51
|
Huang W, Yu J, Liu T, Defnet AE, Zalesak S, Farese AM, MacVittie TJ, Kane MA. Acute Proteomic Changes in Lung after Radiation: Toward Identifying Initiating Events of Delayed Effects of Acute Radiation Exposure in Non-human Primate after Partial Body Irradiation with Minimal Bone Marrow Sparing. HEALTH PHYSICS 2021; 121:384-394. [PMID: 34546219 PMCID: PMC8546870 DOI: 10.1097/hp.0000000000001476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
ABSTRACT Radiation-induced lung injury is a delayed effect of acute radiation exposure resulting in pulmonary pneumonitis and fibrosis. Molecular mechanisms that lead to radiation-induced lung injury remain incompletely understood. Using a non-human primate model of partial body irradiation with minimal bone marrow sparing, lung was analyzed from animals irradiated with 12 Gy at timepoints every 4 d up to 21 d after irradiation and compared to non-irradiated (sham) controls. Tryptic digests of lung tissues were analyzed by liquid chromatography-tandem mass spectrometry followed by pathway analysis. Out of the 3,101 unique proteins that were identified, we found that 252 proteins showed significant and consistent responses across at least three time points post-irradiation, of which 215 proteins showed strong up-regulation while 37 proteins showed down-regulation. Canonical pathways affected by irradiation, changes in proteins that serve as upstream regulators, and proteins involved in key processes including inflammation, fibrosis, and retinoic acid signaling were identified. The proteomic profiling of lung conducted here represents an untargeted systems biology approach to identify acute molecular events in the non-human primate lung that could potentially be initiating events for radiation-induced lung injury.
Collapse
Affiliation(s)
- Weiliang Huang
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, USA
| | - Jianshi Yu
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, USA
| | - Tian Liu
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, USA
| | - Amy E Defnet
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, USA
| | - Stephanie Zalesak
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, USA
| | - Ann M. Farese
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD 21201
| | - Thomas J. MacVittie
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD 21201
| | - Maureen A Kane
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, USA
| |
Collapse
|
52
|
Harriman R, Lewis JS. Bioderived materials that disarm the gut mucosal immune system: Potential lessons from commensal microbiota. Acta Biomater 2021; 133:187-207. [PMID: 34098091 DOI: 10.1016/j.actbio.2021.05.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/25/2021] [Accepted: 05/20/2021] [Indexed: 12/12/2022]
Abstract
Over the course of evolution, mammals and gut commensal microbes have adapted to coexist with each other. This homeostatic coexistence is dependent on an intricate balance between tolerogenic and inflammatory responses directed towards beneficial, commensal microbes and pathogenic intruders, respectively. Immune tolerance towards the gut microflora is largely sustained by immunomodulatory molecules produced by the commensals, which protect the bacteria from immune advances and maintain the gut's unique tolerogenic microenvironment, as well as systemic homeostasis. The identification and characterization of commensal-derived, tolerogenic molecules could lead to their utilization in biomaterials-inspired delivery schemes involving nano/microparticles or hydrogels, and potentially lead to the next generation of commensal-derived therapeutics. Moreover, gut-on-chip technologies could augment the discovery and characterization of influential commensals by providing realistic in vitro models conducive to finicky microbes. In this review, we provide an overview of the gut immune system, describe its intricate relationships with the microflora and identify major genera involved in maintaining tolerogenic responses and peripheral homeostasis. More relevant to biomaterials, we discuss commensal-derived molecules that are known to interface with immune cells and discuss potential strategies for their incorporation into biomaterial-based strategies aimed at culling inflammatory diseases. We hope this review will bridge the current findings in gut immunology, microbiology and biomaterials and spark further investigation into this emerging field. STATEMENT OF SIGNIFICANCE: Despite its tremendous potential to culminate into revolutionary therapeutics, the synergy between immunology, microbiology, and biomaterials has only been explored at a superficial level. Strategic incorporation of biomaterial-based technologies may be necessary to fully characterize and capitalize on the rapidly growing repertoire of immunomodulatory molecules derived from commensal microbes. Bioengineers may be able to combine state-of-the-art delivery platforms with immunomodulatory cues from commensals to provide a more holistic approach to combating inflammatory disease. This interdisciplinary approach could potentiate a neoteric field of research - "commensal-inspired" therapeutics with the promise of revolutionizing the treatment of inflammatory disease.
Collapse
Affiliation(s)
- Rian Harriman
- University of California Davis, Department of Biomedical Engineering, Davis, CA 95616, USA
| | - Jamal S Lewis
- University of California Davis, Department of Biomedical Engineering, Davis, CA 95616, USA.
| |
Collapse
|
53
|
Jacobse J, Li J, Rings EHHM, Samsom JN, Goettel JA. Intestinal Regulatory T Cells as Specialized Tissue-Restricted Immune Cells in Intestinal Immune Homeostasis and Disease. Front Immunol 2021; 12:716499. [PMID: 34421921 PMCID: PMC8371910 DOI: 10.3389/fimmu.2021.716499] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/16/2021] [Indexed: 12/28/2022] Open
Abstract
FOXP3+ regulatory T cells (Treg cells) are a specialized population of CD4+ T cells that restrict immune activation and are essential to prevent systemic autoimmunity. In the intestine, the major function of Treg cells is to regulate inflammation as shown by a wide array of mechanistic studies in mice. While Treg cells originating from the thymus can home to the intestine, the majority of Treg cells residing in the intestine are induced from FOXP3neg conventional CD4+ T cells to elicit tolerogenic responses to microbiota and food antigens. This process largely takes place in the gut draining lymph nodes via interaction with antigen-presenting cells that convert circulating naïve T cells into Treg cells. Notably, dysregulation of Treg cells leads to a number of chronic inflammatory disorders, including inflammatory bowel disease. Thus, understanding intestinal Treg cell biology in settings of inflammation and homeostasis has the potential to improve therapeutic options for patients with inflammatory bowel disease. Here, the induction, maintenance, trafficking, and function of intestinal Treg cells is reviewed in the context of intestinal inflammation and inflammatory bowel disease. In this review we propose intestinal Treg cells do not compose fixed Treg cell subsets, but rather (like T helper cells), are plastic and can adopt different programs depending on microenvironmental cues.
Collapse
Affiliation(s)
- Justin Jacobse
- Department of Pediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, Netherlands
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, United States
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jing Li
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, United States
| | - Edmond H. H. M. Rings
- Department of Pediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, Netherlands
- Department of Pediatrics, Sophia Children’s Hospital, Erasmus University, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Janneke N. Samsom
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jeremy A. Goettel
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, United States
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
54
|
Shawki MA, Elsayed NS, Mantawy EM, Said RS. Promising drug repurposing approach targeted for cytokine storm implicated in SARS-CoV-2 complications. Immunopharmacol Immunotoxicol 2021; 43:395-409. [PMID: 34057871 PMCID: PMC8171013 DOI: 10.1080/08923973.2021.1931302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 04/04/2021] [Indexed: 12/16/2022]
Abstract
A global threat has emerged in 2019 due to the rapid spread of Coronavirus disease (COVID-19). As of January 2021, the number of cases worldwide reached 103 million cases and 2.22 million deaths which were confirmed as the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This global pandemic galvanized the scientific community to study the causative virus (SARS-CoV2) pathogenesis, transmission, and clinical symptoms. Remarkably, the most common complication associated with this disease is the cytokine storm which is responsible for COVID-19 mortality. Thus, targeting the cytokine storm with new medications is needed to hamper COVID-19 complications where the most prominent strategy for the treatment is drug repurposing. Through this strategy, several steps are skipped especially those required for testing drug safety and thus may help in reducing the dissemination of this pandemic. Accordingly, the aim of this review is to outline the pathogenesis, clinical features, and immune complications of SARS-CoV2 in addition to suggesting several repurposed drugs with their plausible mechanism of action for possible management of severe COVID-19 cases.
Collapse
Affiliation(s)
- May Ahmed Shawki
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Noha Salah Elsayed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Eman M. Mantawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Riham S. Said
- Department of Drug Radiation Research, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
55
|
Bos AV, Erkelens MN, Koenders STA, van der Stelt M, van Egmond M, Mebius RE. Clickable Vitamins as a New Tool to Track Vitamin A and Retinoic Acid in Immune Cells. Front Immunol 2021; 12:671283. [PMID: 34305901 PMCID: PMC8298001 DOI: 10.3389/fimmu.2021.671283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/02/2021] [Indexed: 01/24/2023] Open
Abstract
The vitamin A derivative, retinoid acid (RA) is key player in guiding adaptive mucosal immune responses. However, data on the uptake and metabolism of vitamin A within human immune cells has remained largely elusive because retinoids are small, lipophilic molecules which are difficult to detect. To overcome this problem and to be able to study the effect of vitamin A metabolism in human immune cell subsets, we have synthesized novel bio-orthogonal retinoid-based probes (clickable probes), which are structurally and functionally indistinguishable from vitamin A. The probes contain a functional group (an alkyne) to conjugate to a fluorogenic dye to monitor retinoid molecules in real-time in immune cells. We demonstrate, by using flow cytometry and microscopy, that multiple immune cells have the capacity to internalize retinoids to varying degrees, including human monocyte-derived dendritic cells (DCs) and naïve B lymphocytes. We observed that naïve B cells lack the enzymatic machinery to produce RA, but use exogenous retinoic acid to enhance CD38 expression. Furthermore, we showed that human DCs metabolize retinal into retinoic acid, which in co-culture with naïve B cells led to of the induction of CD38 expression. These data demonstrate that in humans, DCs can serve as an exogenous source of RA for naïve B cells. Taken together, through the use of clickable vitamins our data provide valuable insight in the mechanism of vitamin A metabolism and its importance for human adaptive immunity.
Collapse
Affiliation(s)
- Amelie V Bos
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands
| | - Martje N Erkelens
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands
| | - Sebastiaan T A Koenders
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands.,Department of Surgery, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands
| |
Collapse
|
56
|
Nagy NA, de Haas AM, Geijtenbeek TBH, van Ree R, Tas SW, van Kooyk Y, de Jong EC. Therapeutic Liposomal Vaccines for Dendritic Cell Activation or Tolerance. Front Immunol 2021; 12:674048. [PMID: 34054859 PMCID: PMC8155586 DOI: 10.3389/fimmu.2021.674048] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells (DCs) are paramount in initiating and guiding immunity towards a state of activation or tolerance. This bidirectional capacity of DCs sets them at the center stage for treatment of cancer and autoimmune or allergic conditions. Accordingly, many clinical studies use ex vivo DC vaccination as a strategy to boost anti-tumor immunity or to suppress immunity by including vitamin D3, NF-κB inhibitors or retinoic acid to create tolerogenic DCs. As harvesting DCs from patients and differentiating these cells in vitro is a costly and cumbersome process, in vivo targeting of DCs has huge potential as nanoparticulate platforms equipped with activating or tolerogenic adjuvants can modulate DCs in their natural environment. There is a rapid expansion of the choices of nanoparticles and activation- or tolerance-promoting adjuvants for a therapeutic vaccine platform. In this review we highlight the most recent nanomedical approaches aimed at inducing immune activation or tolerance via targeting DCs, together with novel fundamental insights into the mechanisms inherent to fostering anti-tumor or tolerogenic immunity.
Collapse
Affiliation(s)
- Noémi Anna Nagy
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Aram M. de Haas
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Teunis B. H. Geijtenbeek
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Ronald van Ree
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
- Department of Otorhinolaryngology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Sander W. Tas
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, Amsterdam Rheumatology and Immunology Center, University of Amsterdam, Amsterdam, Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Esther C. de Jong
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
57
|
Mayorga C, Palomares F, Cañas JA, Pérez-Sánchez N, Núñez R, Torres MJ, Gómez F. New Insights in Therapy for Food Allergy. Foods 2021; 10:foods10051037. [PMID: 34068667 PMCID: PMC8151532 DOI: 10.3390/foods10051037] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/24/2021] [Accepted: 05/05/2021] [Indexed: 12/23/2022] Open
Abstract
Food allergy is an increasing problem worldwide, with strict avoidance being classically the only available reliable treatment. The main objective of this review is to cover the latest information about the tools available for the diagnosis and treatment of food allergies. In recent years, many efforts have been made to better understand the humoral and cellular mechanisms involved in food allergy and to improve the strategies for diagnosis and treatment. This review illustrates IgE-mediated food hypersensitivity and provides a current description of the diagnostic strategies and advances in different treatments. Specific immunotherapy, including different routes of administration and new therapeutic approaches, such as hypoallergens and nanoparticles, are discussed in detail. Other treatments, such as biologics and microbiota, are also described. Therefore, we conclude that although important efforts have been made in improving therapies for food allergies, including innovative approaches mainly focusing on efficacy and safety, there is an urgent need to develop a set of basic and clinical results to help in the diagnosis and treatment of food allergies.
Collapse
Affiliation(s)
- Cristobalina Mayorga
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga (IBIMA), 29009 Málaga, Spain; (F.P.); (J.A.C.); (R.N.)
- Allergy Clinical Unit, Hospital Regional Universitario de Málaga, 29071 Málaga, Spain; (N.P.-S.); (M.J.T.); (F.G.)
- Correspondence: ; Tel.: +34-951-290-224
| | - Francisca Palomares
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga (IBIMA), 29009 Málaga, Spain; (F.P.); (J.A.C.); (R.N.)
| | - José A. Cañas
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga (IBIMA), 29009 Málaga, Spain; (F.P.); (J.A.C.); (R.N.)
| | - Natalia Pérez-Sánchez
- Allergy Clinical Unit, Hospital Regional Universitario de Málaga, 29071 Málaga, Spain; (N.P.-S.); (M.J.T.); (F.G.)
| | - Rafael Núñez
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga (IBIMA), 29009 Málaga, Spain; (F.P.); (J.A.C.); (R.N.)
| | - María José Torres
- Allergy Clinical Unit, Hospital Regional Universitario de Málaga, 29071 Málaga, Spain; (N.P.-S.); (M.J.T.); (F.G.)
- Medicine Department, Universidad de Málaga-UMA, 29071 Málaga, Spain
| | - Francisca Gómez
- Allergy Clinical Unit, Hospital Regional Universitario de Málaga, 29071 Málaga, Spain; (N.P.-S.); (M.J.T.); (F.G.)
| |
Collapse
|
58
|
Lerner A. The intestinal luminal sources of α-synuclein: a gastroenterologist perspective. Nutr Rev 2021; 80:282-293. [PMID: 33942062 DOI: 10.1093/nutrit/nuab024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease is characterized by nonmotor/motor dysfunction, midbrain dopaminergic neuronal death, and α-synuclein (aSN) deposits. The current hypothesis is that aSN accumulates in the enteric nervous system to reach the brain. However, invertebrate, vertebrate, and nutritional sources of aSN reach the luminal compartment. Submitted to local amyloidogenic forces, the oligomerized proteins' cargo can be sensed and sampled by a specialized mucosal cell to be transmitted to the adjacent enteric nervous system, starting their upward journey to the brain. The present narrative review extends the current mucosal origin of Parkinson's disease, presenting the possibility that the disease starts in the intestinal lumen. If substantiated, eliminating the nutritional sources of aSN (eg, applying a vegetarian diet) might revolutionize the currently used dopaminergic pharmacologic therapy.
Collapse
Affiliation(s)
- Aaron Lerner
- A. Lerner is with the Zabludowicz Center for Autoimmune Diseases, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| |
Collapse
|
59
|
Müller TM, Becker E, Wiendl M, Schulze LL, Voskens C, Völkl S, Kremer AE, Neurath MF, Zundler S. Circulating Adaptive Immune Cells Expressing the Gut Homing Marker α4β7 Integrin Are Decreased in COVID-19. Front Immunol 2021; 12:639329. [PMID: 33959123 PMCID: PMC8093414 DOI: 10.3389/fimmu.2021.639329] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Background Infection with the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes a wide range of symptoms including gastrointestinal manifestations, and intestinal epithelial cells are a target of the virus. However, it is unknown how the intestinal immune system contributes to systemic immune responses in coronavirus disease 2019 (COVID-19). Methods We characterized peripheral blood lymphocytes from patients with active COVID-19 and convalescent patients as well as healthy controls by flow cytometry. Results The frequency and absolute number of circulating memory T and B cells expressing the gut homing integrin α4β7 integrin was reduced during COVID-19, whether gastrointestinal symptoms were present or not. While total IgA-expressing B cells were increased, gut-imprinted B cells with IgA expression were stable. Conclusion COVID-19 is associated with a decrease in circulating adaptive immune cells expressing the key gut homing marker α4β7 suggesting that these cells are preferentially recruited to extra-intestinal tissues independently of α4β7 or that the systemic immune response against SARS-CoV-2 is at least numerically dominated by extraintestinal, particularly pulmonary, immune cell priming.
Collapse
Affiliation(s)
- Tanja M Müller
- Department of Medicine 1 and Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Emily Becker
- Department of Medicine 1 and Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Maximilian Wiendl
- Department of Medicine 1 and Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lisa Lou Schulze
- Department of Medicine 1 and Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Caroline Voskens
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Simon Völkl
- Department of Internal Medicine 5, Hematology and Clinical Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas E Kremer
- Department of Medicine 1 and Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1 and Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian Zundler
- Department of Medicine 1 and Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
60
|
Miah M, Goh I, Haniffa M. Prenatal Development and Function of Human Mononuclear Phagocytes. Front Cell Dev Biol 2021; 9:649937. [PMID: 33898444 PMCID: PMC8060508 DOI: 10.3389/fcell.2021.649937] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
The human mononuclear phagocyte (MP) system, which includes dendritic cells, monocytes, and macrophages, is a critical regulator of innate and adaptive immune responses. During embryonic development, MPs derive sequentially in yolk sac progenitors, fetal liver, and bone marrow haematopoietic stem cells. MPs maintain tissue homeostasis and confer protective immunity in post-natal life. Recent evidence - primarily in animal models - highlight their critical role in coordinating the remodeling, maturation, and repair of target organs during embryonic and fetal development. However, the molecular regulation governing chemotaxis, homeostasis, and functional diversification of resident MP cells in their respective organ systems during development remains elusive. In this review, we summarize the current understanding of the development and functional contribution of tissue MPs during human organ development and morphogenesis and its relevance to regenerative medicine. We outline how single-cell multi-omic approaches and next-generation ex-vivo organ-on-chip models provide new experimental platforms to study the role of human MPs during development and disease.
Collapse
Affiliation(s)
- Mohi Miah
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Issac Goh
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom.,Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Wellcome Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|
61
|
Yang G, Wei J, Liu P, Zhang Q, Tian Y, Hou G, Meng L, Xin Y, Jiang X. Role of the gut microbiota in type 2 diabetes and related diseases. Metabolism 2021; 117:154712. [PMID: 33497712 DOI: 10.1016/j.metabol.2021.154712] [Citation(s) in RCA: 153] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 12/27/2020] [Accepted: 01/20/2021] [Indexed: 02/08/2023]
Abstract
Type 2 diabetes is the fastest-growing metabolic disease in the world. Many clinical studies have found that type 2 diabetes patients have metabolic disorders and chronic inflammatory states accompanied by disturbances in the gut microbiota. The gut microbiota plays an important role in body metabolism and immune regulation, and disturbances in the gut microbiota in conjunction with destruction of the intestinal barrier in type 2 diabetes patients causes damage to multiple organs. Therefore, the gut microbiota may be a new therapeutic target for treating type 2 diabetes and related diseases. In this review, we introduce the characteristics of the gut microbiota in type 2 diabetes and related diseases, as well as highlight the potential molecular mechanisms of their effects on intestinal barrier disruption, metabolic disorders, and chronic inflammation. Finally, we summarize an intestinal microecological therapeutic strategy, with a focus on shaping the intestinal bacteria, to improve the malignant progress of type 2 diabetes and related diseases. AUTHOR SUMMARY: Type 2 diabetes (T2D) is the fastest-growing metabolic disease in the world. Many clinical studies have found that T2D patients have metabolic disorders and chronic inflammatory states, accompanied by disturbances of the gut microbiota and increased intestinal permeability. The number of human gut microbiota is more than 10 times of human cells, and they play an important role in the body's metabolism and immune regulation. The abnormal intestinal metabolites and intestinal barrier disruption caused by the gut microbiota dysbiosis in the T2D facilitate intestinal bacteria and their harmful metabolites entering the circulatory system. The abnormal entering will cause the damage to multiple organs through disturbing insulin sensitivity, glucose metabolism, and immune homeostasis. Therefore, the gut microbiota may be a new therapeutic target for improving T2D and its related diseases. In this review, we introduce the compositional characteristics of the gut microbiota in T2D, and highlight some new molecular mechanisms of their effects on intestinal barrier disruption, metabolic disorders and chronic inflammation in T2D and its related diseases. Finally, we summarize an intestinal microecological therapeutic strategy, with a focus on shaping the intestinal bacteria, to improve the malignant progress of T2D and related diseases.
Collapse
Affiliation(s)
- Ge Yang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China
| | - Jinlong Wei
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China
| | - Pinyi Liu
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China
| | - Qihe Zhang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China
| | - Yuan Tian
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China; Department of Gynecology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Guowen Hou
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China
| | - Lingbin Meng
- Department of Hematology and Medical Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
62
|
Khan I, Daniell H. Oral delivery of therapeutic proteins bioencapsulated in plant cells: preclinical and clinical advances. Curr Opin Colloid Interface Sci 2021; 54. [PMID: 33967586 DOI: 10.1016/j.cocis.2021.101452] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Oral delivery of protein drugs (PDs) made in plant cells could revolutionize current approaches of their production and delivery. Expression of PDs reduces their production cost by elimination of prohibitively expensive fermentation, purification, cold transportation/storage, and sterile injections and increases their shelf life for several years. Ability of plant cell wall to protect PDs from digestive acids/enzymes, commensal bacteria to release PDs in gut lumen after lysis of plant cell wall and role of GALT in inducing tolerance facilitate prevention or treatment allergic, autoimmune diseases or anti-drug antibody responses. Delivery of functional proteins facilitate treatment of inherited or metabolic disorders. Recent advances in making PDs free of antibiotic resistance genes in edible plant cells, long-term storage at ambient temperature maintaining their efficacy, production in cGMP facilities, IND enabling studies for clinical advancement and FDA approval of orally delivered PDs augur well for advancing this novel drug delivery platform technology.
Collapse
Affiliation(s)
- Imran Khan
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Henry Daniell
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
63
|
Li Z, McCafferty KJ, Judd RL. Role of HCA 2 in Regulating Intestinal Homeostasis and Suppressing Colon Carcinogenesis. Front Immunol 2021; 12:606384. [PMID: 33708203 PMCID: PMC7940178 DOI: 10.3389/fimmu.2021.606384] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/05/2021] [Indexed: 12/23/2022] Open
Abstract
Hydroxycarboxylic acid receptor 2 (HCA2) is vital for sensing intermediates of metabolism, including β-hydroxybutyrate and butyrate. It also regulates profound anti-inflammatory effects in various tissues, indicating that HCA2 may serve as an essential therapeutic target for mediating inflammation-associated diseases. Butyrate and niacin, endogenous and exogenous ligands of HCA2, have been reported to play an essential role in maintaining intestinal homeostasis. HCA2, predominantly expressed in diverse immune cells, is also present in intestinal epithelial cells (IECs), where it regulates the intricate communication network between diet, microbiota, and immune cells. This review summarizes the physiological role of HCA2 in intestinal homeostasis and its pathological role in intestinal inflammation and cancer.
Collapse
Affiliation(s)
- Zhuoyue Li
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Kayleen J McCafferty
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Robert L Judd
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| |
Collapse
|
64
|
Zhou Y, Takano T, Wang Y, Li X, Wang R, Wakatsuki Y, Nakajima-Adachi H, Tanokura M, Miyakawa T, Hachimura S. Intestinal regulatory T cell induction by β-elemene alleviates the formation of fat tissue-related inflammation. iScience 2021; 24:101883. [PMID: 33364577 PMCID: PMC7750371 DOI: 10.1016/j.isci.2020.101883] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/20/2020] [Accepted: 11/25/2020] [Indexed: 12/24/2022] Open
Abstract
The role of the intestinal immune system in the inhibition of fat tissue-related inflammation by dietary material is yet to be elucidated. Oral administration of β-elemene, contained in various foodstuffs, downregulated expressions of inflammatory cytokines and increased Foxp3+CD4+ T cells in adipose tissue of obese mice. However, β-elemene did not affect the inflammatory response of adipose tissue in vitro, suggesting that the inhibition observed in vivo was not due to direct interactions of adipose tissue with β-elemene. Instead, β-elemene increased Foxp3+CD4+ T cell population enhancing gene expressions of transforming growth factor β 1, retinaldehyde dehydrogenase 2, integrin αvβ8, and interleukin-10 in intestinal dendritic cells (DCs) in vivo and in vitro. Taken together, this study suggested the therapeutic effects of β-elemene on treating experimental obesity-induced chronic inflammation by adjusting the balance of immune cell populations in fat tissue through the generation of regulatory T cells in the intestinal immune system by modulating DC function.
Collapse
Affiliation(s)
- Yingyu Zhou
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Tomohiro Takano
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Yimei Wang
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Xuyang Li
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Rong Wang
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Yoshio Wakatsuki
- Department of Clinical Bio-regulatory Science, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Haruyo Nakajima-Adachi
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Masaru Tanokura
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Takuya Miyakawa
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Satoshi Hachimura
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
65
|
The demethylase inhibitor GSK-J4 limits inflammatory colitis by promoting de novo synthesis of retinoic acid in dendritic cells. Sci Rep 2021; 11:1342. [PMID: 33446666 PMCID: PMC7809056 DOI: 10.1038/s41598-020-79122-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 11/26/2020] [Indexed: 02/08/2023] Open
Abstract
Dendritic cells (DCs) promote T-cell mediated tolerance to self-antigens and induce inflammation to innocuous-antigens. This dual potential makes DCs fundamental players in inflammatory disorders. Evidence from inflammatory colitis mouse models and inflammatory bowel diseases (IBD) patients indicated that gut inflammation in IBD is driven mainly by T-helper-1 (Th1) and Th17 cells, suggesting an essential role for DCs in the development of IBD. Here we show that GSK-J4, a selective inhibitor of the histone demethylase JMJD3/UTX, attenuated inflammatory colitis by reducing the inflammatory potential and increasing the tolerogenic features of DCs. Mechanistic analyses revealed that GSK-J4 increased activating epigenetic signals while reducing repressive marks in the promoter of retinaldehyde dehydrogenase isoforms 1 and 3 in DCs, enhancing the production of retinoic acid. This, in turn, has an impact on regulatory T cells (Treg) increasing their lineage stability and gut tropism as well as potentiating their suppressive activity. Our results open new avenues for the treatment of IBD patients.
Collapse
|
66
|
Liu EG, Yin X, Swaminathan A, Eisenbarth SC. Antigen-Presenting Cells in Food Tolerance and Allergy. Front Immunol 2021; 11:616020. [PMID: 33488627 PMCID: PMC7821622 DOI: 10.3389/fimmu.2020.616020] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022] Open
Abstract
Food allergy now affects 6%-8% of children in the Western world; despite this, we understand little about why certain people become sensitized to food allergens. The dominant form of food allergy is mediated by food-specific immunoglobulin E (IgE) antibodies, which can cause a variety of symptoms, including life-threatening anaphylaxis. A central step in this immune response to food antigens that differentiates tolerance from allergy is the initial priming of T cells by antigen-presenting cells (APCs), primarily different types of dendritic cells (DCs). DCs, along with monocyte and macrophage populations, dictate oral tolerance versus allergy by shaping the T cell and subsequent B cell antibody response. A growing body of literature has shed light on the conditions under which antigen presentation occurs and how different types of T cell responses are induced by different APCs. We will review APC subsets in the gut and discuss mechanisms of APC-induced oral tolerance versus allergy to food identified using mouse models and patient samples.
Collapse
Affiliation(s)
- Elise G Liu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, United States.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States.,Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, New Haven, CT, United States
| | - Xiangyun Yin
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, United States.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
| | - Anush Swaminathan
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, United States.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States.,Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
67
|
Li C, Dawicki W, Zhang X, Rudulier C, Gordon JR. IL-10- and retinoic acid-induced regulatory dendritic cells are therapeutically equivalent in mouse models of asthma and food allergy. AIMS ALLERGY AND IMMUNOLOGY 2021. [DOI: 10.3934/allergy.2021007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
68
|
Bruellman R, Llorente C. A Perspective Of Intestinal Immune-Microbiome Interactions In Alcohol-Associated Liver Disease. Int J Biol Sci 2021; 17:307-327. [PMID: 33390852 PMCID: PMC7757023 DOI: 10.7150/ijbs.53589] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
Uncovering the intricacies of the gut microbiome and how it interacts with the host immune system has opened up pathways in the search for the treatment of disease conditions. Alcohol-associated liver disease is a major cause of death worldwide. Research has shed light on the breakdown of the protective gut barriers, translocation of gut microbes to the liver and inflammatory immune response to microbes all contributing to alcohol-associated liver disease. This knowledge has opened up avenues for alternative therapies to alleviate alcohol-associated liver disease based on the interaction of the commensal gut microbiome as a key player in the regulation of the immune response. This review describes the relevance of the intestinal immune system, the gut microbiota, and specialized and non-specialized intestinal cells in the regulation of intestinal homeostasis. It also reflects how these components are altered during alcohol-associated liver disease and discusses new approaches for potential future therapies in alcohol-associated liver disease.
Collapse
Affiliation(s)
- Ryan Bruellman
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Cristina Llorente
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
69
|
Rampal R, Wari N, Singh AK, Das U, Bopanna S, Gupta V, Nayak B, Velapandian T, Kedia S, Kumar D, Awasthi A, Ahuja V. Retinoic Acid Is Elevated in the Mucosa of Patients With Active Ulcerative Colitis and Displays a Proinflammatory Role by Augmenting IL-17 and IFNγ Production. Inflamm Bowel Dis 2021; 27:74-83. [PMID: 32469071 DOI: 10.1093/ibd/izaa121] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND All-trans retinoic acid (RA) plays a crucial role in promoting Foxp3+ Treg generation while reciprocally inhibiting Th1/Th17 generation. Our previous research highlighted that in the face of inflammatory conditions, RA plays a contrary role where it aggravates intestinal inflammation by promoting interferon (IFN) γ and interleukin (IL)-17 differentiation in vitro. METHODS In this study we translated our in vitro results into a clinical setting where we estimated mucosal and serum RA levels along with the immunophenotypic profile (IL-17, IFNγ, Foxp3, IL-10) in adaptive (CD4, CD8) and innate-like T cells (mucosal associated invariant T cells and γδ T cells) in patients with ulcerative colitis in remission or with active inflammation. RESULTS This is the first study to estimate RA levels in the human gut and shows that patients with active disease had increased mucosal RA levels as compared with patients in remission (4.0 vs 2.5 ng/mL; P < 0.01) and control patients (3.4 vs 0.8 ng/mL; P < 0.0001). This effect was accompanied by significantly elevated IL-17 and IFNγ in tissue CD4+, CD8+, mucosal associated invariant T+ cells, and γδ + T cells. Moreover, the raised RA levels in patients with active disease showed a positive correlation with proinflammatory cytokines (IL-17, IFNγ) and a negative correlation with IL-10. We also found that RA negatively correlated with IL-9, thereby reinstating our previous finding that RA inhibits Th9 differentiation. CONCLUSIONS These data confirm our previous in vitro results that in the presence of inflammation, RA plays a crucial role in maintaining gut inflammation by upregulating proinflammatory markers.
Collapse
Affiliation(s)
- Ritika Rampal
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| | - Nahidul Wari
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| | - Amit Kumar Singh
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| | - Ujjwalkumar Das
- Department of Ocular Pharmacology, Dr. R. P. Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Sawan Bopanna
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| | - Vipin Gupta
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| | - Baibaswata Nayak
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| | - T Velapandian
- Department of Ocular Pharmacology, Dr. R. P. Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Saurabh Kedia
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| | - Dhiraj Kumar
- Cellular Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Amit Awasthi
- Centre for Human Microbial Ecology, Translational Heath Science and Technology Institute, Haryana, India
| | - Vineet Ahuja
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
70
|
Vitkov L, Minnich B, Knopf J, Schauer C, Hannig M, Herrmann M. NETs Are Double-Edged Swords with the Potential to Aggravate or Resolve Periodontal Inflammation. Cells 2020; 9:E2614. [PMID: 33291407 PMCID: PMC7762037 DOI: 10.3390/cells9122614] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/24/2022] Open
Abstract
Periodontitis is a general term for diseases characterised by inflammatory destruction of tooth-supporting tissues, gradual destruction of the marginal periodontal ligament and resorption of alveolar bone. Early-onset periodontitis is due to disturbed neutrophil extracellular trap (NET) formation and clearance. Indeed, mutations that inactivate the cysteine proteases cathepsin C result in the massive periodontal damage seen in patients with deficient NET formation. In contrast, exaggerated NET formation due to polymorphonuclear neutrophil (PMN) hyper-responsiveness drives the pathology of late-onset periodontitis by damaging and ulcerating the gingival epithelium and retarding epithelial healing. Despite the gingival regeneration, periodontitis progression ends with almost complete loss of the periodontal ligament and subsequent tooth loss. Thus, NETs help to maintain periodontal health, and their dysregulation, either insufficiency or surplus, causes heavy periodontal pathology and edentulism.
Collapse
Affiliation(s)
- Ljubomir Vitkov
- Department of Biosciences, Vascular & Exercise Biology Unit, University of Salzburg, 5020 Salzburg, Austria; (L.V.); (B.M.)
- Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, Saarland University, 66424 Homburg, Germany
| | - Bernd Minnich
- Department of Biosciences, Vascular & Exercise Biology Unit, University of Salzburg, 5020 Salzburg, Austria; (L.V.); (B.M.)
| | - Jasmin Knopf
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (J.K.); (C.S.); (M.H.)
| | - Christine Schauer
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (J.K.); (C.S.); (M.H.)
| | - Matthias Hannig
- Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, Saarland University, 66424 Homburg, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (J.K.); (C.S.); (M.H.)
| |
Collapse
|
71
|
Morante-Palacios O, Fondelli F, Ballestar E, Martínez-Cáceres EM. Tolerogenic Dendritic Cells in Autoimmunity and Inflammatory Diseases. Trends Immunol 2020; 42:59-75. [PMID: 33293219 DOI: 10.1016/j.it.2020.11.001] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/06/2020] [Accepted: 11/08/2020] [Indexed: 12/11/2022]
Abstract
Dendritic cells (DCs), the most efficient antigen-presenting cells, are necessary for the effective activation of naïve T cells. DCs can also acquire tolerogenic functions in vivo and in vitro in response to various stimuli, including interleukin (IL)-10, transforming growth factor (TGF)-β, vitamin D3, corticosteroids, and rapamycin. In this review, we provide a wide perspective on the regulatory mechanisms, including crosstalk with other cell types, downstream signaling pathways, transcription factors, and epigenetics, underlying the acquisition of tolerogenesis by DCs, with a special focus on human studies. Finally, we present clinical assays targeting, or based on, tolerogenic DCs in inflammatory diseases. Our discussion provides a useful resource for better understanding the biology of tolerogenic DCs and their manipulation to improve the immunological fitness of patients with certain inflammatory conditions.
Collapse
Affiliation(s)
- Octavio Morante-Palacios
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain; Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Barcelona, Spain
| | - Federico Fondelli
- Division of Immunology, Germans Trias i Pujol Hospital, LCMN, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Barcelona, Spain; Department of Cell Biology, Physiology, Immunology, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain; Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Barcelona, Spain.
| | - Eva M Martínez-Cáceres
- Division of Immunology, Germans Trias i Pujol Hospital, LCMN, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Barcelona, Spain; Department of Cell Biology, Physiology, Immunology, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain.
| |
Collapse
|
72
|
Nutraceuticals and probiotics in the management of psychiatric and neurological disorders: A focus on microbiota-gut-brain-immune axis. Brain Behav Immun 2020; 90:403-419. [PMID: 32889082 DOI: 10.1016/j.bbi.2020.08.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 08/20/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
|
73
|
Guo HL, Shi FD, Zhou Q, Liu QY, Wang YX, Song Y, Wu ZS, Shi YH, Zhang L, Xu KZ, Song GD. Interleukin-1β Protection Against Experimental Sepsis in Mice. Inflammation 2020; 44:358-370. [PMID: 33044666 PMCID: PMC7548415 DOI: 10.1007/s10753-020-01341-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/21/2020] [Accepted: 09/08/2020] [Indexed: 01/25/2023]
Abstract
The inflammatory response involving interleukin-1β (IL-1β) has been thought to play an important role in the development of late-phase sepsis. However, in this study, we wanted to explore the possibility of using IL-1β to improve the prognosis of sepsis by triggering local differentiation of bone marrow cells (BMCs) into regulatory dendritic cells (DCs) in vivo, thereby reversing the immune paralysis in late-phase sepsis. Sepsis mouse models were induced by cecal ligation and puncture (CLP) and lethal Escherichia coli O18 infection. Mice were injected intraperitoneally with IL-1β after CLP and after the lethal infection. Septic BMCs and liver immune cells were isolated at 0, 3, 6, 9, and 14 days post-CLP. BMCs and liver cells isolated from septic mice treated with IL-1β were adoptively transferred into CLP mice. GFP+-C57BL/6 parabiosis models were established. Serum IL-1β levels were determined by enzyme-linked immunosorbent assay (ELISA) kit, and the number, ratio, and phenotype of immune cells were observed by flow cytometry. IL-1β treatment improved the survival of sepsis and increased the numbers of BMCs and liver immune cells in septic mice. Moreover, IL-1β stimulation increased the number and the percentage of CD11c−CD45RBhigh DCs in septic BM and liver. Adoptive transfer of septic BMCs, liver immune cells, and CD11c−CD45RBhigh DCs treated with IL-1β into CLP mice attenuated sepsis. IL-1β triggered the redistribution of CD11c−CD45RBhigh DCs as well as BMCs in parabiosis models. IL-1β protects against sepsis by stimulating local proliferation and differentiation of BMCs into CD11c−CD45RBhigh DCs at immune organs and non-immune organs.
Collapse
Affiliation(s)
- Hai-Lei Guo
- Department of Burns, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China.,Department of Burns, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Fu-Dong Shi
- Department of Orthopedics, Tangshan People's Hospital, Tangshan, Hebei, People's Republic of China
| | - Qi Zhou
- North China University of Science and Technology Graduate School, Tangshan, Hebei, People's Republic of China
| | - Qing-Yang Liu
- Department of Translational Medicine and Nephrology, Emergency General Hospital, Beijing, 100028, People's Republic of China.
| | - Yue-Xin Wang
- First Department of Orthopedics, Tangshan Worker's Hospital Affiliated to Hebei Medical University, Tangshan, Hebei, People's Republic of China
| | - Yang Song
- Department of Translational Medicine and Nephrology, Emergency General Hospital, Beijing, 100028, People's Republic of China
| | - Zong-Sheng Wu
- Department of Emergency, Zhongda Hospital Affiliated to Southeast University, Nanjing, People's Republic of China
| | - Yu-Hao Shi
- North China University of Science and Technology Graduate School, Tangshan, Hebei, People's Republic of China
| | - Liu Zhang
- Department of Translational Medicine and Nephrology, Emergency General Hospital, Beijing, 100028, People's Republic of China
| | - Kai-Zhi Xu
- Department of Anesthesiology, Tangshan Worker's Hospital Affiliated to Hebei Medical University, Tangshan, Hebei, People's Republic of China
| | - Guo-Dong Song
- Department of Burns, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China. .,Department of Burns, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China.
| |
Collapse
|
74
|
Satomi S, Khanum S, Miller P, Suzuki S, Suganuma H, Heiser A, Gupta SK. Short Communication: Oral Administration of Heat-killed Lactobacillus brevis KB290 in Combination with Retinoic Acid Provides Protection against Influenza Virus Infection in Mice. Nutrients 2020; 12:nu12102925. [PMID: 32987850 PMCID: PMC7600661 DOI: 10.3390/nu12102925] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
Influenza virus type A (IAV) is a seasonal acute respiratory disease virus with severe symptoms, and an effective preventive measure is required. Despite many reports describing the potentially protective effects of lactic acid bacteria, few studies have investigated the effects of nutritional supplement combinations. This study reports the effect of the combined intake of heat-killed Lactobacillus brevis KB290 (KB290) and vitamin A (VA) on mice challenged with a sublethal dose of IAV. For 2 weeks, five groups of mice were fed either placebo, KB290, VA, or a combination of KB290 and VA (KB290+VA). After subsequent IAV challenge, bodyweight and general health were monitored for up to 2 weeks. Viral titres were determined in the lungs of animal subgroups euthanised at days 3, 7, and 14 after IAV challenge. A significant loss was observed in the bodyweights of IAV-infected animals from day 1 post-IAV challenge, whereas the mice fed KB290+VA did not lose any weight after IAV infection, indicating successful protection from the infection. Additionally, mice in the KB290+VA group showed the highest reduction in lung viral titres. In conclusion, the combination of KB290 and VA could be a useful food supplement relevant for protection against seasonal influenza virus infection in humans.
Collapse
Affiliation(s)
- Shohei Satomi
- Department of Nature & Wellness Research, Innovation Division, KAGOME CO., LTD., 17 Nishitomiyama, Nasushiobara, Tochigi 329-2762, Japan; (S.S.); (H.S.)
- Correspondence: (S.S.); (S.K.G.); Tel.: +81-80-8132-3813 (S.S.); +64-06351-8697 (S.K.G.)
| | - Sofia Khanum
- AgResearch Ltd., Hopkirk Research Institute, Grasslands Research Centre, Private Bag 11008, Palmerston North 4442, New Zealand; (S.K.); (P.M.); (A.H.)
| | - Poppy Miller
- AgResearch Ltd., Hopkirk Research Institute, Grasslands Research Centre, Private Bag 11008, Palmerston North 4442, New Zealand; (S.K.); (P.M.); (A.H.)
| | - Shigenori Suzuki
- Department of Nature & Wellness Research, Innovation Division, KAGOME CO., LTD., 17 Nishitomiyama, Nasushiobara, Tochigi 329-2762, Japan; (S.S.); (H.S.)
| | - Hiroyuki Suganuma
- Department of Nature & Wellness Research, Innovation Division, KAGOME CO., LTD., 17 Nishitomiyama, Nasushiobara, Tochigi 329-2762, Japan; (S.S.); (H.S.)
| | - Axel Heiser
- AgResearch Ltd., Hopkirk Research Institute, Grasslands Research Centre, Private Bag 11008, Palmerston North 4442, New Zealand; (S.K.); (P.M.); (A.H.)
| | - Sandeep K Gupta
- AgResearch Ltd., Hopkirk Research Institute, Grasslands Research Centre, Private Bag 11008, Palmerston North 4442, New Zealand; (S.K.); (P.M.); (A.H.)
- Correspondence: (S.S.); (S.K.G.); Tel.: +81-80-8132-3813 (S.S.); +64-06351-8697 (S.K.G.)
| |
Collapse
|
75
|
Iberg CA, Hawiger D. Natural and Induced Tolerogenic Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2020; 204:733-744. [PMID: 32015076 DOI: 10.4049/jimmunol.1901121] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/04/2019] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DCs) are highly susceptible to extrinsic signals that modify the functions of these crucial APCs. Maturation of DCs induced by diverse proinflammatory conditions promotes immune responses, but certain signals also induce tolerogenic functions in DCs. These "induced tolerogenic DCs" help to moderate immune responses such as those to commensals present at specific anatomical locations. However, also under steady-state conditions, some DCs are characterized by inherent tolerogenic properties. The immunomodulatory mechanisms constitutively present in such "natural tolerogenic DCs" help to promote tolerance to peripheral Ags. By extending tolerance initially established in the thymus, these functions of DCs help to regulate autoimmune and other immune responses. In this review we will discuss the mechanisms and functions of natural and induced tolerogenic DCs and offer further insight into how their possible manipulations may ultimately lead to more precise treatments for various immune-mediated conditions and diseases.
Collapse
Affiliation(s)
- Courtney A Iberg
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Daniel Hawiger
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104
| |
Collapse
|
76
|
Olwenyi OA, Acharya A, Routhu NK, Pierzchalski K, Jones JW, Kane MA, Sidell N, Mohan M, Byrareddy SN. Retinoic Acid Improves the Recovery of Replication-Competent Virus from Latent SIV Infected Cells. Cells 2020; 9:E2076. [PMID: 32932813 PMCID: PMC7565696 DOI: 10.3390/cells9092076] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/05/2020] [Accepted: 09/09/2020] [Indexed: 12/31/2022] Open
Abstract
The accurate estimation and eradication of Human Immunodeficiency Virus (HIV) viral reservoirs is limited by the incomplete reactivation of cells harboring the latent replication-competent virus. We investigated whether the in vitro and in vivo addition of retinoic acid (RA) enhances virus replication and improves the detection of latent virus. Peripheral blood mononuclear cells (PBMCs) from naive and anti-retroviral therapy (ART)-treated SIV-infected rhesus macaques (RMs) were cultured in vitro with anti-CD3/CD28 + IL-2 in the presence/absence of RA. Viral RNA and p27 levels were quantified using RT-qPCR and ELISA, respectively. Viral reservoirs were estimated using the Tat/Rev-Induced Limited Dilution Assay (TILDA) and Quantitative Viral Outgrowth Assay (QVOA). In vitro and in vivo measures revealed that there was also an increase in viral replication in RA-treated versus without RA conditions. In parallel, the addition of RA to either CD3/CD28 or phorbol myristate acetate (PMA)/ionomycin during QVOA and TILDA, respectively, was shown to augment reactivation of the replication-competent viral reservoir in anti-retroviral therapy (ART)-suppressed RMs as shown by a greater than 2.3-fold increase for QVOA and 1 to 2-fold increments for multi-spliced RNA per million CD4+ T cells. The use of RA can be a useful approach to enhance the efficiency of current protocols used for in vitro and potentially in vivo estimates of CD4+ T cell latent reservoirs. In addition, flow cytometry analysis revealed that RA improved estimates of various viral reservoir assays by eliciting broad CD4 T-cell activation as demonstrated by elevated CD25 and CD38 but reduced CD69 and PD-1 expressing cells.
Collapse
Affiliation(s)
- Omalla A. Olwenyi
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA; (O.A.O.); (A.A.); (N.K.R.)
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Arpan Acharya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA; (O.A.O.); (A.A.); (N.K.R.)
| | - Nanda Kishore Routhu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA; (O.A.O.); (A.A.); (N.K.R.)
| | - Keely Pierzchalski
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA; (K.P.); (J.W.J.); (M.A.K.)
| | - Jace W. Jones
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA; (K.P.); (J.W.J.); (M.A.K.)
| | - Maureen A. Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA; (K.P.); (J.W.J.); (M.A.K.)
| | - Neil Sidell
- Department of Obstetrics and Gynecology, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Mahesh Mohan
- Texas Biomedical Research Institute, Southwest National Primate Research Institute, San Antonio, TX 78227, USA;
| | - Siddappa N. Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA; (O.A.O.); (A.A.); (N.K.R.)
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| |
Collapse
|
77
|
Segal JP, Mullish BH, Quraishi MN, Iqbal T, Marchesi JR, Sokol H. Mechanisms underpinning the efficacy of faecal microbiota transplantation in treating gastrointestinal disease. Therap Adv Gastroenterol 2020; 13:1756284820946904. [PMID: 32952613 PMCID: PMC7475788 DOI: 10.1177/1756284820946904] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 07/13/2020] [Indexed: 02/04/2023] Open
Abstract
Faecal microbiota transplantation (FMT) is currently a recommended therapy for recurrent/refractory Clostridioides difficile infection (CDI). The success of FMT for CDI has led to interest in its therapeutic potential in many other disorders. The mechanisms that underpin the efficacy of FMT are not fully understood. Importantly, FMT remains a crucial treatment in managing CDI and understanding the mechanisms that underpin its success will be critical to improve its clinical efficacy, safety and usability. Furthermore, a deeper understanding of this may allow us to expose FMT's full potential as a therapeutic tool for other disease states. This review will explore the current understanding of the mechanisms underlying the efficacy of FMT across a variety of diseases.
Collapse
Affiliation(s)
- Jonathan P. Segal
- Departments of Gastroenterology and Hepatology, St Mary’s Hospital, Imperial College Healthcare NHS Trust, South Wharf Rd, London W2 1NY, UK
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, UK
| | - Benjamin H. Mullish
- Departments of Gastroenterology and Hepatology, St Mary’s Hospital, Imperial College Healthcare NHS Trust, London, UK
- Department of Metabolism, Digestion and Reproduction, Division of Digestive Diseases, Imperial College London, UK
| | - Mohammed N. Quraishi
- University of Birmingham Microbiome Treatment Centre, University of Birmingham, Birmingham, UK
- Department of Gastroenterology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Tariq Iqbal
- University of Birmingham Microbiome Treatment Centre, University of Birmingham, Birmingham, UK
- Department of Gastroenterology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Julian R. Marchesi
- Department of Metabolism, Digestion and Reproduction, Division of Digestive Diseases, Imperial College London, UK
- School of Biosciences, Cardiff University, Cardiff, UK
| | - Harry Sokol
- Gastroenterology Department, INSERM, Centre de Recherche Saint Antoine, CRSA, AP-HP, Sorbonne Université, Saint Antoine Hospital, Paris, France
- INRA, UMR1319 Micalis and AgroParisTech, Jouy en Josas, France Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| |
Collapse
|
78
|
Endres K. Retinoic Acid and the Gut Microbiota in Alzheimer's Disease: Fighting Back-to-Back? Curr Alzheimer Res 2020; 16:405-417. [PMID: 30907321 DOI: 10.2174/1567205016666190321163705] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/15/2019] [Accepted: 03/19/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND There is growing evidence that the gut microbiota may play an important role in neurodegenerative diseases such as Alzheimer's disease. However, how these commensals influence disease risk and progression still has to be deciphered. OBJECTIVE The objective of this review was to summarize current knowledge on the interplay between gut microbiota and retinoic acid. The latter one represents one of the important micronutrients, which have been correlated to Alzheimer's disease and are used in initial therapeutic intervention studies. METHODS A selective overview of the literature is given with the focus on the function of retinoic acid in the healthy and diseased brain, its metabolism in the gut, and the potential influence that the bioactive ligand may have on microbiota, gut physiology and, Alzheimer's disease. RESULTS Retinoic acid can influence neuronal functionality by means of plasticity but also by neurogenesis and modulating proteostasis. Impaired retinoid-signaling, therefore, might contribute to the development of diseases in the brain. Despite its rather direct impact, retinoic acid also influences other organ systems such as gut by regulating the residing immune cells but also factors such as permeability or commensal microbiota. These in turn can also interfere with retinoid-metabolism and via the gutbrain- axis furthermore with Alzheimer's disease pathology within the brain. CONCLUSION Potentially, it is yet too early to conclude from the few reports on changed microbiota in Alzheimer's disease to a dysfunctional role in retinoid-signaling. However, there are several routes how microbial commensals might affect and might be affected by vitamin A and its derivatives.
Collapse
Affiliation(s)
- Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
79
|
Stacchiotti V, Rezzi S, Eggersdorfer M, Galli F. Metabolic and functional interplay between gut microbiota and fat-soluble vitamins. Crit Rev Food Sci Nutr 2020; 61:3211-3232. [PMID: 32715724 DOI: 10.1080/10408398.2020.1793728] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gut microbiota is a complex ecosystem seen as an extension of human genome. It represents a major metabolic interface of interaction with food components and xenobiotics in the gastrointestinal (GI) environment. In this context, the advent of modern bacterial genome sequencing technology has enabled the identification of dietary nutrients as key determinants of gut microbial ecosystem able to modulate the host-microbiome symbiotic relationship and its effects on human health. This article provides a literature review on functional and molecular interactions between a specific group of lipids and essential nutrients, e.g., fat-soluble vitamins (FSVs), and the gut microbiota. A two-way relationship appears to emerge from the available literature with important effects on human metabolism, nutrition, GI physiology and immune function. First, FSV directly or indirectly modify the microbial composition involving for example immune system-mediated and/or metabolic mechanisms of bacterial growth or inhibition. Second, the gut microbiota influences at different levels the synthesis, metabolism and transport of FSV including their bioactive metabolites that are either introduced with the diet or released in the gut via entero-hepatic circulation. A better understanding of these interactions, and of their impact on intestinal and metabolic homeostasis, will be pivotal to design new and more efficient strategies of disease prevention and therapy, and personalized nutrition.
Collapse
Affiliation(s)
- Valentina Stacchiotti
- Micronutrient Vitamins and Lipidomics Lab, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Serge Rezzi
- Swiss Vitamin Institute, Epalinges, Switzerland
| | - Manfred Eggersdorfer
- Department of Internal Medicine, University Medical Center Groningen, Groningen, the Netherlands
| | - Francesco Galli
- Micronutrient Vitamins and Lipidomics Lab, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| |
Collapse
|
80
|
Yuzuriha K, Yoshida A, Li S, Kishimura A, Mori T, Katayama Y. Synthesis of peptide conjugates with vitamins for induction of antigen-specific immunotolerance. J Pept Sci 2020; 26:e3275. [PMID: 32671962 DOI: 10.1002/psc.3275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/06/2020] [Accepted: 06/20/2020] [Indexed: 11/07/2022]
Abstract
In this report, we designed conjugates of an antigen peptide with the immunosuppressive vitamins all-trans retinoic acid (ATRA) and vitamin D3 for efficient induction of antigen-specific immunotolerance. We established a synthetic scheme for the preparation of the peptide-vitamin conjugates, which the chemically unstable vitamins tolerated. Among the obtained conjugates, the ATRA conjugate successfully suppressed inflammatory effects in macrophages and dendritic cells and induced antigen presentation in dendritic cells. This synthetic method of conjugate is conceivably applicable to other antigen peptides for induction of antigen-specific immunotolerance.
Collapse
Affiliation(s)
- Kazuki Yuzuriha
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| | - Ayaka Yoshida
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| | - Shunyi Li
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| | - Akihiro Kishimura
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka, 819-0395, Japan.,Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan.,Center for Future Chemistry, Kyushu University, Fukuoka, 819-0395, Japan.,International Research Center for Molecular Systems, Kyushu University, Fukuoka, 819-0395, Japan
| | - Takeshi Mori
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka, 819-0395, Japan.,Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan.,Center for Future Chemistry, Kyushu University, Fukuoka, 819-0395, Japan
| | - Yoshiki Katayama
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka, 819-0395, Japan.,Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan.,Center for Future Chemistry, Kyushu University, Fukuoka, 819-0395, Japan.,International Research Center for Molecular Systems, Kyushu University, Fukuoka, 819-0395, Japan.,Centre for Advanced Medicine Innovation, Kyushu University, Fukuoka, 812-8582, Japan.,Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan City, ROC, 32023, Taiwan
| |
Collapse
|
81
|
Puccetti M, Xiroudaki S, Ricci M, Giovagnoli S. Postbiotic-Enabled Targeting of the Host-Microbiota-Pathogen Interface: Hints of Antibiotic Decline? Pharmaceutics 2020; 12:E624. [PMID: 32635461 PMCID: PMC7408102 DOI: 10.3390/pharmaceutics12070624] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/24/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023] Open
Abstract
Mismanagement of bacterial infection therapies has undermined the reliability and efficacy of antibiotic treatments, producing a profound crisis of the antibiotic drug market. It is by now clear that tackling deadly infections demands novel strategies not only based on the mere toxicity of anti-infective compounds. Host-directed therapies have been the first example as novel treatments with alternate success. Nevertheless, recent advances in the human microbiome research have provided evidence that compounds produced by the microbial metabolism, namely postbiotics, can have significant impact on human health. Such compounds target the host-microbe-pathogen interface rescuing biotic and immune unbalances as well as inflammation, thus providing novel therapeutic opportunities. This work discusses critically, through literature review and personal contributions, these novel nonantibiotic treatment strategies for infectious disease management and resistance prevention, which could represent a paradigm change rocking the foundation of current antibiotic therapy tenets.
Collapse
Affiliation(s)
| | | | | | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, via del Liceo 1, University of Perugia, 06123 Perugia, Italy; (M.P.); (S.X.); (M.R.)
| |
Collapse
|
82
|
Lohrmann F, Forde AJ, Merck P, Henneke P. Control of myeloid cell density in barrier tissues. FEBS J 2020; 288:405-426. [PMID: 32502309 DOI: 10.1111/febs.15436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/21/2020] [Accepted: 06/01/2020] [Indexed: 12/19/2022]
Abstract
The interface between the mammalian host and its environment is formed by barrier tissues, for example, of the skin, and the respiratory and the intestinal tracts. On the one hand, barrier tissues are colonized by site-adapted microbial communities, and on the other hand, they contain specific myeloid cell networks comprising macrophages, dendritic cells, and granulocytes. These immune cells are tightly regulated in function and cell number, indicating important roles in maintaining tissue homeostasis and immune balance in the presence of commensal microorganisms. The regulation of myeloid cell density and activation involves cell-autonomous 'single-loop circuits' including autocrine mechanisms. However, an array of microenvironmental factors originating from nonimmune cells and the microbiota, as well as the microanatomical structure, impose additional layers of regulation onto resident myeloid cells. This review discusses models integrating these factors into cell-specific programs to instruct differentiation and proliferation best suited for the maintenance and renewal of immune homeostasis in the tissue-specific environment.
Collapse
Affiliation(s)
- Florens Lohrmann
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Medical Center - University of Freiburg, Germany.,Institute for Immunodeficiency (IFI), Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Germany.,Spemann Graduate School for Biology and Medicine, University of Freiburg, Germany.,IMM-PACT Clinician Scientist Program, Faculty of Medicine, University of Freiburg, Germany
| | - Aaron J Forde
- Institute for Immunodeficiency (IFI), Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Germany.,Faculty of Biology, university of Freiburg, Germany
| | - Philipp Merck
- Institute for Immunodeficiency (IFI), Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Germany
| | - Philipp Henneke
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Medical Center - University of Freiburg, Germany.,Institute for Immunodeficiency (IFI), Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Germany
| |
Collapse
|
83
|
Ricciuto A, Sherman PM, Laxer RM. Gut microbiota in chronic inflammatory disorders: A focus on pediatric inflammatory bowel diseases and juvenile idiopathic arthritis. Clin Immunol 2020; 215:108415. [DOI: 10.1016/j.clim.2020.108415] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022]
|
84
|
Hantisteanu S, Dicken Y, Negreanu V, Goldenberg D, Brenner O, Leshkowitz D, Lotem J, Levanon D, Groner Y. Runx3 prevents spontaneous colitis by directing the differentiation of anti-inflammatory mononuclear phagocytes. PLoS One 2020; 15:e0233044. [PMID: 32453801 PMCID: PMC7250423 DOI: 10.1371/journal.pone.0233044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/27/2020] [Indexed: 12/23/2022] Open
Abstract
Mice deficient in the transcription factor Runx3 develop a multitude of immune system defects, including early onset colitis. This paper demonstrates that Runx3 is expressed in colonic mononuclear phagocytes (MNP), including resident macrophages (RM) and dendritic cell subsets (cDC2). Runx3 deletion in MNP causes early onset colitis due to their impaired maturation. Mechanistically, the resulting MNP subset imbalance leads to up-regulation of pro-inflammatory genes as occurs in IL10R-deficient RM. In addition, RM and cDC2 display a marked decrease in expression of anti-inflammatory/TGF β-regulated genes and β-catenin signaling associated genes, respectively. MNP transcriptome and ChIP-seq data analysis suggest that a significant fraction of genes affected by Runx3 loss are direct Runx3 targets. Collectively, Runx3 imposes intestinal immune tolerance by regulating maturation of colonic anti-inflammatory MNP, befitting the identification of RUNX3 as a genome-wide associated risk gene for various immune-related diseases in humans, including gastrointestinal tract diseases such as Crohn’s disease and celiac.
Collapse
Affiliation(s)
- Shay Hantisteanu
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Yosef Dicken
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Varda Negreanu
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Dalia Goldenberg
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Ori Brenner
- Veterinary Resources, The Weizmann Institute of Science, Rehovot, Israel
| | - Dena Leshkowitz
- Bioinformatics Unit, The Weizmann Institute of Science, Rehovot, Israel
| | - Joseph Lotem
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Ditsa Levanon
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Yoram Groner
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| |
Collapse
|
85
|
Nance CL, Deniskin R, Diaz VC, Paul M, Anvari S, Anagnostou A. The Role of the Microbiome in Food Allergy: A Review. CHILDREN-BASEL 2020; 7:children7060050. [PMID: 32466620 PMCID: PMC7346163 DOI: 10.3390/children7060050] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/14/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023]
Abstract
Food allergies are common and estimated to affect 8% of children and 11% of adults in the United States. They pose a significant burden—physical, economic and social—to those affected. There is currently no available cure for food allergies. Emerging evidence suggests that the microbiome contributes to the development and manifestations of atopic disease. According to the hygiene hypothesis, children growing up with older siblings have a lower incidence of allergic disease compared with children from smaller families, due to their early exposure to microbes in the home. Research has also demonstrated that certain environmental exposures, such as a farming environment, during early life are associated with a diverse bacterial experience and reduced risk of allergic sensitization. Dysregulation in the homeostatic interaction between the host and the microbiome or gut dysbiosis appears to precede the development of food allergy, and the timing of such dysbiosis is critical. The microbiome affects food tolerance via the secretion of microbial metabolites (e.g., short chain fatty acids) and the expression of microbial cellular components. Understanding the biology of the microbiome and how it interacts with the host to maintain gut homeostasis is helpful in developing smarter therapeutic approaches. There are ongoing trials evaluating the benefits of probiotics and prebiotics, for the prevention and treatment of atopic diseases to correct the dysbiosis. However, the routine use of probiotics as an intervention for preventing allergic disease is not currently recommended. A new approach in microbial intervention is to attempt a more general modification of the gut microbiome, such as with fecal microbiota transplantation. Developing targeted bacterial therapies for food allergy may be promising for both the treatment and prevention of food allergy. Similarly, fecal microbiota transplantation is being explored as a potentially beneficial interventional approach. Overall, targeted bacterial therapies for food allergy may be promising for both the treatment and prevention of food allergy.
Collapse
Affiliation(s)
- Christina L. Nance
- Baylor College of Medicine, Section of Pediatric Immunology, Allergy and Retrovirology, Houston, TX 77030 USA; (C.L.N.); (R.D.); (V.C.D.); (M.P.); (S.A.)
- Texas Children’s Hospital, Department of Pediatrics, Section of Immunology, Allergy and Retrovirology, Houston, TX 77030, USA
| | - Roman Deniskin
- Baylor College of Medicine, Section of Pediatric Immunology, Allergy and Retrovirology, Houston, TX 77030 USA; (C.L.N.); (R.D.); (V.C.D.); (M.P.); (S.A.)
- Texas Children’s Hospital, Department of Pediatrics, Section of Immunology, Allergy and Retrovirology, Houston, TX 77030, USA
| | - Veronica C. Diaz
- Baylor College of Medicine, Section of Pediatric Immunology, Allergy and Retrovirology, Houston, TX 77030 USA; (C.L.N.); (R.D.); (V.C.D.); (M.P.); (S.A.)
- Texas Children’s Hospital, Department of Pediatrics, Section of Immunology, Allergy and Retrovirology, Houston, TX 77030, USA
| | - Misu Paul
- Baylor College of Medicine, Section of Pediatric Immunology, Allergy and Retrovirology, Houston, TX 77030 USA; (C.L.N.); (R.D.); (V.C.D.); (M.P.); (S.A.)
- Texas Children’s Hospital, Department of Pediatrics, Section of Immunology, Allergy and Retrovirology, Houston, TX 77030, USA
| | - Sara Anvari
- Baylor College of Medicine, Section of Pediatric Immunology, Allergy and Retrovirology, Houston, TX 77030 USA; (C.L.N.); (R.D.); (V.C.D.); (M.P.); (S.A.)
- Texas Children’s Hospital, Department of Pediatrics, Section of Immunology, Allergy and Retrovirology, Houston, TX 77030, USA
| | - Aikaterini Anagnostou
- Baylor College of Medicine, Section of Pediatric Immunology, Allergy and Retrovirology, Houston, TX 77030 USA; (C.L.N.); (R.D.); (V.C.D.); (M.P.); (S.A.)
- Texas Children’s Hospital, Department of Pediatrics, Section of Immunology, Allergy and Retrovirology, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
86
|
Yang CD, Cheng ML, Liu W, Zeng DH. Association of serum retinoic acid with depression in patients with acute ischemic stroke. Aging (Albany NY) 2020; 12:2647-2658. [PMID: 32040942 PMCID: PMC7041768 DOI: 10.18632/aging.102767] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/12/2020] [Indexed: 12/17/2022]
Abstract
Retinoic acid (RA), produced by the metabolism of vitamin A, makes effects on depression and stroke. This study was aimed to evaluate the relationship between RA levels in serum and post-stroke depression (PSD). A single-center (Chengdu, China) prospective cohort study was conducted on patients with acute ischemic stroke. The RA serum level was measured at admission. The PSD was assessed in the 3-month follow-up. The RA-PSD relationship was evaluated with conditional logistic regression. In total, 239 ischemic stroke cases and 100 healthy controls were included. The median RA serum level in patients with ischemic stroke was 2.45 ng/ml (interquartile range [IQR], 0.72-4.33), lower(P<0.001) than 3.89 ng/ml of those in control cases ([IQR]: 2.62-5.39). The crude and adjusted odds ratios [OR] (and 95% confidence intervals [CI]) of PSD associated with an IQR increase for RA were 0.54 (0.44, 0.67) and 0.66 (0.52, 0.79), respectively. Higher ORs of PSD associated with reduced RA levels (
Collapse
Affiliation(s)
- Cai-Di Yang
- Department of Neurology, Eastern Hospital, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610101, China
| | - Ming-Li Cheng
- Department of Neurology, People's Hospital of Jianyang, Jianyang 641400, China
| | - Wen Liu
- The Clinical Laboratory Department, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Ding-Hua Zeng
- Department of Neurology, Eastern Hospital, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610101, China
| |
Collapse
|
87
|
Švajger U, Rožman PJ. Recent discoveries in dendritic cell tolerance-inducing pharmacological molecules. Int Immunopharmacol 2020; 81:106275. [PMID: 32044665 DOI: 10.1016/j.intimp.2020.106275] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/31/2020] [Accepted: 01/31/2020] [Indexed: 01/04/2023]
Abstract
Dendritic cells (DCs) represent one of the most important biological tools for cellular immunotherapy purposes. There are an increasing number of phase I and II studies, where regulatory or tolerogenic DCs (TolDCs) are utilized as negative vaccines, with the aim of inducing tolerogenic outcomes in patients with various autoimmune or chronic-inflammatory diseases, as well as in transplant settings. The induction of tolerogenic properties in DCs can be achieved by altering their activation state toward expression of immunosuppressive elements and/or by achieving resistance to maturation, which leads to insufficient co-stimulatory signal delivery and inability to efficiently present antigens. In the past, one of the most efficient ways to induce DC tolerance has been the application of selected pharmacological agents which actively induce a tolerogenic transcription program or inhibit major pro-inflammatory transcription factors such as Nf-κB. Important examples include immunosuppressants such as different corticosteroids, vitamin D3, rapamycin and others. The quality of TolDCs induced by different approaches is becoming a vital issue and recent evidence suggests substantial heterogeneity between variously-generated TolDCs as evidenced by their transcriptomic profile and function. The possibility of various "flavors" of TolDCs encourages future research in discovery of Tol-DC inducing agents to enrich various ways of DC manipulation. This would enable a broader range of tools to manipulate DC toward specific characteristics desirable in different disease settings. In recent years, several novel small molecules have been identified with the capacity to promote DC tolerogenic characteristics. In this review, we will present and discuss these novel findings and also highlight novel understandings of tolerogenic mechanisms by which DC tolerogenicity is induced by already established agents.
Collapse
Affiliation(s)
- Urban Švajger
- Blood Transfusion Center of Slovenia, Šlajmerjeva 6, 1000 Ljubljana, Slovenia.
| | - Primož J Rožman
- Blood Transfusion Center of Slovenia, Šlajmerjeva 6, 1000 Ljubljana, Slovenia
| |
Collapse
|
88
|
Quraishi MN, Shaheen W, Oo YH, Iqbal TH. Immunological mechanisms underpinning faecal microbiota transplantation for the treatment of inflammatory bowel disease. Clin Exp Immunol 2019; 199:24-38. [PMID: 31777058 DOI: 10.1111/cei.13397] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2019] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic gastrointestinal disease that results from a dysregulated immune response against specific environmental triggers in a genetically predisposed individual. Increasing evidence has indicated a causal role for changes in gut microbiota (dysbiosis) contributing to this immune-mediated intestinal inflammation. These mechanisms involve dysregulation of multiple facets of the host immune pathways that are potentially reversible. Faecal microbiota transplantation (FMT) is the transfer of processed stool from a healthy donor into an individual with an illness. FMT has shown promising results in both animal model experiments and clinical studies in IBD in the resolution of intestinal inflammation. The underlying mechanisms, however, are unclear. Insights from these studies have shown interactions between modulation of dysbiosis via changes in abundances of specific members of the gut microbial community and changes in host immunological pathways. Unravelling these causal relationships has promising potential for a translational therapy role to develop targeted microbial therapies and understand the mechanisms that underpin IBD aetiopathogenesis. In this review, we discuss current evidence for the contribution of gut microbiota in the disruption of intestinal immune homeostasis and immunoregulatory mechanisms that are associated with the resolution of inflammation through FMT in IBD.
Collapse
Affiliation(s)
- M N Quraishi
- Centre for Liver and Gastroenterology Research, NIHR Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, UK.,Department of Gastroenterology, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, UK.,University of Birmingham Microbiome Treatment Centre, University of Birmingham, Birmingham, UK
| | - W Shaheen
- Centre for Liver and Gastroenterology Research, NIHR Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, UK.,University of Birmingham Microbiome Treatment Centre, University of Birmingham, Birmingham, UK
| | - Y H Oo
- Centre for Liver and Gastroenterology Research, NIHR Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, UK.,Department of Gastroenterology, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, UK.,Liver Transplant and Hepatobiliary Unit, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, UK
| | - T H Iqbal
- Centre for Liver and Gastroenterology Research, NIHR Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, UK.,Department of Gastroenterology, Queen Elizabeth Hospital, University Hospitals Birmingham, Birmingham, UK.,University of Birmingham Microbiome Treatment Centre, University of Birmingham, Birmingham, UK
| |
Collapse
|
89
|
Vitamin A deficiency impairs the immune response to intranasal vaccination and RSV infection in neonatal calves. Sci Rep 2019; 9:15157. [PMID: 31641172 PMCID: PMC6805856 DOI: 10.1038/s41598-019-51684-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 10/03/2019] [Indexed: 02/08/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection is a leading cause of severe acute lower respiratory tract infection in infants and children worldwide. Vitamin A deficiency (VAD) is one of the most prevalent nutrition-related health problems in the world and is a significant risk factor in the development of severe respiratory infections in infants and young children. Bovine RSV (BRSV) is a primary cause of lower respiratory tract disease in young cattle. The calf model of BRSV infection is useful to understand the immune response to human RSV infection. We have previously developed an amphiphilic polyanhydride nanoparticle (NP)-based vaccine (i.e., nanovaccine) encapsulating the fusion and attachment proteins from BRSV (BRSV-NP). Calves receiving a single, intranasal dose of the BRSV-NP vaccine are partially protected from BRSV challenge. Here, we evaluated the impact of VAD on the immune response to the BRSV-NP vaccine and subsequent challenge with BRSV. Our results show that VAD calves are unable to respond to the mucosal BRSV-NP vaccine, are afforded no protection from BRSV challenge and have significant abnormalities in the inflammatory response in the infected lung. We further show that acute BRSV infection negatively impacts serum and liver retinol, rendering even well-nourished individuals susceptible to VAD. Our results support the use of the calf model for elucidating the impact of nutritional status on mucosal immunity and respiratory viral infection in infants and underline the importance of VA in regulating immunity in the respiratory mucosa.
Collapse
|
90
|
Pejoski D, Ballester M, Auderset F, Vono M, Christensen D, Andersen P, Lambert PH, Siegrist CA. Site-Specific DC Surface Signatures Influence CD4 + T Cell Co-stimulation and Lung-Homing. Front Immunol 2019; 10:1650. [PMID: 31396211 PMCID: PMC6668556 DOI: 10.3389/fimmu.2019.01650] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/03/2019] [Indexed: 11/29/2022] Open
Abstract
Dendritic cells (DCs) that drain the gut and skin are known to favor the establishment of T cell populations that home to the original site of DC-antigen (Ag) encounter by providing soluble “imprinting” signals to T cells in the lymph node (LN). To study the induction of lung T cell-trafficking, we used a protein-adjuvant murine intranasal and intramuscular immunization model to compare in vivo-activated Ag+ DCs in the lung and muscle-draining LNs. Higher frequencies of Ag+ CD11b+ DCs were observed in lung-draining mediastinal LNs (MedLN) compared to muscle-draining inguinal LNs (ILN). Ag+ CD11b+ MedLN DCs were qualitatively superior at priming CD4+ T cells, which then expressed CD49a and CXCR3, and preferentially trafficked into the lung parenchyma. CD11b+ DCs from the MedLN expressed higher levels of surface podoplanin, Trem4, GL7, and the known co-stimulatory molecules CD80, CD86, and CD24. Blockade of specific MedLN DC molecules or the use of sorted DC and T cell co-cultures demonstrated that DC surface phenotype influences the ability to prime T cells that then home to the lung. Thus, the density of dLN Ag+ DCs, and DC surface molecule signatures are factors that can influence the output and differentiation of lung-homing CD4+ T cells.
Collapse
Affiliation(s)
- David Pejoski
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,World Health Organization Collaborating Center for Vaccine Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Marie Ballester
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,World Health Organization Collaborating Center for Vaccine Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Floriane Auderset
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,World Health Organization Collaborating Center for Vaccine Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Maria Vono
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,World Health Organization Collaborating Center for Vaccine Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Dennis Christensen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Andersen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Paul-Henri Lambert
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,World Health Organization Collaborating Center for Vaccine Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Claire-Anne Siegrist
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,World Health Organization Collaborating Center for Vaccine Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
91
|
Daniell H, Kulis M, Herzog RW. Plant cell-made protein antigens for induction of Oral tolerance. Biotechnol Adv 2019; 37:107413. [PMID: 31251968 PMCID: PMC6842683 DOI: 10.1016/j.biotechadv.2019.06.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 05/21/2019] [Accepted: 06/24/2019] [Indexed: 12/15/2022]
Abstract
The gut associated lymphoid tissue has effective mechanisms in place to maintain tolerance to food antigens. These can be exploited to induce antigen-specific tolerance for the prevention and treatment of autoimmune diseases and severe allergies and to prevent serious immune responses in protein replacement therapies for genetic diseases. An oral tolerance approach for the prevention of peanut allergy in infants proved highly efficacious and advances in treatment of peanut allergy have brought forth an oral immunotherapy drug that is currently awaiting FDA approval. Several other protein antigens made in plant cells are in clinical development. Plant cell-made proteins are protected in the stomach from acids and enzymes after their oral delivery because of bioencapsulation within plant cell wall, but are released to the immune system upon digestion by gut microbes. Utilization of fusion protein technologies facilitates their delivery to the immune system, oral tolerance induction at low antigen doses, resulting in efficient induction of FoxP3+ and latency-associated peptide (LAP)+ regulatory T cells that express immune suppressive cytokines such as IL-10. LAP and IL-10 expression represent potential biomarkers for plant-based oral tolerance. Efficacy studies in hemophilia dogs support clinical development of oral delivery of bioencapsulated antigens to prevent anti-drug antibody formation. Production of clinical grade materials in cGMP facilities, stability of antigens in lyophilized plant cells for several years when stored at ambient temperature, efficacy of oral delivery of human doses in large animal models and lack of toxicity augur well for clinical advancement of this novel drug delivery concept.
Collapse
Affiliation(s)
- Henry Daniell
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Michael Kulis
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Roland W Herzog
- Department of Pediatrics, Indiana University, Indianapolis, IN 46202, USA.
| |
Collapse
|
92
|
Swaims-Kohlmeier A, Haddad LB, Li ZRT, Brookmeyer KA, Baker JM, Widom CS, Lamousin JC, Chi KH, Chen CY, Kersh EN, Johnson JA, Herbst-Kralovetz MM, Hogben M, Ofotokun I, Kohlmeier JE. Chronic immune barrier dysregulation among women with a history of violence victimization. JCI Insight 2019; 4:126097. [PMID: 31092736 DOI: 10.1172/jci.insight.126097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 04/11/2019] [Indexed: 11/17/2022] Open
Abstract
We explored the association between violence victimization and increased risk for acquiring sexually transmitted infections (STIs) in women by measuring cellular immune barrier properties from the female reproductive tract. STI-negative participants reporting repeated prior victimization occurrences through the lifetime trauma and victimization history (LTVH) instrument were more likely to exhibit alterations in barrier homeostasis and the composition of critical immune mediators irrespective of demographic parameters or presence of bacterial vaginosis. By combining cellular data with mixed-effect linear modeling, we uncovered differences in local T cells, MHCII+ antigen-presenting cells, and epithelial cells indicative of altered trafficking behavior, increased immunosuppressive function, and decreased barrier integrity at sites of STI exposure that correlate most strongly with LTVH score. These data evidence a biological link between a history of violence victimization and risk of STI acquisition through immune dysregulation in the female reproductive tract.
Collapse
Affiliation(s)
- Alison Swaims-Kohlmeier
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | - Zheng-Rong Tiger Li
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kathryn A Brookmeyer
- Social and Behavioral Research and Evaluation Branch, Division of STD Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - James M Baker
- Department of Basic Medical Sciences, Department of Obstetrics and Gynecology, The University of Arizona College of Medicine, Phoenix, Arizona, USA
| | - Cathy Spatz Widom
- Psychology Department, John Jay College of Criminal Justice, and Graduate Center, City University of New York, New York, New York, USA
| | - James C Lamousin
- Department of Mental Health, South Mississippi State Hospital, and Choices PLLC, Purvis, Mississippi, USA
| | - Kai-Hua Chi
- Laboratory Branch, Division of STD Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Cheng Y Chen
- Laboratory Branch, Division of STD Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Ellen N Kersh
- Laboratory Branch, Division of STD Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jeffrey A Johnson
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Melissa M Herbst-Kralovetz
- Department of Basic Medical Sciences, Department of Obstetrics and Gynecology, The University of Arizona College of Medicine, Phoenix, Arizona, USA
| | - Matthew Hogben
- Social and Behavioral Research and Evaluation Branch, Division of STD Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Igho Ofotokun
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jacob E Kohlmeier
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
93
|
Belarif L, Danger R, Kermarrec L, Nerrière-Daguin V, Pengam S, Durand T, Mary C, Kerdreux E, Gauttier V, Kucik A, Thepenier V, Martin JC, Chang C, Rahman A, Guen NSL, Braudeau C, Abidi A, David G, Malard F, Takoudju C, Martinet B, Gérard N, Neveu I, Neunlist M, Coron E, MacDonald TT, Desreumaux P, Mai HL, Le Bas-Bernardet S, Mosnier JF, Merad M, Josien R, Brouard S, Soulillou JP, Blancho G, Bourreille A, Naveilhan P, Vanhove B, Poirier N. IL-7 receptor influences anti-TNF responsiveness and T cell gut homing in inflammatory bowel disease. J Clin Invest 2019; 129:1910-1925. [PMID: 30939120 DOI: 10.1172/jci121668] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 02/21/2019] [Indexed: 12/16/2022] Open
Abstract
It remains unknown what causes inflammatory bowel disease (IBD), including signaling networks perpetuating chronic gastrointestinal inflammation in Crohn's disease (CD) and ulcerative colitis (UC), in humans. According to an analysis of up to 500 patients with IBD and 100 controls, we report that key transcripts of the IL-7 receptor (IL-7R) pathway are accumulated in inflamed colon tissues of severe CD and UC patients not responding to either immunosuppressive/corticosteroid, anti-TNF, or anti-α4β7 therapies. High expression of both IL7R and IL-7R signaling signature in the colon before treatment is strongly associated with nonresponsiveness to anti-TNF therapy. While in mice IL-7 is known to play a role in systemic inflammation, we found that in humans IL-7 also controlled α4β7 integrin expression and imprinted gut-homing specificity on T cells. IL-7R blockade reduced human T cell homing to the gut and colonic inflammation in vivo in humanized mouse models, and altered effector T cells in colon explants from UC patients grown ex vivo. Our findings show that failure of current treatments for CD and UC is strongly associated with an overexpressed IL-7R signaling pathway and point to IL-7R as a relevant therapeutic target and potential biomarker to fill an unmet need in clinical IBD detection and treatment.
Collapse
Affiliation(s)
| | - Richard Danger
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France
| | - Laetitia Kermarrec
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France
| | - Véronique Nerrière-Daguin
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France
| | | | - Tony Durand
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France
| | | | | | | | - Aneta Kucik
- Blizard Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | | | - Jerome C Martin
- Precision Immunology Institute.,Tisch Cancer Institute.,Department of Oncological Sciences
| | - Christie Chang
- Precision Immunology Institute.,Tisch Cancer Institute.,Department of Oncological Sciences
| | - Adeeb Rahman
- Precision Immunology Institute.,Charles Bronfman Institute for Personalized Medicine, and.,Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nina Salabert-Le Guen
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,CHU Nantes, Laboratoire d'Immunologie, Center for Immuno Monitoring Nantes-Atlantique (CIMNA), Nantes, France.,LabEx Immunograft Oncology (IGO), Nantes, France.,Université de Nantes, Faculté de Médecine, Nantes, France
| | - Cécile Braudeau
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,CHU Nantes, Laboratoire d'Immunologie, Center for Immuno Monitoring Nantes-Atlantique (CIMNA), Nantes, France.,LabEx Immunograft Oncology (IGO), Nantes, France
| | - Ahmed Abidi
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Université de Tunis El Manar, Laboratoire de génétique, immunologie et pathologies humaines, Faculté des sciences de Tunis, Tunis, Tunisia
| | - Grégoire David
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France
| | - Florent Malard
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France
| | - Celine Takoudju
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France
| | - Bernard Martinet
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France
| | - Nathalie Gérard
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France
| | - Isabelle Neveu
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France.,CHU Nantes, IMAD, Nantes, France
| | - Michel Neunlist
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France.,CHU Nantes, IMAD, Nantes, France
| | - Emmanuel Coron
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France.,CHU Nantes, IMAD, Nantes, France
| | - Thomas T MacDonald
- Blizard Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Pierre Desreumaux
- Hepato-Gastroenterology Department, Claude Huriez Hospital, University of Lille 2, Lille, France
| | - Hoa-Le Mai
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France
| | - Stephanie Le Bas-Bernardet
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France
| | - Jean-François Mosnier
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,CHU Nantes, Service d'Anatomie et Cytologie Pathologiques, Nantes, France
| | - Miriam Merad
- Precision Immunology Institute.,Tisch Cancer Institute.,Department of Oncological Sciences.,Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Régis Josien
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France.,CHU Nantes, Laboratoire d'Immunologie, Center for Immuno Monitoring Nantes-Atlantique (CIMNA), Nantes, France.,Université de Nantes, Faculté de Médecine, Nantes, France
| | - Sophie Brouard
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France
| | - Jean-Paul Soulillou
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France
| | - Gilles Blancho
- Centre de Recherche en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalier Universitaire de Nantes (CHU Nantes), Nantes, France
| | - Arnaud Bourreille
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France.,CHU Nantes, IMAD, Nantes, France
| | - Philippe Naveilhan
- Institut des Maladies de l'Appareil Digestif (IMAD), The Enteric Nervous System in Gut and Brain Disorders, Université de Nantes, INSERM, Nantes, France.,CHU Nantes, IMAD, Nantes, France
| | | | | |
Collapse
|
94
|
Huang W, Yu J, Jones JW, Carter CL, Jackson IL, Vujaskovic Z, MacVittie TJ, Kane MA. Acute Proteomic Changes in the Lung After WTLI in a Mouse Model: Identification of Potential Initiating Events for Delayed Effects of Acute Radiation Exposure. HEALTH PHYSICS 2019; 116:503-515. [PMID: 30652977 PMCID: PMC6384149 DOI: 10.1097/hp.0000000000000956] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Radiation-induced lung injury is a delayed effect of acute radiation exposure resulting in pulmonary pneumonitis and fibrosis. Molecular mechanisms that lead to radiation-induced lung injury remain incompletely understood. Using a murine model of whole-thorax lung irradiation, C57BL/6J mice were irradiated at 8, 10, 12, and 14 Gy and assayed at day 1, 3, and 6 postexposure and compared to nonirradiated (sham) controls. Tryptic digests of lung tissues were analyzed by liquid chromatography-tandem mass spectrometry on a Waters nanoLC instrument coupled to a Thermo Scientific Q Exactive hybrid quadrupole-orbitrap mass spectrometer. Pathway and gene ontology analysis were performed with Qiagen Ingenuity, Panther GO, and DAVID databases. A number of trends were identified in the proteomic data, including protein changes greater than 10 fold, protein changes that were consistently up regulated or down regulated at all time points and dose levels interrogated, time and dose dependency of protein changes, canonical pathways affected by irradiation, changes in proteins that serve as upstream regulators, and proteins involved in key processes including inflammation, radiation, and retinoic acid signaling. The proteomic profiling conducted here represents an untargeted systems biology approach to identify acute molecular events that could potentially be initiating events for radiation-induced lung injury.
Collapse
Affiliation(s)
- Weiliang Huang
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Jianshi Yu
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Jace W. Jones
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Claire L. Carter
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - I. Lauren Jackson
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| | - Zeljko Vujaskovic
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| | - Thomas J. MacVittie
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| | - Maureen A. Kane
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
- Correspondence: Maureen A. Kane, Ph.D., University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, 20 N. Pine Street, Room 723, Baltimore, MD 21201, Phone: (410) 706-5097, Fax: (410) 706-0886,
| |
Collapse
|
95
|
Hoeppli RE, MacDonald KN, Leclair P, Fung VCW, Mojibian M, Gillies J, Rahavi SMR, Campbell AIM, Gandhi SK, Pesenacker AM, Reid G, Lim CJ, Levings MK. Tailoring the homing capacity of human Tregs for directed migration to sites of Th1-inflammation or intestinal regions. Am J Transplant 2019; 19:62-76. [PMID: 29766641 DOI: 10.1111/ajt.14936] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 04/17/2018] [Accepted: 05/06/2018] [Indexed: 01/25/2023]
Abstract
Cell-based therapy with CD4+ FOXP3+ regulatory T cells (Tregs) is a promising strategy to limit organ rejection and graft-vs-host disease. Ongoing clinical applications have yet to consider how human Tregs could be modified to direct their migration to specific inflammation sites and/or tissues for more targeted immunosuppression. We show here that stable, homing-receptor-tailored human Tregs can be generated from thymic Tregs isolated from pediatric thymus or adult blood. To direct migration to Th1-inflammatory sites, addition of interferon-γ and IL-12 during Treg expansion produced suppressive, epigenetically stable CXCR3+ TBET+ FOXP3+ T helper (Th)1-Tregs. CXCR3 remained expressed after injection in vivo and Th1-Tregs migrated efficiently towards CXCL10 in vitro. To induce tissue-specific migration, addition of retinoic acid (RA) during Treg expansion induced expression of the gut-homing receptors α4β7-integrin and CCR9. FOXP3+ RA-Tregs had elevated expression of the functional markers latency-associated peptide and glycoprotein A repetitions predominant, increased suppressive capacity in vitro and migrated efficiently to healthy and inflamed intestine after injection into mice. Homing-receptor-tailored Tregs were epigenetically stable even after long-term exposure to inflammatory conditions, suppressive in vivo and characterized by Th1- or gut-homing-specific transcriptomes. Tailoring human thymic Treg homing during in vitro expansion offers a new and clinically applicable approach to improving the potency and specificity of Treg therapy.
Collapse
Affiliation(s)
- R E Hoeppli
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - K N MacDonald
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - P Leclair
- Department of Pediatrics, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - V C W Fung
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - M Mojibian
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - J Gillies
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - S M R Rahavi
- Department of Pediatrics, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - A I M Campbell
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - S K Gandhi
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - A M Pesenacker
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - G Reid
- Department of Pediatrics, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - C J Lim
- Department of Pediatrics, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - M K Levings
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
96
|
Barbalho SM, Goulart RDA, Batista GLDSA. Vitamin A and inflammatory bowel diseases: from cellular studies and animal models to human disease. Expert Rev Gastroenterol Hepatol 2019; 13:25-35. [PMID: 30791845 DOI: 10.1080/17474124.2019.1543588] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Vitamin A (VA) and metabolites such as Retinoic Acid (RA) and all-trans-RA (at-RA) are crucial in the modulation of the immune system and may be determinative in the balance of the immune responses. Inflammatory bowel diseases (IBD) consist of chronic relapsing and heterogeneous disorders with not well-known etiology. Due to its role in inflammatory processes, VA may be helpful in the treatment of IBD. Area covered: As VA plays a significant role in the inflammatory processes, this review aims to show the potential role of this vitamin in IBD, searching for cellular studies, animal models, and studies with humans. Expert commentary: Many studies have described the importance of alternative therapeutic approaches for IBD. Due to its role in the immune system, VA may also exert an indispensable role in the IBD. Nevertheless, some authors have shown that these compounds could stimulate the release of pro-inflammatory cytokines. For these reasons, more studies should be performed to establish the precise mechanisms of VA and its metabolites in systemic and intestinal inflammation.
Collapse
Affiliation(s)
- Sandra Maria Barbalho
- a School of Medicine , University of Marília (UNIMAR) , São Paulo , Brazil.,b Department of Biochemistry and Nutrition , Faculty of Food Technology of Marília (FATEC) , São Paulo , Brazil
| | | | | |
Collapse
|
97
|
de la Fuente‐Granada M, Olguín‐Alor R, Ortega‐Francisco S, Bonifaz LC, Soldevila G. Inhibins regulate peripheral regulatory T cell induction through modulation of dendritic cell function. FEBS Open Bio 2019; 9:137-147. [PMID: 30652081 PMCID: PMC6325588 DOI: 10.1002/2211-5463.12555] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/13/2018] [Accepted: 11/16/2018] [Indexed: 01/15/2023] Open
Abstract
We have previously reported that the absence of inhibins results in impaired dendritic cell (DC) maturation and function, leading to decreased T cell activation and diminished delayed-type hypersensitivity responses. Here, we investigated the role of inhibins in peripheral regulatory T cell (Treg) induction in vitro and in vivo. Inhibin deficient (Inhα-/-) mice showed an increased percentage of peripherally induced Tregs in colonic lamina propria and mesenteric lymph nodes, compared to Inhα+/+ mice, which correlated with increased expression of PD-L1 in CD103+ and CD8α+ DCs. Lipopolysaccharide-stimulated bone marrow-derived and ex vivo spleen- and lymph node-purified CD11c+ Inhα-/- DCs induced higher Tregs in vitro. Moreover, in vivo anti-DEC205-ovalbumin (OVA) DC targeting of mice with adoptively transferred OVA-specific T cells showed enhanced induced peripheral Treg conversion in Inhα-/- mice. These data identify inhibins as key regulators of peripheral T cell tolerance.
Collapse
Affiliation(s)
| | - Roxana Olguín‐Alor
- Departamento de InmunologíaInstituto de Investigaciones BiomédicasUNAMMexico CityMexico
- Laboratorio Nacional de Citometría de FlujoInstituto de Investigaciones BiomédicasUNAMMexico CityMexico
| | | | - Laura C. Bonifaz
- Unidad de Investigación Médica en InmunoquímicaInstituto Mexicano del Seguro SocialCentro Médico Nacional Siglo XXIMexico CityMexico
| | - Gloria Soldevila
- Departamento de InmunologíaInstituto de Investigaciones BiomédicasUNAMMexico CityMexico
| |
Collapse
|
98
|
Yan N, Xu J, Zhao C, Wu Y, Gao F, Li C, Zhou W, Xiao T, Zhou X, Shao Q, Xia S. Human umbilical cord-derived mesenchymal stem cells ameliorate the enteropathy of food allergies in mice. Exp Ther Med 2018; 16:4445-4456. [PMID: 30546392 PMCID: PMC6256969 DOI: 10.3892/etm.2018.6763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 08/09/2018] [Indexed: 12/13/2022] Open
Abstract
Food allergy prevalence has steadily increased worldwide over the past decades and immunotherapeutic treatment strategies are gaining attention. Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) exhibit similar immune regulatory properties to bone marrow-derived MSCs. hUC-MCSs can be prepared with fewer ethical constraints and are potential candidates for allergic disorder therapies. The current study aimed to investigate potential antiallergic properties of hUC-MSCs in mice with ovalbumin (OVA)-induced food allergy. Administration of hUC-MSCs cells intraperitoneally combined with oral gavage of the culture medium significantly alleviated OVA-induced diarrhea symptoms. Additionally, this treatment significantly decreased IgE levels and the percentage of T helper 2 cells in the blood, which were increased in mice with OVA-induced food allergy. The mRNA levels of the inflammatory cytokines interleukin-4 and tumor necrosis factor-α, and inflammatory cell infiltration in mouse colons were significantly decreased in hUC-MSCs-treated animals compared with mice with OVA-induced food allergy. Goblet cells were detected in colons of allergy-induced mice and their numbers were reduced following treatment with hUC-MSCs. In addition, treatment with hUC-MSCs reestablished the gut flora. The results revealed that hUC-MSCs may have a potential application in food allergy therapy.
Collapse
Affiliation(s)
- Nannan Yan
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jie Xu
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Chuanxiang Zhao
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yi Wu
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Fengwei Gao
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Ci Li
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wenhui Zhou
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Tengfei Xiao
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xiaoming Zhou
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Qixiang Shao
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
99
|
Li P, Wang J, Cao M, Deng Q, Jiang S, Wu MX, Lu L. Topical Application of a Vitamin A Derivative and Its Combination With Non-ablative Fractional Laser Potentiates Cutaneous Influenza Vaccination. Front Microbiol 2018; 9:2570. [PMID: 30425691 PMCID: PMC6218415 DOI: 10.3389/fmicb.2018.02570] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/09/2018] [Indexed: 12/21/2022] Open
Abstract
Skin contains a large number of antigen presenting cells, making intradermal (ID) injection one of the most effective ways for vaccine administration. However, although current adjuvants may cause severe local reactions and inflammations in the skin, no adjuvant has been approved for ID vaccination so far. Here, we report that topical application of all-trans retinoic acid (ATRA), a vitamin A derivative produced in the human body, augmented cutaneous influenza vaccination. The adjuvant effects were evaluated in a murine vaccination/challenge model by using A/California/07/2009 pandemic vaccine (09V) or a seasonal influenza vaccine (SIV). ATRA drove a Th2-biased immune response, as demonstrated by profoundly elevated IgG1 titer rather than IgG2 titer. Combining ATRA with a non-ablative fractional laser (NAFL), which represents a new category of vaccine adjuvant utilizing physical stimuli to induce self-immune stimulators, further enhanced the efficacy of influenza vaccines with a more balanced Th1/Th2 immune response. The dual adjuvant strengthened cross-reactive immune responses against both homogenous and heterogeneous influenza viral strains. Analysis of gene expression profile showed that ATRA/NAFL stimulated upregulation of cytosolic nucleic acid sensors and their downstream factors, leading to a synergistic elevation of type I interferon expression. Consistent with this finding, knocking out IRF3 or IRF7, two key downstream regulatory factors in most nucleic acid sensing pathways, resulted in a significant decrease in the adjuvant effect of ATRA/NAFL. Thus, our study demonstrates that the self molecule ATRA could boost cutaneous influenza vaccination either alone or ideally in combination with NAFL.
Collapse
Affiliation(s)
- Peiyu Li
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences & Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, United States
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People’s Hospital, Guangdong Medical University, Shenzhen, China
| | - Ji Wang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, United States
- The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Miao Cao
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences & Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Qiwen Deng
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People’s Hospital, Guangdong Medical University, Shenzhen, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences & Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| | - Mei X. Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA, United States
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, United States
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences & Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
100
|
|