38321225 PMCID: PMC10950786 DOI: 10.1038/s43587-024-00572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024]
Abstract
The incidence of intestinal diseases increases with age, yet the mechanisms governing gut aging and its link to diseases, such as colorectal cancer (CRC), remain elusive. In this study, while considering age, sex and proximal-distal variations, we used a multi-omics approach in non-human primates (Macaca fascicularis) to shed light on the heterogeneity of intestinal aging and identify potential regulators of gut aging. We explored the roles of several regulators, including those from tryptophan metabolism, in intestinal function and lifespan in Caenorhabditis elegans. Suggesting conservation of region specificity, tryptophan metabolism via the kynurenine and serotonin (5-HT) pathways varied between the proximal and distal colon, and, using a mouse colitis model, we observed that distal colitis was more sensitive to 5-HT treatment. Additionally, using proteomics analysis of human CRC samples, we identified links between gut aging and CRC, with high HPX levels predicting poor prognosis in older patients with CRC. Together, this work provides potential targets for preventing gut aging and associated diseases.
Collapse
Grants
- P40 OD010440 NIH HHS
- National Natural Science Foundation of China (National Science Foundation of China)
- National Key R&D Program of China,2022YFA1303200, 2018YFC2000305; The 135 Project of West China Hospital, ZYJC21005, ZYGD20010 and ZYYC23013.
- Natural Science Foundation of Sichuan Province,2023NSFSC1196
- Natural Science Foundation of Sichuan Province,2021YFS0134
- National Clinical Research Center for Geriatrics of West China Hospital, Z2021JC005
- The 135 Project of West China Hospital, ZYYC23025.
- National Key R&D Program of China, 2019YFA0110203;
- National Clinical Research Center for Geriatrics of West China Hospital, Z2021JC006;
Collapse
Affiliation(s)
- Xinyuan Wang
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yaru Luo
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Siyu He
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Lu
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yanqiu Gong
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Li Gao
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shengqiang Mao
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaohui Liu
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Na Jiang
- Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Qianlun Pu
- Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Du
- Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Shu
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shan Hai
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shuangqing Li
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hai-Ning Chen
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Zhao
- Department of Rheumatology and Immunology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Xie
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shiqian Qi
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Lei
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongbo Hu
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Heng Xu
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zong-Guang Zhou
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Biao Dong
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Huiyuan Zhang
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Yan Zhang
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Lunzhi Dai
- National Clinical Research Center for Geriatrics, Center for Immunology and Hematology and General Practice Ward/International Medical Center Ward, General Practice Medical Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
27
|
Wheeler AE, Stoeger V, Owens RM. Lab-on-chip technologies for exploring the gut-immune axis in metabolic disease. LAB ON A CHIP 2024; 24:1266-1292. [PMID: 38226866 DOI: 10.1039/d3lc00877k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The continued rise in metabolic diseases such as obesity and type 2 diabetes mellitus poses a global health burden, necessitating further research into factors implicated in the onset and progression of these diseases. Recently, the gut-immune axis, with diet as a main regulator, has been identified as a possible role player in their development. Translation of conventional 2D in vitro and animal models is however limited, while human studies are expensive and preclude individual mechanisms from being investigated. Lab-on-chip technology therefore offers an attractive new avenue to study gut-immune interactions. This review provides an overview of the influence of diet on gut-immune interactions in metabolic diseases and a critical analysis of the current state of lab-on-chip technology to study this axis. While there has been progress in the development of "immuno-competent" intestinal lab-on-chip models, with studies showing the ability of the technology to provide mechanical cues, support longer-term co-culture of microbiota and maintain in vivo-like oxygen gradients, platforms which combine all three and include intestinal and immune cells are still lacking. Further, immune cell types and inclusion of microenvironment conditions which enable in vivo-like immune cell dynamics as well as host-microbiome interactions are limited. Future model development should focus on combining these conditions to create an environment capable of hosting more complex microbiota and immune cells to allow further study into the effects of diet and related metabolites on the gut-immune ecosystem and their role in the prevention and development of metabolic diseases in humans.
Collapse
Affiliation(s)
- Alexandra E Wheeler
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| | - Verena Stoeger
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| |
Collapse
|
28
|
Nagao I, Nakazawa M, Ambrosini YM. Three-Dimensional Morphogenesis in Canine Gut-on-a-Chip Using Intestinal Organoids Derived from Inflammatory Bowel Disease Patients. J Vis Exp 2024:10.3791/65720. [PMID: 38407238 PMCID: PMC10994191 DOI: 10.3791/65720] [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] [Indexed: 02/27/2024] Open
Abstract
Canine intestines possess similarities in anatomy, microbiology, and physiology to those of humans, and dogs naturally develop spontaneous intestinal disorders similar to humans. Overcoming the inherent limitation of three-dimensional (3D) organoids in accessing the apical surface of the intestinal epithelium has led to the generation of two-dimensional (2D) monolayer cultures, which expose the accessible luminal surface using cells derived from the organoids. The integration of these organoids and organoid-derived monolayer cultures into a microfluidic Gut-on-a-Chip system has further evolved the technology, allowing for the development of more physiologically relevant dynamic in vitro intestinal models. In this study, we present a protocol for generating 3D morphogenesis of canine intestinal epithelium using primary intestinal tissue samples obtained from dogs affected by inflammatory bowel disease (IBD). We also outline a protocol for generating and maintaining 2D monolayer cultures and intestine-on-a-chip systems using cells derived from the 3D intestinal organoids. The protocols presented in this study serve as a foundational framework for establishing a microfluidic Gut-on-a-Chip system specifically designed for canines. By laying the groundwork for this innovative approach, we aim to expand the application of these techniques in biomedical and translational research, aligning with the principles of the One Health Initiative. By utilizing this approach, we can develop more physiologically relevant dynamic in vitro models for studying intestinal physiology in both dogs and humans. This has significant implications for biomedical and pharmaceutical applications, as it can aid in the development of more effective treatments for intestinal diseases in both species.
Collapse
Affiliation(s)
- Itsuma Nagao
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University; Department of Veterinary Internal Medicine, Graduate School of Agricultural and Life Sciences, The University of Tokyo
| | - Meg Nakazawa
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University
| | - Yoko M Ambrosini
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University;
| |
Collapse
|
29
|
Bu L, Li Y, Wang C, Jiang Y, Suo H. Preventive effect of Lacticaseibacillus rhamnosus 2016SWU.05.0601 and its postbiotic elements on dextran sodium sulfate-induced colitis in mice. Front Microbiol 2024; 15:1342705. [PMID: 38374921 PMCID: PMC10876090 DOI: 10.3389/fmicb.2024.1342705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/15/2024] [Indexed: 02/21/2024] Open
Abstract
Microbial-based therapies are one of the hotspots in the field of ulcerative colitis research. The lactic acid bacteria and their postbiotics occupy a key position in microbial therapies, however, the mechanism by which they alleviate ulcerative colitis in mice is unknown. We investigated the effects of Lacticaseibacillus rhamnosus 2016SWU.05.0601 (Lr-0601) and its postbiotics on male Kunming mice with dextran sulfate sodium salt (DSS)-induced ulcerative colitis (UC). The results showed that Lr-0601 significantly alleviated the deterioration of UC and restored the expression of intestinal mechanical barrier proteins. In addition, Lr-0601 significantly reduced the expression of inflammatory cytokines in the body and regulated the expression of key regulatory genes of the NF-κB-iNOS/COX-2 signaling pathway in colon tissues to a large extent. Our results suggest that supplementation with Lr-0601 and its postbiotics can effectively prevent DSS-induced UC and have a beneficial effect on intestinal health, which also provides new insights and research bases for the prevention as well as the treatment of ulcerative colitis and other diseases related to intestinal barrier dysfunction and other diseases.
Collapse
Affiliation(s)
- Linli Bu
- College of Food Science, Southwest University, Chongqing, China
| | - Yang Li
- College of Food Science, Southwest University, Chongqing, China
| | - Chen Wang
- College of Food Science, Southwest University, Chongqing, China
- Modern “Chuan Cai Yu Wei” Food Industry Innovation Research Institute, Chongqing, China
| | - Yuhang Jiang
- College of Food Science, Southwest University, Chongqing, China
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing, China
- Modern “Chuan Cai Yu Wei” Food Industry Innovation Research Institute, Chongqing, China
| |
Collapse
|
30
|
Gudi RR, Johnson BM, Gaudreau MC, Sun W, Ball L, Vasu C. Intestinal permeability and inflammatory features of juvenile age correlate with the eventual systemic autoimmunity in lupus-prone female SWR × NZB F1 (SNF1) mice. Immunology 2024; 171:235-249. [PMID: 37947218 PMCID: PMC10842200 DOI: 10.1111/imm.13713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023] Open
Abstract
The incidence of systemic lupus erythematosus (SLE) is about nine times higher in women than in men, and the underlying mechanisms that contribute to this gender bias are not fully understood. Previously, using lupus-prone (SWR × NZB)F1 (SNF1) mice, we have shown that the intestinal immune system could play a role in the initiation and progression of disease in SLE, and depletion of gut microbiota produces more pronounced disease protection in females than in males. Here, we show that the gut permeability features of lupus-prone female SNF1 mice at juvenile ages directly correlate with the expression levels of pro-inflammatory factors, faecal IgA abundance and nAg reactivity and the eventual systemic autoantibody levels and proteinuria onset. Furthermore, we observed that the disease protection achieved in female SNF1 mice upon depletion of gut microbiota correlates with the diminished gut inflammatory protein levels, intestinal permeability and circulating microbial DNA levels. However, faecal microbiota transplant from juvenile male and females did not result in modulation of gut inflammatory features or permeability. Overall, these observations suggest that the early onset of intestinal inflammation, systemic autoantibody production and clinical stage disease in lupus-prone females is linked to higher gut permeability in them starting at as early as juvenile age. While the higher gut permeability in juvenile lupus-prone females is dependent on the presence of gut microbes, it appears to be independent of the composition of gut microbiota.
Collapse
Affiliation(s)
- Radhika R Gudi
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Benjamin M Johnson
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Marie-Claude Gaudreau
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Wei Sun
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lauren Ball
- Department of Pharmacology, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Chenthamarakshan Vasu
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
31
|
Sardelli L, Campanile M, Boeri L, Donnaloja F, Fanizza F, Perottoni S, Petrini P, Albani D, Giordano C. A novel on-a-chip system with a 3D-bioinspired gut mucus suitable to investigate bacterial endotoxins dynamics. Mater Today Bio 2024; 24:100898. [PMID: 38204482 PMCID: PMC10776420 DOI: 10.1016/j.mtbio.2023.100898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 11/20/2023] [Accepted: 12/01/2023] [Indexed: 01/12/2024] Open
Abstract
The possible pathogenic impact of pro-inflammatory molecules produced by the gut microbiota is one of the hypotheses considered at the basis of the biomolecular dialogue governing the microbiota-gut-brain axis. Among these molecules, lipopolysaccharides (LPS) produced by Gram-negative gut microbiota strains may have a potential key role due to their toxic effects in both the gut and the brain. In this work, we engineered a new dynamic fluidic system, the MINERVA device (MI-device), with the potential to advance the current knowledge of the biological mechanisms regulating the microbiota-gut molecular crosstalk. The MI-device supported the growth of bacteria that are part of the intestinal microbiota under dynamic conditions within a 3D moving mucus model, with features comparable to the physiological conditions (storage modulus of 80 ± 19 Pa, network mesh size of 41 ± 3 nm), without affecting their viability (∼ 109 bacteria/mL). The integration of a fluidically optimized and user-friendly design with a bioinspired microenvironment enabled the sterile extraction and quantification of the LPS produced within the mucus by bacteria (from 423 ± 34 ng/mL to 1785 ± 91 ng/mL). Compatibility with commercially available Transwell-like inserts allows the user to precisely control the transport phenomena that occur between the two chambers by selecting the pore density of the insert membrane without changing the design of the system. The MI-device is able to provide the flow of sterile medium enriched with LPS directly produced by bacteria, opening up the possibility of studying the effects of bacteria-derived molecules on cells in depth, as well as the assessment and characterization of their effects in a physiological or pathological scenario.
Collapse
Affiliation(s)
- L. Sardelli
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - M. Campanile
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - L. Boeri
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - F. Donnaloja
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - F. Fanizza
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - S. Perottoni
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - P. Petrini
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| | - D. Albani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - C. Giordano
- Department of Chemistry, Materials and Chemical Engineering ‘Giulio Natta,’ Politecnico di Milano, Milan, Italy
| |
Collapse
|
32
|
Huang FC, Huang SC. The Hazards of Probiotics on Gut-Derived Pseudomonas aeruginosa Sepsis in Mice Undergoing Chemotherapy. Biomedicines 2024; 12:253. [PMID: 38397855 PMCID: PMC10886725 DOI: 10.3390/biomedicines12020253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/04/2024] [Accepted: 01/11/2024] [Indexed: 02/25/2024] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is a leading cause of nosocomial infections associated with a high mortality rate and represents a serious threat to human health and the increasing frequency of antimicrobial resistance. Cancer patients are more vulnerable to invasive infection due to ulcerative lesions in mucosal surfaces and immune suppression secondary to chemotherapy. In our in vitro study, we observed that probiotics have the potential to yield beneficial effects on intestinal epithelial cells infected with P. aeruginosa. Additionally, probiotics were found to confer advantageous effects on the innate immunity of mice suffering from Salmonella-induced colitis. As a result, we sought to investigate the impact of probiotics on gut-derived P. aeruginosa sepsis induced by chemotherapy. Following chemotherapy, gut-derived P. aeruginosa sepsis was induced in female C57BL/6 mice aged 6-8 weeks, which were raised under specific-pathogen-free (SPF) conditions in an animal center. Prior to the induction of the sepsis model, the mice were administered 1 × 108 colony-forming units (CFU) of the probiotics, namely Lactobacillus rhamnosus GG (LGG) and Bifidobacterium longum (BL) via oral gavage. We observed that LGG or BL amplified the inflammatory mRNA expression in mice undergoing chemotherapy and suffering from gut-derived P. aeruginosa sepsis. This led to a heightened severity of colitis, as indicated by histological examination. Meanwhile, there was a notable decrease in the expression of antimicrobial peptide mRNA along with reduced levels of zonulin and claudin-2 protein staining within mucosal tissue. These alterations facilitated the translocation of bacteria to the liver, spleen, and bloodstream. To our astonishment, the introduction of probiotics exacerbated gut-derived P. aeruginosa sepsis in mice undergoing chemotherapy. Conclusively, we must be prudent when using probiotics in mice receiving chemotherapy complicated with gut-derived P. aeruginosa sepsis.
Collapse
Affiliation(s)
- Fu-Chen Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Shun-Chen Huang
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| |
Collapse
|
33
|
Hahn S, Kim G, Jin SM, Kim JH. Protective effects of metformin in the pro-inflammatory cytokine induced intestinal organoids injury model. Biochem Biophys Res Commun 2024; 690:149291. [PMID: 38006803 DOI: 10.1016/j.bbrc.2023.149291] [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: 11/07/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 11/27/2023]
Abstract
Pathogenesis of inflammatory bowel disease (IBD) accompanies disrupted intestinal tight junctions. However, many approaches of therapeutics for IBD are focused only on anti-inflammatory effects and most cellular experiments are based on two-dimensional cell lines which have insufficient circumstances of intestine. Thus, here, we used three-dimensional structure intestinal organoids to investigate effects of metformin in the in vitro IBD condition. In this study, we focused on both tight junctions and the levels of inflammatory cytokines. Metformin enhances the intestinal barrier in injured intestine via upregulation of AMP-activated protein kinase, dysfunction of which contributes to the pathogenesis of intestinal diseases. We aim to investigate the effects of metformin on cytokine-induced injured intestinal organoids. Tumor necrosis factor-alpha (TNF-α) was used to induce intestinal injury in an organoid model, and the effects of metformin were assessed. Cell viability and levels of inflammatory cytokines were quantified in addition to tight junction markers. Furthermore, 4 kDa FITC-dextran was used to assess intestinal permeability. The upregulation of inflammatory cytokine levels was alleviated by metformin, which also restored the intestinal epithelium permeability in TNF-α-treated injury organoids. We confirmed that claudin-2 and claudin-7, representative tight junction markers, were also protected by metformin treatment. This study confirms the protective effects of metformin, which could be used as a therapeutic strategy for inflammatory intestinal diseases.
Collapse
Affiliation(s)
- Soojung Hahn
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06355, South Korea; Division of Endocrinology and Metabolism, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06355, South Korea.
| | - Gyuri Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06355, South Korea.
| | - Sang-Man Jin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06355, South Korea.
| | - Jae Hyeon Kim
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06355, South Korea; Division of Endocrinology and Metabolism, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06355, South Korea.
| |
Collapse
|
34
|
Wang H, Ning X, Zhao F, Zhao H, Li D. Human organoids-on-chips for biomedical research and applications. Theranostics 2024; 14:788-818. [PMID: 38169573 PMCID: PMC10758054 DOI: 10.7150/thno.90492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/09/2023] [Indexed: 01/05/2024] Open
Abstract
Human organoids-on-chips (OrgOCs) are the synergism of human organoids (HOs) technology and microfluidic organs-on-chips (OOCs). OOCs can mimic extrinsic characteristics of organs, such as environmental clues of living tissue, while HOs are more amenable to biological analysis and genetic manipulation. By spatial cooperation, OrgOCs served as 3D organotypic living models allowing them to recapitulate critical tissue-specific properties and forecast human responses and outcomes. It represents a giant leap forward from the regular 2D cell monolayers and animal models in the improved human ecological niche modeling. In recent years, OrgOCs have offered potential promises for clinical studies and advanced the preclinical-to-clinical translation in medical and industrial fields. In this review, we highlight the cutting-edge achievements in OrgOCs, introduce the key features of OrgOCs architectures, and share the revolutionary applications in basic biology, disease modeling, preclinical assay and precision medicine. Furthermore, we discuss how to combine a wide range of disciplines with OrgOCs and accelerate translational applications, as well as the challenges and opportunities of OrgOCs in biomedical research and applications.
Collapse
Affiliation(s)
- Hui Wang
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Xiufan Ning
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Feng Zhao
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Hui Zhao
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Dong Li
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| |
Collapse
|
35
|
Hsu SCJ, Luu TU, Smith TD, Liu WF. Macro- and micro-scale culture environment differentially regulate the effects of crowding on macrophage function. Biotechnol Bioeng 2024; 121:306-316. [PMID: 37792882 DOI: 10.1002/bit.28554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/25/2023] [Accepted: 08/21/2023] [Indexed: 10/06/2023]
Abstract
Macrophages hold vital roles in immune defense, wound healing, and tissue homeostasis, and have the exquisite ability to sense and respond to dynamically changing cues in their microenvironment. Much of our understanding of their behavior has been derived from studies performed using in vitro culture systems, in which the cell environment can be precisely controlled. Recent advances in miniaturized culture platforms also offer the ability to recapitulate some features of the in vivo environment and analyze cellular responses at the single-cell level. Since macrophages are sensitive to their surrounding environments, the specific conditions in both macro- and micro-scale cultures likely contribute to observed responses. In this study, we investigate how the presence of neighboring cells influence macrophage activation following proinflammatory stimulation in both bulk and micro-scale culture. We found that in bulk cultures, higher seeding density negatively regulated the average TNF-α secretion from individual macrophages in response to inflammatory agonists, and this effect was partially caused by the reduced cell-to-media volume ratio. In contrast, studies conducted using microwells to isolate single cells and groups of cells revealed that increasing numbers of cells positively influences their inflammatory activation, suggesting that the absolute cell numbers in the system may be important. In addition, a single inflammatory cell enhanced the inflammatory state of a small group of cells. Overall, this work helps to better understand how variations of macroscopic and microscopic culture environments influence studies in macrophage biology and provides insight into how the presence of neighboring cells and the soluble environment influences macrophage activation.
Collapse
Affiliation(s)
- Ssu-Chieh J Hsu
- Department of Biomedical Engineering, University of California, Irvine, California, USA
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, University of California, Irvine, California, USA
| | - Thuy U Luu
- Department of Biomedical Engineering, University of California, Irvine, California, USA
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, University of California, Irvine, California, USA
| | - Tim D Smith
- Department of Biomedical Engineering, University of California, Irvine, California, USA
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, University of California, Irvine, California, USA
| | - Wendy F Liu
- Department of Biomedical Engineering, University of California, Irvine, California, USA
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, University of California, Irvine, California, USA
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, California, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| |
Collapse
|
36
|
Kriaa A, Mariaule V, De Rudder C, Jablaoui A, Sokol H, Wilmes P, Maguin E, Rhimi M. From animal models to gut-on-chip: the challenging journey to capture inter-individual variability in chronic digestive disorders. Gut Microbes 2024; 16:2333434. [PMID: 38536705 PMCID: PMC10978023 DOI: 10.1080/19490976.2024.2333434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/10/2024] Open
Abstract
Chronic digestive disorders are of increasing incidence worldwide with expensive treatments and no available cure. Available therapeutic schemes mainly rely on symptom relief, with large degrees of variability in patients' response to such treatments, underlining the need for new therapeutic strategies. There are strong indications that the gut microbiota's contribution seems to be a key modulator of disease activity and patients' treatment responses. Hence, efforts have been devoted to understanding host-microbe interactions and the mechanisms underpinning such variability. Animal models, being the gold standard, provide valuable mechanistic insights into host-microbe interactions. However, they are not exempt from limitations prompting the development of alternative methods. Emerging microfluidic technologies and gut-on-chip models were shown to mirror the main features of gut physiology and disease state, reflect microbiota modification, and include functional readouts for studying host responses. In this commentary, we discuss the relevance of animal models in understanding host-microbe interactions and how gut-on-chip technology holds promises for addressing patient variability in responses to chronic digestive disease treatment.
Collapse
Affiliation(s)
- Aicha Kriaa
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Vincent Mariaule
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Charlotte De Rudder
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Amin Jablaoui
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Harry Sokol
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Sorbonne Université, Paris, France
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Emmanuelle Maguin
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Moez Rhimi
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| |
Collapse
|
37
|
Hayes JA, Lunger AW, Sharma AS, Fernez MT, Carrier RL, Koppes AN, Koppes R, Woolston BM. Engineered bacteria titrate hydrogen sulfide and induce concentration-dependent effects on the host in a gut microphysiological system. Cell Rep 2023; 42:113481. [PMID: 37980564 PMCID: PMC10791167 DOI: 10.1016/j.celrep.2023.113481] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/05/2023] [Accepted: 11/06/2023] [Indexed: 11/21/2023] Open
Abstract
Hydrogen sulfide (H2S) is a gaseous microbial metabolite whose role in gut diseases is debated, with contradictory results stemming from experimental difficulties associated with accurate dosing and measuring H2S and the use of model systems that do not accurately represent the human gut environment. Here, we engineer Escherichia coli to titrate H2S across the physiological range in a gut microphysiological system (chip) supportive of the co-culture of microbes and host cells. The chip is engineered to maintain H2S gas tension and enables visualization of co-culture in real time with confocal microscopy. Engineered strains colonize the chip and are metabolically active for 2 days, during which they produce H2S across a 16-fold range and induce changes in host gene expression and metabolism in an H2S-concentration-dependent manner. These results validate a platform for studying the mechanisms underlying microbe-host interactions by enabling experiments that are infeasible with current animal and in vitro models.
Collapse
Affiliation(s)
- Justin A Hayes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Anna W Lunger
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Aayushi S Sharma
- Department of Mechanical and Industrial Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA; Department of Bioengineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Matthew T Fernez
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Rebecca L Carrier
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA; Department of Bioengineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA; Department of Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Abigail N Koppes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA; Department of Bioengineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Ryan Koppes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| | - Benjamin M Woolston
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| |
Collapse
|
38
|
Li H, Liu S, Zhang K, Zhu X, Dai J, Lu Y. Gut microbiome and plasma metabolome alterations in myopic mice. Front Microbiol 2023; 14:1251243. [PMID: 38179454 PMCID: PMC10764480 DOI: 10.3389/fmicb.2023.1251243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
Background Myopia is one of the most common eye diseases leading to blurred distance vision. Inflammatory diseases could trigger or exacerbate myopic changes. Although gut microbiota bacteria are associated with various inflammatory diseases, little is known about its role in myopia. Materials and methods The mice were randomly divided into control and model groups, with the model group being attached-30D lens onto the eyes for 3 weeks. Then, mouse cecal contents and plasma were collected to analyze their intestinal microbiota and plasma metabolome. Results We identified that the microbial composition differed considerably between the myopic and non-myopic mice, with the relative abundance of Firmicutes phylum decreased obviously while that of Actinobacteria phylum was increased in myopia. Furthermore, Actinobacteria and Bifidobacterium were positively correlated with axial lengths (ALs) of eyeballs while negatively correlated with refractive diopters. Untargeted metabolomic analysis identified 141 differentially expressed metabolites, and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis revealed considerable enrichment mainly in amino acid metabolism pathways. Notably, pathways involved glutamate metabolism including "Glutamine and D-glutamate metabolism" and "Alanine, aspartate and glutamate metabolism" was changed dramatically, which presented as the concentrations of L-Glutamate and L-Glutamine decreased obviously in myopia. Interestingly, microbiome dysbiosis and metabolites alternations in myopia have a disrupting gut barrier feature. We further demonstrated that the gut barrier function was impaired in myopic mice manifesting in decreased expression of Occludin, ZO-1 and increased permeation of FITC-dextran. Discussion Myopic mice had obviously altered gut microbiome and metabolites profiles compared to non-myopic mice. The dysbiosis and plasma metabolomics shift in myopia had an interrupting gut barrier feature. Our study provides new insights into the possible role of the gut microbiota in myopia and reinforces the potential feasibility of microbiome-based therapies in myopia.
Collapse
Affiliation(s)
- Hao Li
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Shuyu Liu
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Keke Zhang
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Xiangjia Zhu
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Jinhui Dai
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi Lu
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| |
Collapse
|
39
|
Tataru C, Livni M, Marean-Reardon C, Franco MC, David M. Cytokine induced inflammatory bowel disease model using organ-on-a-chip technology. PLoS One 2023; 18:e0289314. [PMID: 38091316 PMCID: PMC10718466 DOI: 10.1371/journal.pone.0289314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 07/14/2023] [Indexed: 12/18/2023] Open
Abstract
Over 2 million people in North America suffer from inflammatory bowel disease (IBD), a chronic and idiopathic inflammatory condition. While previous research has primarily focused on studying immune cells as a cause and therapeutic target for IBD, recent findings suggest that non-immune cells may also play a crucial role in mediating cytokine and chemokine signaling, and therefore IBD symptoms. In this study, we developed an organ-on-a-chip co-culture model of Caco2 epithelial and HUVEC endothelial cells and induced inflammation using pro-inflammatory cytokines TNF-α and IFN-γ. We tested different concentration ranges and delivery orientations (apical vs. basal) to develop a consistently inducible inflammatory response model. We then measured pro-inflammatory cytokines and chemokines IL-6, IL-8, and CXCL-10, as well as epithelial barrier integrity. Our results indicate that this model 1. induces IBD-like cytokine secretion in non-immune cells and 2. decreases barrier integrity, making it a feasible and reliable model to test the direct actions of potential anti-inflammatory therapeutics on epithelial and endothelial cells.
Collapse
Affiliation(s)
- Christine Tataru
- Oregon State University, College of Science, Microbiology, Corvallis, OR, United States of America
| | - Maya Livni
- Oregon State University, College of Science, Microbiology, Corvallis, OR, United States of America
| | - Carrie Marean-Reardon
- Oregon State University, College of Science, Biochemistry and Biophysics, Corvallis, OR, United States of America
| | - Maria Clara Franco
- Oregon State University, College of Science, Biochemistry and Biophysics, Corvallis, OR, United States of America
- Florida International University, Herbert Wertheim College of Medicine, Center for Translational Science, Port St. Lucie, FL, United States of America
| | - Maude David
- Oregon State University, College of Science, Microbiology, Corvallis, OR, United States of America
- Oregon State University, College of Pharmacy, Corvallis, OR, United States of America
| |
Collapse
|
40
|
Moon HR, Surianarayanan N, Singh T, Han B. Microphysiological systems as reliable drug discovery and evaluation tools: Evolution from innovation to maturity. BIOMICROFLUIDICS 2023; 17:061504. [PMID: 38162229 PMCID: PMC10756708 DOI: 10.1063/5.0179444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024]
Abstract
Microphysiological systems (MPSs), also known as organ-on-chip or disease-on-chip, have recently emerged to reconstitute the in vivo cellular microenvironment of various organs and diseases on in vitro platforms. These microfluidics-based platforms are developed to provide reliable drug discovery and regulatory evaluation testbeds. Despite recent emergences and advances of various MPS platforms, their adoption of drug discovery and evaluation processes still lags. This delay is mainly due to a lack of rigorous standards with reproducibility and reliability, and practical difficulties to be adopted in pharmaceutical research and industry settings. This review discusses the current and potential use of MPS platforms in drug discovery processes while considering the context of several key steps during drug discovery processes, including target identification and validation, preclinical evaluation, and clinical trials. Opportunities and challenges are also discussed for the broader dissemination and adoption of MPSs in various drug discovery and regulatory evaluation steps. Addressing these challenges will transform long and expensive drug discovery and evaluation processes into more efficient discovery, screening, and approval of innovative drugs.
Collapse
Affiliation(s)
- Hye-Ran Moon
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | | | - Tarun Singh
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Bumsoo Han
- Author to whom correspondence should be addressed:. Tel: +1-765-494-5626
| |
Collapse
|
41
|
Huang S, Chen J, Cui Z, Ma K, Wu D, Luo J, Li F, Xiong W, Rao S, Xiang Q, Shi W, Song T, Deng J, Yin Y, Tan C. Lachnospiraceae-derived butyrate mediates protection of high fermentable fiber against placental inflammation in gestational diabetes mellitus. SCIENCE ADVANCES 2023; 9:eadi7337. [PMID: 37922350 PMCID: PMC10624355 DOI: 10.1126/sciadv.adi7337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/04/2023] [Indexed: 11/05/2023]
Abstract
Inflammation-associated insulin resistance is a key trigger of gestational diabetes mellitus (GDM), but the underlying mechanisms and effective interventions remain unclear. Here, we report the association of placental inflammation (tumor necrosis factor-α) and abnormal maternal glucose metabolism in patients with GDM, and a high fermentable dietary fiber (HFDF; konjac) could reduce GDM development through gut flora-short-chain fatty acid-placental inflammation axis in GDM mouse model. Mechanistically, HFDF increases abundances of Lachnospiraceae and butyrate, reduces placental-derived inflammation by enhancing gut barrier and inhibiting the transfer of bacterial-derived lipopolysaccharide, and ultimately resists high-fat diet-induced insulin resistance. Lachnospiraceae and butyrate have similar anti-GDM and anti-placental inflammation effects, and they can ameliorate placental function and pregnancy outcome effects probably by dampening placental immune dysfunction. These findings demonstrate the involvement of important placental inflammation-related mechanisms in the progression of GDM and the great potential of HFDFs to reduce susceptibility to GDM through gut-flora-placenta axis.
Collapse
Affiliation(s)
- Shuangbo Huang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jianzhao Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Zhijuan Cui
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Kaidi Ma
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Deyuan Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jinxi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Fuyong Li
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Department of Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen 518000, China
- Department of Obstetrics and Gynecology, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen 518000, China
- Huazhong Agricultural University, College of Animal Science and Technology, Wuhan 430070, China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Wenyu Xiong
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Sujuan Rao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Quanhang Xiang
- Department of Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen 518000, China
| | - Wei Shi
- Department of Obstetrics and Gynecology, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen 518000, China
| | - Tongxing Song
- Huazhong Agricultural University, College of Animal Science and Technology, Wuhan 430070, China
| | - Jinping Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yulong Yin
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Chengquan Tan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
42
|
Kwee BJ, Li X, Nguyen XX, Campagna C, Lam J, Sung KE. Modeling immunity in microphysiological systems. Exp Biol Med (Maywood) 2023; 248:2001-2019. [PMID: 38166397 PMCID: PMC10800123 DOI: 10.1177/15353702231215897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024] Open
Abstract
There is a need for better predictive models of the human immune system to evaluate safety and efficacy of immunomodulatory drugs and biologics for successful product development and regulatory approvals. Current in vitro models, which are often tested in two-dimensional (2D) tissue culture polystyrene, and preclinical animal models fail to fully recapitulate the function and physiology of the human immune system. Microphysiological systems (MPSs) that can model key microenvironment cues of the human immune system, as well as of specific organs and tissues, may be able to recapitulate specific features of the in vivo inflammatory response. This minireview provides an overview of MPS for modeling lymphatic tissues, immunity at tissue interfaces, inflammatory diseases, and the inflammatory tumor microenvironment in vitro and ex vivo. Broadly, these systems have utility in modeling how certain immunotherapies function in vivo, how dysfunctional immune responses can propagate diseases, and how our immune system can combat pathogens.
Collapse
Affiliation(s)
- Brian J Kwee
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19711, USA
| | - Xiaoqing Li
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Xinh-Xinh Nguyen
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Courtney Campagna
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA
| | - Johnny Lam
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Kyung E Sung
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
43
|
Ma Q, Zhang X, Xu X, Lu Y, Chen Q, Chen Y, Liu C, Chen K. Long-term oral administration of burdock fructooligosaccharide alleviates DSS-induced colitis in mice by mediating anti-inflammatory effects and protection of intestinal barrier function. Immun Inflamm Dis 2023; 11:e1092. [PMID: 38018589 PMCID: PMC10664397 DOI: 10.1002/iid3.1092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Ulcerative colitis, a typical subtype of inflammatory bowel disease, can cause many serious complications. Burdock fructooligosaccharide (BFO), a linear inulin with a purity of 99.439% and a molecular weight of 2345 Da, demonstrates anti-inflammatory and immunomodulatory properties. METHODS The Kunming mice were divided into two experimental models: a normal pretreatment model and a colitis experimental model. During the experimental treatment period, we assessed changes in weight and disease activity index (DAI), quantified the intestinal index, and determined myeloperoxidase (MPO) activity and reactive oxide species (ROS) levels in colitis mice. We also photographed colon morphology to investigate alterations in the integrity of the intestinal barrier function. Finally, we performed ELISA and qRT-PCR to evaluate the anti-inflammatory effect of BFO treatment on colitis mice. RESULT The long-term oral administration of BFO alone exhibited protective effects by preventing disruption of the intestinal functional structure and increasing the colon index in mice. However, in a dextran sodium sulfate (DSS)-induced colitis mouse model, BFO administration facilitated quick recovery of body weight and effectively reduced the DAI, especially in the BFO-H group (500 mg/kg/day). BFO treatment maintained the integrity of the intestinal barrier by attenuating the crypt distortion and increasing the goblet cells count It restored the DSS-induced colon shortening and reduced the symptoms of colitis. These effects may be attributed to the appropriate concentrations of BFO effectively inhibiting MPO activity, clearing excessive ROS, and relieving spleen abnormalitie. BFO also attenuated the overexpression and excessive secretion of inflammatory cytokines (TNF-α, IL-1β, IL-6, and MCP-1) induced by DSS, reduced intestinal inflammation, and consequently protected the intestinal barrier function. CONCLUSION BFO effectively alleviated the symptoms of DSS-induced colitis by mediating anti-inflammatory effects and protecting the intestinal barrier integrity, thereby potentially facilitating the utilization of safer and more efficacious polysaccharides for managing chronic inflammatory diseases.
Collapse
Affiliation(s)
- Qunfei Ma
- School of Life ScienceShandong UniversityQingdaoChina
- Department of PhysiologyNaval Medical UniversityShanghaiChina
| | - Xiujuan Zhang
- School of Life ScienceShandong UniversityQingdaoChina
| | - Xuan Xu
- School of Life ScienceShandong UniversityQingdaoChina
| | - Yan Lu
- School of Life ScienceShandong UniversityQingdaoChina
- Clinical Laboratory Medicine DepartmentJining No. 1 People's HospitalJiningChina
| | - Qiang Chen
- Burdock Biotechnology (Dezhou) Co., LtdDezhouChina
| | - Yiru Chen
- Burdock Biotechnology (Dezhou) Co., LtdDezhouChina
| | - Chunyan Liu
- Provincial Engineering Laboratory for Screening and Re‐Evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of PharmacyDrug Research & Development Center, Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical CollegeWuhuChina
| | - Kaoshan Chen
- School of Life ScienceShandong UniversityQingdaoChina
- Provincial Engineering Laboratory for Screening and Re‐Evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of PharmacyDrug Research & Development Center, Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical CollegeWuhuChina
| |
Collapse
|
44
|
Li M, Wang Z, Fu S, Sun N, Li W, Xu Y, Han X, Zhang J, Miao J. Taurine reduction of injury from neutrophil infiltration ameliorates Streptococcus uberis-induced mastitis. Int Immunopharmacol 2023; 124:111028. [PMID: 37857121 DOI: 10.1016/j.intimp.2023.111028] [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: 08/01/2023] [Revised: 09/14/2023] [Accepted: 10/04/2023] [Indexed: 10/21/2023]
Abstract
Mastitis is a common disease of dairy cows characterized by infiltration of leukocytes, especially neutrophils, resulting in increased permeability of the blood-milk barrier (BMB). Taurine, a functional nutrient, has been shown to have anti-inflammatory and antioxidant effects. Here, we investigated the regulatory effects and mechanisms of taurine on the complex immune network of the mammary gland in Streptococcus uberis (S. uberis) infection. We found that taurine had no direct effect on CXCL2-mediated neutrophil chemotaxis. However, it inhibited MAPK and NF-κB signalings by modulating the activity of TAK1 downstream of TLR2, thereby reducing CXCL2 expression in macrophages to reduce neutrophil recruitment in S. uberis infection. Further, the AMPK/Nrf2 signaling pathway was activated by taurine to help mitigate oxidative damage, apoptosis and disruption of tight junctions in mammary epithelial cells caused by hypochlorous acid, a strong oxidant produced by neutrophils, thus protecting the integrity of the mammary epithelial barrier. Taurine protects the BMB from damage caused by neutrophils via blocking the macrophage-CXCL2-neutrophil signaling axis and increasing the antioxidant capacity of mammary epithelial cells.
Collapse
Affiliation(s)
- Ming Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Zhenglei Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Shaodong Fu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Naiyan Sun
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Weizhen Li
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Yuanyuan Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangan Han
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Jinqiu Zhang
- National Research Center for Veterinary Vaccine Engineering and Technology of China, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| | - Jinfeng Miao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
45
|
Zhang J, Huang YJ, Trapecar M, Wright C, Schneider K, Kemmit J, Hernandez-Gordillo V, Yoon JY, Alm EJ, Breault DT, Trumper D, Griffith LG. An immune-competent human gut microphysiological system enables inflammation-modulation of Faecalibacterium prausnitzii. RESEARCH SQUARE 2023:rs.3.rs-3373576. [PMID: 37886530 PMCID: PMC10602192 DOI: 10.21203/rs.3.rs-3373576/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Crosstalk of microbes with human gut epithelia and immune cells is crucial for gut health. However, there is no existing system for a long-term co-culture of human innate immune cells with epithelium and oxygen-intolerant commensal microbes, hindering the understanding of microbe-immune interactions in a controlled manner. Here, we establish a gut epithelium-microbe-immune microphysiological system to maintain the long-term continuous co-culture of Faecalibacterium prausnitzii/Faecalibacterium duncaniae with colonic epithelium, antigen-presenting cells (APCs, herein dendritic cells and macrophages), with CD4+ naïve T cells circulating underneath the colonic epithelium. Multiplex cytokine assays suggested that APCs contribute to the elevated level of cytokines and chemokines being secreted into both apical and basolateral compartments. In contrast, the absence of APCs does not allow reliable detection of these cytokines. In the presence of APCs, F. prausnitzii increased the transcription of pro-inflammatory genes such as toll-like receptor 1 (TLR1) and interferon alpha 1 (IFNA1) in the colonic epithelium, but no significant change on the secreted cytokines. In contrast, integration of CD4+ naïve T cells reverses this effect by decreasing the transcription of TLR1, IFNA1, and indoleamine 2,3-dioxygenase, and increasing the F. prausnitzii-induced secretion of pro-inflammatory cytokines such as IL-8, MCP-1/CCL2, and IL1A. These results highlight the contribution of individual innate immune cells in the regulation of the immune response triggered by the gut commensal F. prausnitzii. The successful integration of defined populations of immune cells in this gut microphysiological system demonstrated the usefulness of the GuMI physiomimetic platform to study microbe-epithelial-immune interactions in health and disease.
Collapse
Affiliation(s)
- Jianbo Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Yu-Ja Huang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Martin Trapecar
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Charles Wright
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kirsten Schneider
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - John Kemmit
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Jun Young Yoon
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Yonsei University, Seoul, South Korea
| | - Eric J. Alm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David T. Breault
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - David Trumper
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Linda G. Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
46
|
Aizpurua O, Blijleven K, Trivedi U, Gilbert MTP, Alberdi A. Unravelling animal-microbiota evolution on a chip. Trends Microbiol 2023; 31:995-1002. [PMID: 37217368 DOI: 10.1016/j.tim.2023.04.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 05/24/2023]
Abstract
Whether and how microorganisms have shaped the evolution of their animal hosts is a major question in biology. Although many animal evolutionary processes appear to correlate with changes in their associated microbial communities, the mechanistic processes leading to these patterns and their causal relationships are still far from being resolved. Gut-on-a-chip models provide an innovative approach that expands beyond the potential of conventional microbiome profiling to study how different animals sense and react to microbes by comparing responses of animal intestinal tissue models to different microbial stimuli. This complementary knowledge can contribute to our understanding of how host genetic features facilitate or prevent different microbiomes from being assembled, and in doing so elucidate the role of host-microbiota interactions in animal evolution.
Collapse
Affiliation(s)
- Ostaizka Aizpurua
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark.
| | - Kees Blijleven
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark
| | - Urvish Trivedi
- Department of Biology, Section of Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - M Thomas P Gilbert
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark; University Museum, NTNU, Trondheim, Norway
| | - Antton Alberdi
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
47
|
Oyama A, Takaki A, Adachi T, Wada N, Takeuchi Y, Onishi H, Shiraha H, Okada H, Otsuka M. Oxidative stress-related markers as prognostic factors for patients with primary sclerosing cholangitis in Japan. Hepatol Int 2023; 17:1215-1224. [PMID: 37493884 PMCID: PMC10522747 DOI: 10.1007/s12072-023-10557-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/27/2023] [Indexed: 07/27/2023]
Abstract
BACKGROUND/PURPOSE Primary sclerosing cholangitis (PSC) is a rare chronic liver disease. The mechanisms and prediction of PSC progression are unclear. Recent investigations have shown that general conditions, such as oxidative stress, affect the course of chronic diseases. We investigated the clinical course and oxidative stress-related condition of PSC to determine prognostic factors. METHODS We recruited 58 patients with PSC (mean age; 37.4 years, mean observation period; 1382 days) who visited our department from 2003 to 2021. Clinical characteristics were investigated to define prognostic factors. Oxidative stress status was evaluated using two types of markers: an oxidative stress marker (serum reactive oxygen metabolite; dROM) and an antioxidant marker (serum OXY adsorbent test; OXY). RESULTS The revised Mayo risk, Child-Pugh, model for end-stage liver disease-sodium (MELD-Na) scores or fibrosis-related FIB-4 index significantly predicted poor overall survival. High intestinal immunoglobulin A (IgA) levels predicted poor survival. Among patients with high and intermediate revised Mayo risk scores, those with physiologically high dROM levels showed better survival than those with lower dROM levels. In this population, dROM was negatively correlated with AST and IgA, which are both correlated with survival. CONCLUSIONS High and intermediate revised Mayo risk score group predicted a poor clinical course in PSC. Additionally, the Child-Pugh score, MELD-Na score, FIB-4 index, and serum IgA were significantly correlated with survival. In patients with high and intermediate revised Mayo risk scores, physiologically high oxidative stress status correlated with low IgA levels and a good prognosis.
Collapse
Affiliation(s)
- Atsushi Oyama
- Department of Gastroenterology and Hepatology, Okayama University, Faculty of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Akinobu Takaki
- Department of Gastroenterology and Hepatology, Okayama University, Faculty of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| | - Takuya Adachi
- Department of Gastroenterology and Hepatology, Okayama University, Faculty of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Nozomu Wada
- Department of Gastroenterology and Hepatology, Okayama University, Faculty of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Yasuto Takeuchi
- Department of Gastroenterology and Hepatology, Okayama University, Faculty of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Hideki Onishi
- Department of Gastroenterology and Hepatology, Okayama University, Faculty of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Hidenori Shiraha
- Department of Gastroenterology and Hepatology, Okayama University, Faculty of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Hiroyuki Okada
- Department of Gastroenterology and Hepatology, Okayama University, Faculty of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Motoyuki Otsuka
- Department of Gastroenterology and Hepatology, Okayama University, Faculty of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| |
Collapse
|
48
|
Wang K, Lin X, Wang T, Zhang X, Cheng W, Xu F, Wang L, Li B, Wang M, Wang W, Zhang M, Ding S, Jin G, Zhu Y, Yang W, Hu A, Zhao Q. Synergistic effects of low-dose arsenic and N-methyl-N'-nitro-N-nitrosoguanidine co-exposure by altering gut microbiota and intestinal metabolic profile in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 263:115195. [PMID: 37418937 DOI: 10.1016/j.ecoenv.2023.115195] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 07/09/2023]
Abstract
Biological organisms are exposed to low-dose arsenic or N-nitro compounds (NOCs) alone or in combination worldwide, especially in areas with high cancer prevalence through drinking water or food exposure; however, information on their combined exposure effects is limited. Here, we conducted an in-depth study of the effects on the gut microbiota, metabolomics, and signaling pathways using rat models exposed to arsenic or N-methyl-N'-nitro-N-nitrosoguanidine (MNNG), one of the most active carcinogenic NOCs, separately or in combination with metabolomics and high-throughput sequencing. Compared to exposure alone, combined exposure to arsenic and MNNG exacerbated damage to gastric tissue morphology, interfered with intestinal microflora and substance metabolism, and exerted a stronger carcinogenic effect. This may be related to intestinal microbiota disorders, including Dyella, Oscillibacter, Myroides, and metabolic pathways such as glycine, serine, and threonine metabolism, arginine biosynthesis, central carbon metabolism in cancer, and purine and pyrimidine metabolism, thereby enhancing the cancer-causing effects of gonadotrophin-releasing hormone (GnRH), P53, and Wnt signaling pathways.
Collapse
Affiliation(s)
- Kexin Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Xiao Lin
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Tingting Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Xiaohui Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Wenli Cheng
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Fang Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Li Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Bin Li
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Min Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Wuqi Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Meng Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Shaopeng Ding
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Guoqing Jin
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Yuting Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Wanshui Yang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Anla Hu
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China.
| | - Qihong Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China.
| |
Collapse
|
49
|
Zhou Y, Duan L, Zeng Y, Song X, Pan K, Niu L, Pu Y, Li J, Khalique A, Fang J, Jing B, Zeng D, Shen B, Ni X. The panda-derived Lactiplantibacillus plantarum BSG201683 improves LPS-induced intestinal inflammation and epithelial barrier disruption in vitro. BMC Microbiol 2023; 23:249. [PMID: 37674107 PMCID: PMC10481503 DOI: 10.1186/s12866-023-02928-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 07/03/2023] [Indexed: 09/08/2023] Open
Abstract
Captive pandas are suffering from intestinal infection due to intestinal microbiota characterized by a high abundance of Enterobacteriaceae induced by long-term captivity. Probiotic supplements showed improvement in intestinal barrier function and inflammation. However, the effects of panda-derived probiotics on the intestinal epithelium and inflammation have not been elucidated. In the present study, lipopolysaccharide (LPS) impaired Caco-2 and RAW264.7 inflammatory models were applied to assess the protection of Lactiplantibacillus plantarum BSG201683 (L. plantarum G83) on barrier disruption and inflammation. The results showed that treatment with L. plantarum G83 significantly decreased the paracellular permeability to fluorescein isothiocyanate conjugated dextran (MW 4000, FITC-D4) after LPS induction. Meanwhile, L. plantarum G83 alleviated the reduction in tight junction (TJ) proteins and downregulated proinflammatory cytokines caused by LPS in Caco-2 cells. L. plantarum G83 also significantly decreased the expression and secretion of pro-inflammatory cytokines in LPS-induced RAW264.7 cells. In addition, the IL-10 increased in both Caco-2 and RAW264.7 cells after L. plantarum G83 treatment. The phagocytosis activity of RAW264.7 cells was significantly increased after L. plantarum G83 treatment. Toll-like receptor 4/ nuclear factor kappa-B (TLR4/NF-κB) signaling pathways were significantly down-regulated after L. plantarum G83 intervention, and the phosphorylation of NF-κB/p65 was consistent with this result. Our findings suggest that L. plantarum G83 improves intestinal inflammation and epithelial barrier disruption in vitro.
Collapse
Affiliation(s)
- Yi Zhou
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Department of Urology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 611130, Sichuan, China
| | - Ling Duan
- Animal Feed Affairs of Sichuan Province, Sichuan Provincial Department of Agriculture and Rural Affairs, Chengdu, 610041, Sichuan, China
| | - Yan Zeng
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Xu Song
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Kangcheng Pan
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Lili Niu
- Chengdu Wildlife Institute, Chengdu Zoo, Chengdu, 610081, Sichuan, China
| | - Yang Pu
- Chengdu Wildlife Institute, Chengdu Zoo, Chengdu, 610081, Sichuan, China
| | - Jiakun Li
- Department of Urology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 611130, Sichuan, China
| | - Abdul Khalique
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Jing Fang
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Bo Jing
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Dong Zeng
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Bairong Shen
- Department of Urology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 611130, Sichuan, China.
| | - Xueqin Ni
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
50
|
Ding N, Xiao H, Zhen L, Li H, Zhang Z, Ge J, Jia H. Systemic cytokines inhibition with Imp7 siRNA nanoparticle ameliorates gut injury in a mouse model of ventilator-induced lung injury. Biomed Pharmacother 2023; 165:115237. [PMID: 37516020 DOI: 10.1016/j.biopha.2023.115237] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 07/31/2023] Open
Abstract
Mechanical ventilation (MV) may negatively affect the lungs and cause the release of inflammatory mediators, resulting in extra-pulmonary organ dysfunction. Studies have revealed systemically elevated levels of proinflammatory cytokines in animal models of ventilator-induced lung injury (VILI); however, whether these cytokines have an effect on gut injury and the mechanisms involved remain unknown. In this study, VILI was generated in mice with high tidal volume mechanical ventilation (20 ml/kg). Tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and IL-6 concentrations in serum and gut measured by ELISA showed significant elevation in the VILI mice. Significant increases in gut injury and PANoptosis were observed in the VILI mice, which were positively correlated with the serum levels of TNF-α, IL-1β, and IL-6. The VILI mice displayed intestinal barrier defects, decreased expressions of occludin and zonula occludin-1 (ZO-1), and increased expression of claudin-2 and the activation of myosin light chain (MLC). Importantly, intratracheal administration of Imp7 siRNA nanoparticle effectively inhibited cytokines production and protected mice from VILI-induced gut injury. These data provide evidence of systemic cytokines contributing to gut injury following VILI and highlight the possibility of targeting cytokines inhibition via Imp7 siRNA nanoparticle as a potential therapeutic intervention for alleviating gut injury following VILI.
Collapse
Affiliation(s)
- Ning Ding
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China; Department of Anesthesiology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China.
| | - Hui Xiao
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Lixiao Zhen
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Huiqing Li
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China; Department of Anesthesiology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Zengzhen Zhang
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China; Department of Anesthesiology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Junke Ge
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China; Department of Intensive Care Medicine, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haiyan Jia
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China; Department of Intensive Care Medicine, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|