1
|
Guvenc F, Danska JS. The intestinal microbiome in type 1 diabetes: bridging early childhood exposures with translational advances. Curr Opin Immunol 2025; 94:102553. [PMID: 40179800 DOI: 10.1016/j.coi.2025.102553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 04/05/2025]
Abstract
Type 1 diabetes (T1D) results from T cell-mediated destruction of pancreatic β-cells, requiring lifelong insulin therapy and glycemic monitoring. While genetic risk, particularly HLA class II, is well established, rising T1D incidence and earlier onset suggest environmental modifiers. Mouse models show that microbiome alterations influence β-cell autoimmunity, and human studies link microbiome composition to T1D, though specific microbial regulators remain unidentified. We examine host-microbiome interactions, including studies implicating enteroviruses in modulating islet autoimmunity. Mechanistic discoveries of microbial effects on diabetes have emerged from mouse model studies. We consider clinical applications, including microbiota-targeted therapies and biomarkers of microbiome-immune crosstalk. Future research should integrate microbial, genetic, environmental, and immune data using multi-omic approaches. Collaborative efforts combining immunology, microbiology, and clinical metadata will drive discovery and precision medicine in T1D.
Collapse
Affiliation(s)
- Furkan Guvenc
- Hospital for Sick Children Research Institute, Program in Genetics and Genome Biology, Department of Immunology, University of Toronto, ON, Canada
| | - Jayne S Danska
- Hospital for Sick Children Research Institute, Program in Genetics and Genome Biology, Department of Immunology, University of Toronto, ON, Canada; Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, ON, Canada.
| |
Collapse
|
2
|
Li Y, Gunderson RC, Xu Z, Ai W, Shen F, Ye J, Xu B, Michie SA. Mucosal Addressin Cell Adhesion Molecule-1 Mediates T Cell Migration into Pancreas-Draining Lymph Nodes for Initiation of the Autoimmune Response in Type 1 Diabetes. Int J Mol Sci 2024; 25:11350. [PMID: 39518902 PMCID: PMC11545416 DOI: 10.3390/ijms252111350] [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: 09/04/2024] [Revised: 10/17/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease that is caused by autoreactive T cell-mediated destruction of insulin-producing β cells in the pancreatic islets. Although naive autoreactive T cells are initially primed by islet antigens in pancreas-draining lymph nodes (pan-LNs), the adhesion molecules that recruit T cells into pan-LNs are unknown. We show that high endothelial venules in pan-LNs of young nonobese diabetic mice have a unique adhesion molecule profile that includes strong expression of mucosal addressin cell adhesion molecule-1 (MAdCAM-1). Anti-MAdCAM-1 antibody blocked more than 80% of the migration of naive autoreactive CD4+ T cells from blood vessels into pan-LNs. Transient blockade of MAdCAM-1 in young nonobese diabetic mice led to increased numbers of autoreactive regulatory CD4+ T cells in pan-LNs and pancreas and to long-lasting protection from T1D. These results indicate the importance of MAdCAM-1 in the development of T1D and suggest MAdCAM-1 as a potential therapeutic target for treating T1D.
Collapse
Affiliation(s)
- Yankui Li
- Department of Vascular Surgery, Tianjin Medical University Second Hospital, Tianjin 300211, China
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.C.G.); (Z.X.); (W.A.); (B.X.); (S.A.M.)
- Department of Surgery, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Rachel C. Gunderson
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.C.G.); (Z.X.); (W.A.); (B.X.); (S.A.M.)
| | - Zeyu Xu
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.C.G.); (Z.X.); (W.A.); (B.X.); (S.A.M.)
- Department of Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45219, USA
| | - Wenjia Ai
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.C.G.); (Z.X.); (W.A.); (B.X.); (S.A.M.)
- Department of Surgery, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Fanru Shen
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.C.G.); (Z.X.); (W.A.); (B.X.); (S.A.M.)
- Department of Surgery, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Jiayu Ye
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA;
| | - Baohui Xu
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.C.G.); (Z.X.); (W.A.); (B.X.); (S.A.M.)
- Department of Surgery, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sara A. Michie
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.C.G.); (Z.X.); (W.A.); (B.X.); (S.A.M.)
| |
Collapse
|
3
|
Yang S, Tong L, Li X, Zhang Y, Chen H, Zhang W, Zhang H, Chen Y, Chen R. A novel clinically relevant human fecal microbial transplantation model in humanized mice. Microbiol Spectr 2024; 12:e0043624. [PMID: 39162553 PMCID: PMC11448399 DOI: 10.1128/spectrum.00436-24] [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: 02/16/2024] [Accepted: 07/12/2024] [Indexed: 08/21/2024] Open
Abstract
The intact immune system of mice exhibits resistance to colonization by exogenous microorganisms, but the gut microbiota profiles of the humanized mice and the patterns of human fecal microbiota colonization remain unexplored. Humanized NCG (huNCG) mice were constructed by injected CD34 +stem cells. 16S rRNA sequencing and fecal microbiota transplantation (FMT) technologies were used to detect the differences in microbiota and selective colonization ability for exogenous community colonization among three mice cohorts (C57BL/6J, NCG, and huNCG). Flow cytometry analysis showed that all huNCG mice had over 25% hCD45 +in peripheral blood. 16S rRNA gene sequence analysis showed that compared with NCG mice, the gut microbiota of huNCG mice were significantly altered. After FMT, the principal coordinates analysis (PCoA) showed that the gut microbial composition of huNCG mice (huNCG-D9) was similar to that of donors. The relative abundance of Firmicutes and Bacteroidetes were significantly increased in huNCG mice compared to NCG mice. Further comparison of ASV sequences revealed that Bacteroides plebeius, Bacteroides finegoldii, Escherichia fergusonii, Escherichia albertii, Klebsiella pneumoniae, and Klebsiella variicola exhibited higher abundance and stability in huNCG mice after FMT. Furthermore, PICRUSt2 analysis showed that huNCG mice had significantly enhanced metabolism and immunity. This study demonstrated that humanized mice are more conducive to colonization within the human gut microbiota, which provides a good method for studying the association between human diseases and microbiota.IMPORTANCEThe gut microbiota and biomarkers of humanized mice are systematically revealed for the first time. The finding that human fecal microbiota colonize humanized mice more stably provides new insights into the study of interactions between immune responses and gut microbiota.
Collapse
Affiliation(s)
- Shuai Yang
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Linglin Tong
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xin Li
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuchen Zhang
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hao Chen
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wei Zhang
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - He Zhang
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Chen
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Renjin Chen
- College of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
4
|
Herold KC, Delong T, Perdigoto AL, Biru N, Brusko TM, Walker LSK. The immunology of type 1 diabetes. Nat Rev Immunol 2024; 24:435-451. [PMID: 38308004 PMCID: PMC7616056 DOI: 10.1038/s41577-023-00985-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2023] [Indexed: 02/04/2024]
Abstract
Following the seminal discovery of insulin a century ago, treatment of individuals with type 1 diabetes (T1D) has been largely restricted to efforts to monitor and treat metabolic glucose dysregulation. The recent regulatory approval of the first immunotherapy that targets T cells as a means to delay the autoimmune destruction of pancreatic β-cells highlights the critical role of the immune system in disease pathogenesis and tends to pave the way for other immune-targeted interventions for T1D. Improving the efficacy of such interventions across the natural history of the disease will probably require a more detailed understanding of the immunobiology of T1D, as well as technologies to monitor residual β-cell mass and function. Here we provide an overview of the immune mechanisms that underpin the pathogenesis of T1D, with a particular emphasis on T cells.
Collapse
Affiliation(s)
- Kevan C Herold
- Department of Immunobiology, Yale University, New Haven, CT, USA.
- Department of Internal Medicine, Yale University, New Haven, CT, USA.
| | - Thomas Delong
- Anschutz Medical Campus, University of Colorado, Denver, CO, USA
| | - Ana Luisa Perdigoto
- Department of Internal Medicine, Yale University, New Haven, CT, USA
- Internal Medicine, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Noah Biru
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Lucy S K Walker
- Institute of Immunity & Transplantation, University College London, London, UK.
- Division of Infection & Immunity, University College London, London, UK.
| |
Collapse
|
5
|
Xie QY, Oh S, Wong A, Yau C, Herold KC, Danska JS. Immune responses to gut bacteria associated with time to diagnosis and clinical response to T cell-directed therapy for type 1 diabetes prevention. Sci Transl Med 2023; 15:eadh0353. [PMID: 37878676 DOI: 10.1126/scitranslmed.adh0353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 10/02/2023] [Indexed: 10/27/2023]
Abstract
Immune-targeted therapies have efficacy for treatment of autoinflammatory diseases. For example, treatment with the T cell-specific anti-CD3 antibody teplizumab delayed disease onset in participants at high risk for type 1 diabetes (T1D) in the TrialNet 10 (TN-10) trial. However, heterogeneity in therapeutic responses in TN-10 and other immunotherapy trials identifies gaps in understanding disease progression and treatment responses. The intestinal microbiome is a potential source of biomarkers associated with future T1D diagnosis and responses to immunotherapy. We previously reported that antibody responses to gut commensal bacteria were associated with T1D diagnosis, suggesting that certain antimicrobial immune responses may help predict disease onset. Here, we investigated anticommensal antibody (ACAb) responses against a panel of taxonomically diverse intestinal bacteria species in sera from TN-10 participants before and after teplizumab or placebo treatment. We identified IgG2 responses to three species that were associated with time to T1D diagnosis and with teplizumab treatment responses that delayed disease onset. These antibody responses link human intestinal bacteria with T1D progression, adding predictive value to known T1D risk factors. ACAb analysis provides a new approach to elucidate heterogeneity in responses to immunotherapy and identify individuals who may benefit from teplizumab, recently approved by the U.S. Food and Drug Administration for delaying T1D onset.
Collapse
Affiliation(s)
- Quin Yuhui Xie
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5T2S8, Canada
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G1X8, Canada
| | - Sean Oh
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G1X8, Canada
| | - Anthony Wong
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G1X8, Canada
| | - Christopher Yau
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G1X8, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario M5T2S8, Canada
| | - Kevan C Herold
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA
- Department of Internal Medicine, Yale University, New Haven, CT 06520, USA
| | - Jayne S Danska
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5T2S8, Canada
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G1X8, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario M5T2S8, Canada
| |
Collapse
|
6
|
Karampitsakos T, Juan-Guardela BM, Tzouvelekis A, Herazo-Maya JD. Precision medicine advances in idiopathic pulmonary fibrosis. EBioMedicine 2023; 95:104766. [PMID: 37625268 PMCID: PMC10469771 DOI: 10.1016/j.ebiom.2023.104766] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/07/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a highly heterogeneous, unpredictable and ultimately lethal chronic lung disease. Over the last decade, two anti-fibrotic agents have been shown to slow disease progression, however, both drugs are administered uniformly with minimal consideration of disease severity and inter-individual molecular, genetic, and genomic differences. Advances in biological understanding of disease endotyping and the emergence of precision medicine have shown that "a one-size-fits-all approach" to the management of chronic lung diseases is no longer appropriate. While precision medicine approaches have revolutionized the management of other diseases such as lung cancer and asthma, the implementation of precision medicine in IPF clinical practice remains an unmet need despite several reports demonstrating a large number of diagnostic, prognostic and theragnostic biomarker candidates in IPF. This review article aims to summarize our current knowledge of precision medicine in IPF and highlight barriers to translate these research findings into clinical practice.
Collapse
Affiliation(s)
- Theodoros Karampitsakos
- Division of Pulmonary, Critical Care and Sleep Medicine, Ubben Center for Pulmonary Fibrosis Research, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Brenda M Juan-Guardela
- Division of Pulmonary, Critical Care and Sleep Medicine, Ubben Center for Pulmonary Fibrosis Research, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | | | - Jose D Herazo-Maya
- Division of Pulmonary, Critical Care and Sleep Medicine, Ubben Center for Pulmonary Fibrosis Research, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
7
|
Zhu J, Jin J, Qi Q, Li L, Zhou J, Cao L, Wang L. The association of gut microbiome with recurrent pregnancy loss: A comprehensive review. Drug Discov Ther 2023; 17:157-169. [PMID: 37357394 DOI: 10.5582/ddt.2023.01010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
The steady-state gut microbiome not only promotes the metabolism and absorption of nutrients that are difficult to digest by the host itself, but also participates in systemic metabolism. Once the dynamic balance is disturbed, the gut microbiome may lead to a variety of diseases. Recurrent pregnancy loss (RPL) affects 1-2% of women of reproductive age, and its prevalence has increased in recent years. According to the literature review, the gut microbiome is a new potential driver of the pathophysiology of recurrent abortion, and the gut microbiome has emerged as a new candidate for clinical prevention and treatment of RPL. However, few studies have concentrated on the direct correlation between RPL and the gut microbiome, and the mechanisms by which the gut microbiome influences recurrent miscarriage need further investigation. In this review, the effects of the gut microbiome on RPL were discussed and found to be associated with inflammatory response, the disruption of insulin signaling pathway and the formation of insulin resistance, maintenance of immunological tolerance at the maternal-fetal interface due to the interference with the immune imbalance of Treg/Th17 cells, and obesity.
Collapse
Affiliation(s)
- Jun Zhu
- The Affiliated Wenling Hospital of Wenzhou Medical University, Zhejiang, China
| | - Jiaxi Jin
- The Affiliated Wenling Hospital of Wenzhou Medical University, Zhejiang, China
| | - Qing Qi
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Lisha Li
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Jing Zhou
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
| | - Liwen Cao
- Center for Reproductive Medicine, Zhoushan Women and Children Hospital, Zhejiang, China
| | - Ling Wang
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| |
Collapse
|
8
|
Hogan G, Winer BY, Ahodantin J, Sellau J, Huang T, Douam F, Funaki M, Chiriboga L, Su L, Ploss A. Persistent hepatitis B virus and HIV coinfections in dually humanized mice engrafted with human liver and immune system. J Med Virol 2023; 95:e28930. [PMID: 37403703 PMCID: PMC11298785 DOI: 10.1002/jmv.28930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 07/06/2023]
Abstract
Chronic hepatitis B (CHB), caused by hepatitis B virus (HBV), remains a major medical problem. HBV has a high propensity for progressing to chronicity and can result in severe liver disease, including fibrosis, cirrhosis, and hepatocellular carcinoma. CHB patients frequently present with viral coinfection, including human immunodeficiency virus type (HIV) and hepatitis delta virus. About 10% of chronic HIV carriers are also persistently infected with HBV, which can result in more exacerbated liver disease. Mechanistic studies of HBV-induced immune responses and pathogenesis, which could be significantly influenced by HIV infection, have been hampered by the scarcity of immunocompetent animal models. Here, we demonstrate that humanized mice dually engrafted with components of a human immune system and a human liver supported HBV infection, which was partially controlled by human immune cells, as evidenced by lower levels of serum viremia and HBV replication intermediates in the liver. HBV infection resulted in priming and expansion of human HLA-restricted CD8+ T cells, which acquired an activated phenotype. Notably, our dually humanized mice support persistent coinfections with HBV and HIV, which opens opportunities for analyzing immune dysregulation during HBV and HIV coinfection, and preclinical testing of novel immunotherapeutics.
Collapse
Affiliation(s)
- Glenn Hogan
- Lewis Thomas Laboratory, Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Benjamin Y Winer
- Lewis Thomas Laboratory, Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - James Ahodantin
- Division of Virology, Pathogenesis and Cancer, Institute of Human Virology, Departments of Pharmacology, Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Julie Sellau
- Lewis Thomas Laboratory, Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Tiffany Huang
- Lewis Thomas Laboratory, Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Florian Douam
- Lewis Thomas Laboratory, Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Masaya Funaki
- Division of Virology, Pathogenesis and Cancer, Institute of Human Virology, Departments of Pharmacology, Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Luis Chiriboga
- Department of Pathology, New York University Medical Center, New York, New York, USA
| | - Lishan Su
- Division of Virology, Pathogenesis and Cancer, Institute of Human Virology, Departments of Pharmacology, Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alexander Ploss
- Lewis Thomas Laboratory, Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|
9
|
RAGE antagonism with azeliragon improves xenograft rejection by T cells in humanized mice. Clin Immunol 2022; 245:109165. [DOI: 10.1016/j.clim.2022.109165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/21/2022]
|
10
|
Application of germ-free NOD-scid IL2rg null mice as a humanized model for tumor microbiome precision medicine. SCIENCE CHINA. LIFE SCIENCES 2021; 64:644-647. [PMID: 33644835 DOI: 10.1007/s11427-020-1876-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 01/05/2021] [Indexed: 12/23/2022]
|
11
|
Ploss A, Strick-Marchand H, Li W. Animal Models for Hepatitis B: Does the Supply Meet the Demand? Gastroenterology 2021; 160:1437-1442. [PMID: 33352166 PMCID: PMC8035324 DOI: 10.1053/j.gastro.2020.11.056] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, New Jersey.
| | - Hélène Strick-Marchand
- Innate Immunity Unit and, Institut National de la Santé et de la Recherche Médicale U1223, Institut Pasteur, Paris, France
| | - Wenhui Li
- National Institute of Biological Sciences and, Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| |
Collapse
|
12
|
Humanization of Immunodeficient Animals for the Modeling of Transplantation, Graft Versus Host Disease, and Regenerative Medicine. Transplantation 2021; 104:2290-2306. [PMID: 32068660 PMCID: PMC7590965 DOI: 10.1097/tp.0000000000003177] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The humanization of animals is a powerful tool for the exploration of human disease pathogenesis in biomedical research, as well as for the development of therapeutic interventions with enhanced translational potential. Humanized models enable us to overcome biologic differences that exist between humans and other species, while giving us a platform to study human processes in vivo. To become humanized, an immune-deficient recipient is engrafted with cells, tissues, or organoids. The mouse is the most well studied of these hosts, with a variety of immunodeficient strains available for various specific uses. More recently, efforts have turned to the humanization of other animal species such as the rat, which offers some technical and immunologic advantages over mice. These advances, together with ongoing developments in the incorporation of human transgenes and additional mutations in humanized mouse models, have expanded our opportunities to replicate aspects of human allotransplantation and to assist in the development of immunotherapies. In this review, the immune and tissue humanization of various species is presented with an emphasis on their potential for use as models for allotransplantation, graft versus host disease, and regenerative medicine.
Collapse
|
13
|
How to overcome the side effects of tumor immunotherapy. Biomed Pharmacother 2020; 130:110639. [PMID: 33658124 DOI: 10.1016/j.biopha.2020.110639] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/12/2020] [Accepted: 08/16/2020] [Indexed: 12/17/2022] Open
Abstract
The incidence of cancer is increasing year by year. Cancer has become one of the health threats of modern people. Simply relying on the surgery, chemotherapy or radiotherapy, not only the survival rate is not high, but also the quality of life of patients is not much better. Fortunately, the emergence and rapid development of cancer immunotherapy have brought more and more exciting results. However, when scientists think it is possible to overcome cancer, they find that not all cancer patients can benefit from immunotherapy, that is to say, the overall efficiency of immunotherapy is not high. Drug resistance and side effects of immunotherapy cannot be ignored. In order to overcome these difficulties, scientists continue to improve the strategy of immunotherapy and find that combination therapy can effectively reduce the incidence of drug resistance. They also found that by reprogramming tumor blood vessels, activating ferroptosis, utilizing thioredoxin, FATP2 and other substances, the therapeutic effect can be improved and side effects can be alleviated. This article reviews the principles of immunotherapy, new strategies to overcome drug resistance of cancer immunotherapy, and how to improve the efficacy of immunotherapy and reduce side effects.
Collapse
|
14
|
Animal Models Used in Hepatitis C Virus Research. Int J Mol Sci 2020; 21:ijms21113869. [PMID: 32485887 PMCID: PMC7312079 DOI: 10.3390/ijms21113869] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
The narrow range of species permissive to infection by hepatitis C virus (HCV) presents a unique challenge to the development of useful animal models for studying HCV, as well as host immune responses and development of chronic infection and disease. Following earlier studies in chimpanzees, several unique approaches have been pursued to develop useful animal models for research while avoiding the important ethical concerns and costs inherent in research with chimpanzees. Genetically related hepatotropic viruses that infect animals are being used as surrogates for HCV in research studies; chimeras of these surrogate viruses harboring specific regions of the HCV genome are being developed to improve their utility for vaccine testing. Concurrently, genetically humanized mice are being developed and continually advanced using human factors known to be involved in virus entry and replication. Further, xenotransplantation of human hepatocytes into mice allows for the direct study of HCV infection in human liver tissue in a small animal model. The current advances in each of these approaches are discussed in the present review.
Collapse
|
15
|
O’Connell AK, Douam F. Humanized Mice for Live-Attenuated Vaccine Research: From Unmet Potential to New Promises. Vaccines (Basel) 2020; 8:E36. [PMID: 31973073 PMCID: PMC7157703 DOI: 10.3390/vaccines8010036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/11/2020] [Accepted: 01/13/2020] [Indexed: 01/24/2023] Open
Abstract
Live-attenuated vaccines (LAV) represent one of the most important medical innovations in human history. In the past three centuries, LAV have saved hundreds of millions of lives, and will continue to do so for many decades to come. Interestingly, the most successful LAVs, such as the smallpox vaccine, the measles vaccine, and the yellow fever vaccine, have been isolated and/or developed in a purely empirical manner without any understanding of the immunological mechanisms they trigger. Today, the mechanisms governing potent LAV immunogenicity and long-term induced protective immunity continue to be elusive, and therefore hamper the rational design of innovative vaccine strategies. A serious roadblock to understanding LAV-induced immunity has been the lack of suitable and cost-effective animal models that can accurately mimic human immune responses. In the last two decades, human-immune system mice (HIS mice), i.e., mice engrafted with components of the human immune system, have been instrumental in investigating the life-cycle and immune responses to multiple human-tropic pathogens. However, their use in LAV research has remained limited. Here, we discuss the strong potential of LAVs as tools to enhance our understanding of human immunity and review the past, current and future contributions of HIS mice to this endeavor.
Collapse
Affiliation(s)
| | - Florian Douam
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, MA 02118, USA;
| |
Collapse
|
16
|
Li Y, Di Santo JP. Modeling Infectious Diseases in Mice with a "Humanized" Immune System. Microbiol Spectr 2019; 7:10.1128/microbiolspec.bai-0019-2019. [PMID: 30953434 PMCID: PMC11590424 DOI: 10.1128/microbiolspec.bai-0019-2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Indexed: 01/07/2023] Open
Abstract
Human immune system (HIS) mice are created by transplanting human immune cells or their progenitor cells into highly immunodeficient recipient mouse hosts, thereby "humanizing" their immune systems. Over past decades, the field of HIS mice has evolved rapidly, as modifications of existing immunodeficient mouse strains have been developed, resulting in increasing levels of human tissue engraftment as humanization is optimized. Current HIS mouse models not only permit elevated levels of human cell engraftment but also demonstrate graft stability. As such, HIS mice are being extensively used to study the human innate and adaptive immune response against microbial infections in vivo. Compared to nonhumanized animal models, which are frequently infected with surrogate or adapted microbes, the HIS mouse models allow the analysis of interactions between human immune cells and bona fide pathogenic microbes, making them a more clinically relevant model. This article reviews the development of HIS mice and covers the different strategies used to humanize mice, as well as discussing the use of HIS mice for studying bacterial infections that cause human disease.
Collapse
Affiliation(s)
- Yan Li
- Innate Immunity Unit, Immunology Department, Institut Pasteur, Paris, France
- Inserm U1223, Paris, France
| | - James P Di Santo
- Innate Immunity Unit, Immunology Department, Institut Pasteur, Paris, France
- Inserm U1223, Paris, France
| |
Collapse
|
17
|
Douam F, Ziegler CGK, Hrebikova G, Fant B, Leach R, Parsons L, Wang W, Gaska JM, Winer BY, Heller B, Shalek AK, Ploss A. Selective expansion of myeloid and NK cells in humanized mice yields human-like vaccine responses. Nat Commun 2018; 9:5031. [PMID: 30487575 PMCID: PMC6262001 DOI: 10.1038/s41467-018-07478-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 10/29/2018] [Indexed: 12/21/2022] Open
Abstract
Mice engrafted with components of a human immune system have become widely-used models for studying aspects of human immunity and disease. However, a defined methodology to objectively measure and compare the quality of the human immune response in different models is lacking. Here, by taking advantage of the highly immunogenic live-attenuated yellow fever virus vaccine YFV-17D, we provide an in-depth comparison of immune responses in human vaccinees, conventional humanized mice, and second generation humanized mice. We demonstrate that selective expansion of human myeloid and natural killer cells promotes transcriptomic responses akin to those of human vaccinees. These enhanced transcriptomic profiles correlate with the development of an antigen-specific cellular and humoral response to YFV-17D. Altogether, our approach provides a robust scoring of the quality of the human immune response in humanized mice and highlights a rational path towards developing better pre-clinical models for studying the human immune response and disease.
Collapse
Affiliation(s)
- Florian Douam
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Washington Road, Princeton, NJ, 08544, USA
| | - Carly G K Ziegler
- Institute for Medical Engineering & Science (IMES), MIT, Cambridge, MA, 02139, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA, 02139, USA
- Graduate Program in Biophysics, Harvard Medical School, Boston, MA, 02139, USA
| | - Gabriela Hrebikova
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Washington Road, Princeton, NJ, 08544, USA
| | - Bruno Fant
- Department of Psychiatry, Center for Neurobiology and Behavior, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Robert Leach
- Lewis Sigler Institute for Integrative Genomics, Genomics Core, Carl Icahn Laboratory, Princeton University, Princeton, NJ, 19104, USA
| | - Lance Parsons
- Lewis Sigler Institute for Integrative Genomics, Genomics Core, Carl Icahn Laboratory, Princeton University, Princeton, NJ, 19104, USA
| | - Wei Wang
- Lewis Sigler Institute for Integrative Genomics, Genomics Core, Carl Icahn Laboratory, Princeton University, Princeton, NJ, 19104, USA
| | - Jenna M Gaska
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Washington Road, Princeton, NJ, 08544, USA
| | - Benjamin Y Winer
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Washington Road, Princeton, NJ, 08544, USA
| | - Brigitte Heller
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Washington Road, Princeton, NJ, 08544, USA
| | - Alex K Shalek
- Institute for Medical Engineering & Science (IMES), MIT, Cambridge, MA, 02139, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA, 02139, USA
- Graduate Program in Biophysics, Harvard Medical School, Boston, MA, 02139, USA
| | - Alexander Ploss
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Washington Road, Princeton, NJ, 08544, USA.
| |
Collapse
|
18
|
Diosgenin promotes antitumor immunity and PD-1 antibody efficacy against melanoma by regulating intestinal microbiota. Cell Death Dis 2018; 9:1039. [PMID: 30305604 PMCID: PMC6179990 DOI: 10.1038/s41419-018-1099-3] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/12/2018] [Accepted: 09/14/2018] [Indexed: 12/30/2022]
Abstract
Diosgenin, a natural steroidal saponin, can exert antitumor effect by regulating immune function and improving intestinal microbiota. The response to anti-PD-1 immunotherapy is associated with intestinal microbiota and effector T cells in tumor microenvironment. We hypothesize that the modulation of diosgenin on intestinal microbiota can facilitate antitumor immunity and the therapeutic efficacy of PD-1 antibody. In melanoma-bearing C57BL/6 mice, we observed that the anti-melanoma effect of diosgenin relied more on antitumor immunity than direct tumor inhibition activity evidenced by obvious CD4+/CD8+ T-cell infiltration and IFN-γ expression in tumor tissues, and it could improve the compositions of intestinal microbiota. Antibiotics impaired the therapeutic efficacy and immunity responses of diosgenin through disturbing intestinal microbiota, indicating the importance of intestinal microbiota in diosgenin’s in vivo antitumor activity. More importantly, the combined administration of PD-1 antibody with diosgenin aggravated the tumor necrosis and apoptosis by eliciting augmented T-cell responses. Taken together, diosgenin can be used as a microecological regulator to induce antitumor immunity and improve the efficacy of immune checkpoint antibody, making it more suitable for the treatment of malignant tumors.
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW The incidence of type 1 diabetes (T1D) is rising drastically for the past decades at a rate that cannot be explained by genetic changes alone. Environmental changes are considered to be the main drivers of this change. Recently, the gut microbiota has been suggested as a missing link between known environmental disease modulators and T1D promotion. Lifestyle factors have changed over time and have altered the gut microbiota-host interaction affecting T1D development. The purpose of this review is to discuss recent data emphasizing the modulatory potential of early lifestyle factors on gut microbiota and to elucidate their implication for T1D. RECENT FINDINGS Recent findings show that lifestyle factors, especially those that affect the early establishment of gut homeostasis and the education of the immune system, are crucial disease modulators. Changing lifestyle factors affecting the early establishment of gut homeostasis are suggested to be key drivers of the rising T1D incidence.
Collapse
Affiliation(s)
- Elke Gülden
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
20
|
Abstract
Although immunotherapy has been remarkably effective across multiple cancer types, there continues to be a significant number of non-responding patients. A possible factor proposed to influence the efficacy of immunotherapies is the gut microbiome. We discuss the results and implications of recent research on the relationship between the gut microbiome, our immune systems, and immune checkpoint inhibitor therapies including anti-CTLA-4 Ab and anti-PD-1 Ab. While the investigations all exhibit interesting results and conclusions, we find little congruence in the specific bacteria that were found favorable for antitumor responses. It is unclear whether the inconsistencies are due to differential approaches in study design (pre-clinical or clinical subjects, anti-CTLA-4 Ab or anti-PD-1 Ab), experimental methods and measurements (metagenomics sequencing and clustering variations) or subject population dynamics (differential cancer types and baseline characteristics). Moreover, we note studies regarding particular bacterial commensals and autoimmune diseases, which challenge findings from these investigations. We conclude that with the current research, clinical investigators can appreciate the critical role of gut microbiota in mediating immunostimulant response. However, prospective research exploring the biochemical mechanisms which commensal bacteria communicate with each other and the immune system is imperative to understand how they can be adjusted properly for higher immunotherapy response.
Collapse
Affiliation(s)
- Audrey Humphries
- a Department of Medicine , Division of Hematology/Oncology, University of California San Francisco , San Francisco , CA , USA
| | - Adil Daud
- a Department of Medicine , Division of Hematology/Oncology, University of California San Francisco , San Francisco , CA , USA
| |
Collapse
|
21
|
Abstract
Type 1 diabetes (T1D) is a multifactorial autoimmune disease whose etiology involves complex interactions between the immune system and the intestinal microbiota. Recent studies by Rouxel et al. (2017) suggest that innate-like mucosal-associated invariant T cells (MAIT cells) may link gut integrity, the microbiota, and T1D.
Collapse
Affiliation(s)
- Elke Gülden
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA
| | - Noah Palm
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA
| | - Kevan C Herold
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|