101
|
Epstein-Barr Virus DNA Exacerbates Colitis Symptoms in a Mouse Model of Inflammatory Bowel Disease. Viruses 2021; 13:v13071272. [PMID: 34210024 PMCID: PMC8310145 DOI: 10.3390/v13071272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
Infection with EBV has been associated with various inflammatory disorders including inflammatory bowel diseases (IBD). Contribution of this virus to intestinal disease processes has not been assessed. We previously detected that EBV DNA triggers proinflammatory responses via the activation of endosomal Toll-like receptor (TLR) signaling. Hence, to examine the colitogenic potential of EBV DNA, we used the dextran sodium sulfate (DSS) mouse colitis model. C57BL/6J mice received either DSS-containing or regular drinking water. Mice were then administered EBV DNA by rectal gavage. Administration of EBV DNA to the DSS-fed mice aggravated colonic disease activity as well as increased the damage to the colon histologic architecture. Moreover, we observed enhanced expression of IL-17A, IFNγ and TNFα in colon tissues from the colitis mice (DSS-treated) given the EBV DNA compared to the other groups. This group also had a marked decrease in expression of the CTLA4 immunoregulatory marker. On the other hand, we observed enhanced expression of endosomal TLRs in colon tissues from the EBV DNA-treated colitis mice. These findings indicate that EBV DNA exacerbates proinflammatory responses in colitis. The ubiquity of EBV in the population indicates that possible similar responses may be of pertinence in a relevant proportion of IBD patients.
Collapse
|
102
|
Poholek AC. Tissue-Specific Contributions to Control of T Cell Immunity. Immunohorizons 2021; 5:410-423. [PMID: 34103371 PMCID: PMC10876086 DOI: 10.4049/immunohorizons.2000103] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/17/2021] [Indexed: 11/19/2022] Open
Abstract
T cells are critical for orchestrating appropriate adaptive immune responses and maintaining homeostasis in the face of persistent nonpathogenic Ags. T cell function is controlled in part by environmental signals received upon activation and derived from the tissue environment in which Ag is encountered. Indeed, tissue-specific environments play important roles in controlling the T cell response to Ag, and recent evidence suggests that tissue draining lymph nodes can mirror those local differences. Thus, tissue-specific immunity may begin at priming in secondary lymph nodes, where local signals have an important role in T cell fate. In this study, we discuss the tissue-specific signals that may impact T cell differentiation and function, including the microbiome, metabolism, and tissue-specific innate cell imprinting. We argue that these individual contributions create tissue-specific niches that likely play important roles in T cell differentiation and function controlling the outcome of the response to Ags.
Collapse
Affiliation(s)
- Amanda C Poholek
- Division of Pediatric Rheumatology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA; and Department of Immunology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
103
|
Carrasco E, Gómez de Las Heras MM, Gabandé-Rodríguez E, Desdín-Micó G, Aranda JF, Mittelbrunn M. The role of T cells in age-related diseases. Nat Rev Immunol 2021; 22:97-111. [PMID: 34099898 DOI: 10.1038/s41577-021-00557-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 12/11/2022]
Abstract
Age-related T cell dysfunction can lead to failure of immune tolerance mechanisms, resulting in aberrant T cell-driven cytokine and cytotoxic responses that ultimately cause tissue damage. In this Review, we discuss the role of T cells in the onset and progression of age-associated conditions, focusing on cardiovascular disorders, metabolic dysfunction, neuroinflammation and defective tissue repair and regeneration. We present different mechanisms by which T cells contribute to inflammageing and might act as modulators of age-associated diseases, including through enhanced pro-inflammatory and cytotoxic activity, defective clearance of senescent cells or regulation of the gut microbiota. Finally, we propose that 'resetting' immune system tolerance or targeting pathogenic T cells could open up new therapeutic opportunities to boost resilience to age-related diseases.
Collapse
Affiliation(s)
- Elisa Carrasco
- Departamento de Biología, Facultad de Ciencias (UAM); Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Biología Molecular, Facultad de Ciencias (UAM); Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel M Gómez de Las Heras
- Departamento de Biología Molecular, Facultad de Ciencias (UAM); Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Enrique Gabandé-Rodríguez
- Departamento de Biología Molecular, Facultad de Ciencias (UAM); Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Gabriela Desdín-Micó
- Departamento de Biología Molecular, Facultad de Ciencias (UAM); Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Juan Francisco Aranda
- Departamento de Biología Molecular, Facultad de Ciencias (UAM); Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Maria Mittelbrunn
- Departamento de Biología Molecular, Facultad de Ciencias (UAM); Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain. .,Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain.
| |
Collapse
|
104
|
Scheau C, Caruntu C, Badarau IA, Scheau AE, Docea AO, Calina D, Caruntu A. Cannabinoids and Inflammations of the Gut-Lung-Skin Barrier. J Pers Med 2021; 11:494. [PMID: 34072930 PMCID: PMC8227007 DOI: 10.3390/jpm11060494] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/20/2021] [Accepted: 05/28/2021] [Indexed: 12/13/2022] Open
Abstract
Recent studies have identified great similarities and interferences between the epithelial layers of the digestive tract, the airways and the cutaneous layer. The relationship between these structures seems to implicate signaling pathways, cellular components and metabolic features, and has led to the definition of a gut-lung-skin barrier. Inflammation seems to involve common features in these tissues; therefore, analyzing the similarities and differences in the modulation of its biomarkers can yield significant data promoting a better understanding of the particularities of specific signaling pathways and cellular effects. Cannabinoids are well known for a wide array of beneficial effects, including anti-inflammatory properties. This paper aims to explore the effects of natural and synthetic cannabinoids, including the components of the endocannabinoid system, in relation to the inflammation of the gut-lung-skin barrier epithelia. Recent advancements in the use of cannabinoids as anti-inflammatory substances in various disorders of the gut, lungs and skin are detailed. Some studies have reported mixed or controversial results, and these have also been addressed in our paper.
Collapse
Affiliation(s)
- Cristian Scheau
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.S.); (C.C.); (I.A.B.)
| | - Constantin Caruntu
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.S.); (C.C.); (I.A.B.)
- Department of Dermatology, “Prof. N. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Ioana Anca Badarau
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.S.); (C.C.); (I.A.B.)
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania;
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania;
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| |
Collapse
|
105
|
Wu S, Liu X, Jiang R, Yan X, Ling Z. Roles and Mechanisms of Gut Microbiota in Patients With Alzheimer's Disease. Front Aging Neurosci 2021; 13:650047. [PMID: 34122039 PMCID: PMC8193064 DOI: 10.3389/fnagi.2021.650047] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/30/2021] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is the most common age-related progressive neurodegenerative disease, characterized by a decline in cognitive function and neuronal loss, and is caused by several factors. Numerous clinical and experimental studies have suggested the involvement of gut microbiota dysbiosis in patients with AD. The altered gut microbiota can influence brain function and behavior through the microbiota-gut-brain axis via various pathways such as increased amyloid-β deposits and tau phosphorylation, neuroinflammation, metabolic dysfunctions, and chronic oxidative stress. With no current effective therapy to cure AD, gut microbiota modulation may be a promising therapeutic option to prevent or delay the onset of AD or counteract its progression. Our present review summarizes the alterations in the gut microbiota in patients with AD, the pathogenetic roles and mechanisms of gut microbiota in AD, and gut microbiota-targeted therapies for AD. Understanding the roles and mechanisms between gut microbiota and AD will help decipher the pathogenesis of AD from novel perspectives and shed light on novel therapeutic strategies for AD.
Collapse
Affiliation(s)
- Shaochang Wu
- Department of Geriatrics, Lishui Second People’s Hospital, Lishui, China
| | - Xia Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ruilai Jiang
- Department of Geriatrics, Lishui Second People’s Hospital, Lishui, China
| | - Xiumei Yan
- Department of Geriatrics, Lishui Second People’s Hospital, Lishui, China
| | - Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Microbe & Host Health, Linyi University, Linyi, China
| |
Collapse
|
106
|
Kanova M, Kohout P. Serotonin-Its Synthesis and Roles in the Healthy and the Critically Ill. Int J Mol Sci 2021; 22:ijms22094837. [PMID: 34063611 PMCID: PMC8124334 DOI: 10.3390/ijms22094837] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/25/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) plays two important roles in humans-one central and the other peripheral-depending on the location of the 5-HT pools of on either side of the blood-brain barrier. In the central nervous system it acts as a neurotransmitter, controlling such brain functions as autonomic neural activity, stress response, body temperature, sleep, mood and appetite. This role is very important in intensive care, as in critically ill patients multiple serotoninergic agents like opioids, antiemetics and antidepressants are frequently used. High serotonin levels lead to altered mental status, deliria, rigidity and myoclonus, together recognized as serotonin syndrome. In its role as a peripheral hormone, serotonin is unique in controlling the functions of several organs. In the gastrointestinal tract it is important for regulating motor and secretory functions. Apart from intestinal motility, energy metabolism is regulated by both central and peripheral serotonin signaling. It also has fundamental effects on hemostasis, vascular tone, heart rate, respiratory drive, cell growth and immunity. Serotonin regulates almost all immune cells in response to inflammation, following the activation of platelets.
Collapse
Affiliation(s)
- Marcela Kanova
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Ostrava, 70852 Ostrava-Poruba, Czech Republic
- Institute of Physiology and Pathophysiology, Faculty of Medicine, University of Ostrava, Syllabova 19, 70300 Ostrava-Vítkovice, Czech Republic
- Correspondence: ; Tel.: +420-59737-2707
| | - Pavel Kohout
- Department of Internal Medicine, 3rd Faculty of Medicine, Charles University Prague and Teaching Thomayer Hospital, 14059 Prague, Czech Republic;
| |
Collapse
|
107
|
Hatscher L, Lehmann CHK, Purbojo A, Onderka C, Liang C, Hartmann A, Cesnjevar R, Bruns H, Gross O, Nimmerjahn F, Ivanović-Burmazović I, Kunz M, Heger L, Dudziak D. Select hyperactivating NLRP3 ligands enhance the T H1- and T H17-inducing potential of human type 2 conventional dendritic cells. Sci Signal 2021; 14:14/680/eabe1757. [PMID: 33906973 DOI: 10.1126/scisignal.abe1757] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The detection of microorganisms and danger signals by pattern recognition receptors on dendritic cells (DCs) and the consequent formation of inflammasomes are pivotal for initiating protective immune responses. Although the activation of inflammasomes leading to secretion of the cytokine IL-1β is typically accompanied by pyroptosis (an inflammatory form of lytic programmed cell death), some cells can survive and exist in a state of hyperactivation. Here, we found that the conventional type 2 DC (cDC2) subset is the major human DC subset that is transcriptionally and functionally poised for inflammasome formation and response without pyroptosis. When cDC2 were stimulated with ligands that relatively weakly activated the inflammasome, the cells did not enter pyroptosis but instead secreted IL-12 family cytokines and IL-1β. These cytokines induced prominent T helper type 1 (TH1) and TH17 responses that were superior to those seen in response to Toll-like receptor (TLR) stimulation alone or to stronger, classical inflammasome ligands. These findings not only define the human cDC2 subpopulation as a prime target for the treatment of inflammasome-dependent inflammatory diseases but may also inform new approaches for adjuvant and vaccine development.
Collapse
Affiliation(s)
- Lukas Hatscher
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052 Erlangen, Germany
| | - Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052 Erlangen, Germany
| | - Ariawan Purbojo
- Department of Pediatric Cardiac Surgery, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Constantin Onderka
- Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Chunguang Liang
- Chair of Medical Informatics, Friedrich-Alexander University Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Arndt Hartmann
- Department of Pathology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Robert Cesnjevar
- Department of Pediatric Cardiac Surgery, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Heiko Bruns
- Department of Internal Medicine 5-Hematology/Oncology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Olaf Gross
- Institute of Neuropathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Falk Nimmerjahn
- Institute of Genetics, Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany
| | - Ivana Ivanović-Burmazović
- Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nürnberg, 91058 Erlangen, Germany.,Department Chemistry, Ludwigs Maximilians University, 81377 Munich, Germany
| | - Meik Kunz
- Chair of Medical Informatics, Friedrich-Alexander University Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052 Erlangen, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052 Erlangen, Germany. .,Institute of Genetics, Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany.,Deutsches Zentrum Immuntherapie, 91054 Erlangen, Germany.,Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg, 91054 Erlangen, Germany.,Medical Immunology Campus Erlangen, 91054 Erlangen, Germany
| |
Collapse
|
108
|
Abstract
PURPOSE OF REVIEW Although the gut microbiome plays a crucial role in the maintenance of health, it is hypothesized to drive morbidity and mortality in critically ill patients. This review describes the relationship between the gut microbiome and the immune system in critical illness. RECENT FINDINGS The gut microbiome is converted to a pathobiome in the ICU, characterized by decreased microbial diversity and pathogen predominance. These changes are induced by a pathologic microenvironment and are further exacerbated by common medical treatments initiated in the ICU. The conversion of the microbiome to a pathobiome has direct consequences on the regulation of inflammation and immunity by loss of beneficial host responses and initiation of maladaptive changes that can further propagate critical illness. SUMMARY The gut microbiome is dramatically altered in the ICU. In light of constant crosstalk between the microbiome and the host immune system, the pathobiome may play a key mechanistic role in driving a maladaptive response in critically ill patients. The pathobiome represents a potential therapeutic target in the management of critical illness whereby restoration of a healthier microbiome may directly alter the host inflammatory response, which could lead to improved patient outcomes.
Collapse
Affiliation(s)
- Ashley A Miniet
- Division of Critical Care Medicine, Department of Pediatrics, Emory University School of Medicine
- Children's Healthcare of Atlanta at Egleston
| | - Jocelyn R Grunwell
- Division of Critical Care Medicine, Department of Pediatrics, Emory University School of Medicine
- Children's Healthcare of Atlanta at Egleston
| | - Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
109
|
Li X, Colamatteo A, Kalafati L, Kajikawa T, Wang H, Lim JH, Bdeir K, Chung KJ, Yu X, Fusco C, Porcellini A, De Simone S, Matarese G, Chavakis T, De Rosa V, Hajishengallis G. The DEL-1/β3 integrin axis promotes regulatory T cell responses during inflammation resolution. J Clin Invest 2021; 130:6261-6277. [PMID: 32817592 DOI: 10.1172/jci137530] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023] Open
Abstract
FOXP3+CD4+ regulatory T cells (Tregs) are critical for immune homeostasis and respond to local tissue cues, which control their stability and function. We explored here whether developmental endothelial locus-1 (DEL-1), which, like Tregs, increases during resolution of inflammation, promotes Treg responses. DEL-1 enhanced Treg numbers and function at barrier sites (oral and lung mucosa). The underlying mechanism was dissected using mice lacking DEL-1 or expressing a point mutant thereof, or mice with T cell-specific deletion of the transcription factor RUNX1, identified by RNA sequencing analysis of the DEL-1-induced Treg transcriptome. Specifically, through interaction with αvβ3 integrin, DEL-1 promoted induction of RUNX1-dependent FOXP3 expression and conferred stability of FOXP3 expression upon Treg restimulation in the absence of exogenous TGF-β1. Consistently, DEL-1 enhanced the demethylation of the Treg-specific demethylated region (TSDR) in the mouse Foxp3 gene and the suppressive function of sorted induced Tregs. Similarly, DEL-1 increased RUNX1 and FOXP3 expression in human conventional T cells, promoting their conversion into induced Tregs with increased TSDR demethylation, enhanced stability, and suppressive activity. We thus uncovered a DEL-1/αvβ3/RUNX1 axis that promotes Treg responses at barrier sites and offers therapeutic options for modulating inflammatory/autoimmune disorders.
Collapse
Affiliation(s)
- Xiaofei Li
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alessandra Colamatteo
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II," Naples, Italy
| | - Lydia Kalafati
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,National Center for Tumor Diseases, Dresden, Germany, and German Cancer Research Center, Heidelberg, Germany
| | - Tetsuhiro Kajikawa
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hui Wang
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jong-Hyung Lim
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Khalil Bdeir
- Department of Pathology and Laboratory Medicine and
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Xiang Yu
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Clorinda Fusco
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II," Naples, Italy
| | - Antonio Porcellini
- Dipartimento di Biologia, Università degli Studi di Napoli "Federico II," Complesso Universitario di Monte Santangelo, Naples, Italy
| | - Salvatore De Simone
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Naples, Italy
| | - Giuseppe Matarese
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II," Naples, Italy.,Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Naples, Italy
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Veronica De Rosa
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Naples, Italy.,Unità di Neuroimmunologia, Fondazione Santa Lucia, Rome, Italy
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
110
|
Weidmann MD, Ofori K, Rai AJ. Laboratory Biomarkers in the Management of Patients With COVID-19. Am J Clin Pathol 2021; 155:333-342. [PMID: 33107558 PMCID: PMC7665296 DOI: 10.1093/ajcp/aqaa205] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES Laboratory testing and the measurement of appropriate biomarkers play a critical role in managing patients with coronavirus disease 2019 (COVID-19), allowing for disease diagnosis, monitoring progression, prognostication, prediction of treatment response, and risk stratification. We sought to characterize these effects on a more detailed, mechanistic level. METHODS We reviewed the literature and identified a multitude of reports that describe the unique effects of this virus and its devastating consequences to multiple organ systems in COVID-19 patients. RESULTS There are specific alterations in biomarkers related to coagulation, depopulation of T-cell subtypes, the cytokine storm and inflammation, and kidney and cardiac dysfunction. CONCLUSIONS Laboratory measurement of specific parameters and the use of appropriate prognostic, predictive, and monitoring biomarkers afford clinicians the ability to make informed medical decisions and guide therapy for patients afflicted with this dreaded disease.
Collapse
Affiliation(s)
- Maxwell D Weidmann
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Kenneth Ofori
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Alex J Rai
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
111
|
Sasaki T, Nagata R, Takahashi S, Takei Y. Effects of RORγt overexpression on the murine central nervous system. Neuropsychopharmacol Rep 2021; 41:102-110. [PMID: 33547881 PMCID: PMC8182958 DOI: 10.1002/npr2.12162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 01/07/2021] [Accepted: 01/19/2021] [Indexed: 12/18/2022] Open
Abstract
Objective T helper 17 (Th17) cells are a subset of CD4+ T cells that produce interleukin (IL)‐17A. Recent studies showed that an increase in circulating IL‐17A causes cognitive dysfunction, although it is unknown how increased systemic IL‐17A affects brain function. Using transgenic mice overexpressing RORγt, a transcription factor essential for differentiation of Th17 cells (RORγt Tg mice), we examined changes in the brain caused by chronically increased IL‐17A resulting from excessive activation of Th17 cells. Results RORγt Tg mice exhibited elevated Rorc and IL‐17A mRNA expression in the colon, as well as a chronic increase in circulating IL‐17A. We found that the immunoreactivity of Iba1 and density of microglia were lower in the dentate gyrus of RORγt Tg mice compared with wild‐type mice. However, GFAP+ astrocytes were unchanged in the hippocampi of RORγt Tg mice. Levels of synaptic proteins were not significantly different between RORγt Tg and wild‐type mouse brains. In addition, novel object location test results indicated no difference in preference between these mice. Conclusion Our findings indicate that a continuous increase of IL‐17A in response to RORγt overexpression resulted in decreased microglia activity in the dentate gyrus, but had only a subtle effect on murine hippocampal functions. Using transgenic mice overexpressing RORγt, a transcription factor essential for differentiation of Th17 cells, we examined changes in the brain caused by chronically increased IL‐17A resulting from excessive activation of Th17 cells. RORγt Tg mice exhibited elevated Rorc and IL‐17A mRNA expression in the colon, as well as a chronic increase in circulating IL‐17A. Our findings indicate that a continuous increase of IL‐17A in response to RORγt overexpression resulted in decreased microglia activity in the dentate gyrus but had a subtle effect on murine hippocampal functions.![]()
Collapse
Affiliation(s)
- Tetsuya Sasaki
- Faculty of Medicine, Department of Anatomy and Neuroscience, University of Tsukuba, Tsukuba, Ibaraki, Japan.,PhD Program of Neurosciences, Degree Program of Comprehensive Human Sciences, Graduate School of Comprehensive Human Sciences,, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Rei Nagata
- Faculty of Medicine, Department of Anatomy and Neuroscience, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Satoru Takahashi
- Faculty of Medicine, Department of Anatomy and Embryology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yosuke Takei
- Faculty of Medicine, Department of Anatomy and Neuroscience, University of Tsukuba, Tsukuba, Ibaraki, Japan.,PhD Program of Neurosciences, Degree Program of Comprehensive Human Sciences, Graduate School of Comprehensive Human Sciences,, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
112
|
Regen T, Isaac S, Amorim A, Núñez NG, Hauptmann J, Shanmugavadivu A, Klein M, Sankowski R, Mufazalov IA, Yogev N, Huppert J, Wanke F, Witting M, Grill A, Gálvez EJC, Nikolaev A, Blanfeld M, Prinz I, Schmitt-Kopplin P, Strowig T, Reinhardt C, Prinz M, Bopp T, Becher B, Ubeda C, Waisman A. IL-17 controls central nervous system autoimmunity through the intestinal microbiome. Sci Immunol 2021; 6:6/56/eaaz6563. [PMID: 33547052 DOI: 10.1126/sciimmunol.aaz6563] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/24/2020] [Indexed: 12/17/2022]
Abstract
Interleukin-17A- (IL-17A) and IL-17F-producing CD4+ T helper cells (TH17 cells) are implicated in the development of chronic inflammatory diseases, such as multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). TH17 cells also orchestrate leukocyte invasion of the central nervous system (CNS) and subsequent tissue damage. However, the role of IL-17A and IL-17F as effector cytokines is still confused with the encephalitogenic function of the cells that produce these cytokines, namely, TH17 cells, fueling a long-standing debate in the neuroimmunology field. Here, we demonstrated that mice deficient for IL-17A/F lose their susceptibility to EAE, which correlated with an altered composition of their gut microbiota. However, loss of IL-17A/F in TH cells did not diminish their encephalitogenic capacity. Reconstitution of a wild-type-like intestinal microbiota or reintroduction of IL-17A specifically into the gut epithelium of IL-17A/F-deficient mice reestablished their susceptibility to EAE. Thus, our data demonstrated that IL-17A and IL-17F are not encephalitogenic mediators but rather modulators of intestinal homeostasis that indirectly alter CNS-directed autoimmunity.
Collapse
Affiliation(s)
- Tommy Regen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Sandrine Isaac
- Department of Genomics and Health, Center for Advanced Research in Public Health, FISABIO, Valencia, Spain
| | - Ana Amorim
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Judith Hauptmann
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Arthi Shanmugavadivu
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Matthias Klein
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Roman Sankowski
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany.,Berta-Ottenstein-Program for Clinician Scientists, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ilgiz A Mufazalov
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Nir Yogev
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Jula Huppert
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Florian Wanke
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Michael Witting
- Research Unit Analytical BioGeoChemistry, Helmholtz Center Munich, Munich, Germany.,Chair of Analytical Food Chemistry, TUM School of Life Sciences, Technical University Munich, Munich, Germany
| | - Alexandra Grill
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Eric J C Gálvez
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Alexei Nikolaev
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Michaela Blanfeld
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Philippe Schmitt-Kopplin
- Research Unit Analytical BioGeoChemistry, Helmholtz Center Munich, Munich, Germany.,Chair of Analytical Food Chemistry, TUM School of Life Sciences, Technical University Munich, Munich, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany.,Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Center for NeuroModulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Carles Ubeda
- Department of Genomics and Health, Center for Advanced Research in Public Health, FISABIO, Valencia, Spain.,Center of Biomedical Research Network (CIBER), Epidemiology and Public Health, Madrid, Spain
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany. .,Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
113
|
Mikami A, Ogita T, Namai F, Shigemori S, Sato T, Shimosato T. Oral Administration of Flavonifractor plautii, a Bacteria Increased With Green Tea Consumption, Promotes Recovery From Acute Colitis in Mice via Suppression of IL-17. Front Nutr 2021; 7:610946. [PMID: 33614691 PMCID: PMC7890079 DOI: 10.3389/fnut.2020.610946] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022] Open
Abstract
Flavonifractor plautii (FP) has been reported to participate in the metabolism of catechins in the human gut. However, there is limited information on the immune regulatory effects of this bacterium. We confirmed that the administration of green tea increases the abundance of FP in the gut microbiota and investigated the effect of FP in a mouse colitis model. Mice were orally administered FP for 10 consecutive days; colonic inflammation was evaluated daily on the basis of stool consistency, gross rectal bleeding, and body weight. In the dextran sodium sulfate model, FP-exposed animals exhibited lower levels of inflammation and strong inhibition of interleukin (IL)-17 signaling. Moreover, lipoteichoic acid from FP was identified as the active component mediating IL-17 suppression. Thus, oral administration of FP appears to modulate gut inflammation and represents a viable and inexpensive oral microbial therapeutic.
Collapse
Affiliation(s)
- Ayane Mikami
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Tasuku Ogita
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Fu Namai
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Suguru Shigemori
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Takashi Sato
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Takeshi Shimosato
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| |
Collapse
|
114
|
Qu SL, Chen L, Wen XS, Zuo JP, Wang XY, Lu ZJ, Yang YF. Suppression of Th17 cell differentiation via sphingosine-1-phosphate receptor 2 by cinnamaldehyde can ameliorate ulcerative colitis. Biomed Pharmacother 2021; 134:111116. [PMID: 33341041 DOI: 10.1016/j.biopha.2020.111116] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022] Open
Abstract
Ulcerative colitis (UC) is chronic disease characterized by diffuse inflammation of the mucosa of the colon and rectum. Although the etiology is unknown, dysregulation of the intestinal mucosal immune system is closely related to UC. Cinnamaldehyde (CA) is a major active compound from cinnamon, is known as its anti-inflammatory and antibacterial. However, little research focused on its regulatory function on immune cells in UC. Therefore, we set out to explore the modulating effects of CA on immune cells in UC. We found that CA reduced the progression of colitis through controlling the production of proinflammatory cytokines and inhibiting the proportion of Th17 cells. Furthermore, the liquid chromatography-mass spectrometry (LC-MS) method was employed for analyzing and differentiating metabolites, data showed that sphingolipid pathway has a great influence on the effect of CA on UC. Meanwhile, sphingosine-1-phosphate receptor 2 (S1P2) and Rho-GTP protein levels were downregulated in colonic tissues after CA treatment. Moreover, in vitro assays showed that CA inhibited Th17 cell differentiation and downregulated of S1P2 and Rho-GTP signaling. Notably, we found that treatment with S1P2 antagonist (JTE-013) weakened the inhibitory effect of CA on Th17 cells. Furthermore, S1P2 deficiency (S1P2-/-) blocked the effect of CA on Th17 cell differentiation. In addition, CA can also improve inflammation via lncRNA H19 and MIAT. To sum up, this study provides clear evidence that CA can ameliorate ulcerative colitis through suppressing Th17 cells via S1P2 pathway and regulating lncRNA H19 and MIAT, which further supports S1P2 as a potential drug target for immunity-mediated UC.
Collapse
Affiliation(s)
- Shu-Lan Qu
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Long Chen
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xue-Shan Wen
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jian-Ping Zuo
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Laboratory of Anti-inflammation and Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiao-Yu Wang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Laboratory of Anti-inflammation and Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Zhi-Jie Lu
- Department of Anesthesiology and Intensive Care Unit, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China.
| | - Yi-Fu Yang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
115
|
Nelson JW, Phillips SC, Ganesh BP, Petrosino JF, Durgan DJ, Bryan RM. The gut microbiome contributes to blood-brain barrier disruption in spontaneously hypertensive stroke prone rats. FASEB J 2021; 35:e21201. [PMID: 33496989 PMCID: PMC8238036 DOI: 10.1096/fj.202001117r] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/09/2020] [Accepted: 11/03/2020] [Indexed: 11/11/2022]
Abstract
In recent years, it has become apparent that the gut microbiome can influence the functioning and pathological states of organs and systems throughout the body. In this study, we tested the hypothesis that the gut microbiome has a major role in the disruption of the blood-brain barrier (BBB) in the spontaneously hypertensive stroke prone rats (SHRSP), an animal model for hypertensive cerebral small vessel disease (CSVD). Loss of BBB is thought to be an early and initiating component to the full expression of CSVD in animal models and humans. To test this hypothesis, newly born SHRSP pups were placed with foster dams of the SHRSP strain or dams of the WKY strain, the control strain that does not demonstrate BBB dysfunction or develop hypertensive CSVD. Similarly, WKY pups were placed with foster dams of the same or opposite strain. The rationale for cross fostering is that the gut microbiomes are shaped by environmental bacteria of the foster dam and the nesting surroundings. Analysis of the bacterial genera in feces, using 16S rRNA analysis, demonstrated that the gut microbiome in the rat pups was influenced by the foster dam. SHRSP offspring fostered on WKY dams had systolic blood pressures (SBPs) that were significantly decreased by 26 mmHg (P < .001) from 16-20 weeks, compared to SHRSP offspring fostered on SHRSP dams. Similarly WKY offspring fostered on SHRSP dams had significantly increased SBP compared to WKY offspring fostered on WKY dams, although the magnitude of SBP change was not as robust. At ~20 weeks of age, rats fostered on SHRSP dams showed enhanced inflammation in distal ileum regardless of the strain of the offspring. Disruption of BBB integrity, an early marker of CSVD onset, was improved in SHRSPs that were fostered on WKY dams when compared to the SHRSP rats fostered on SHRSP dams. Although SHRSP is a genetic model for CSVD, environmental factors such as the gut microbiota of the foster dam have a major influence in the loss of BBB integrity.
Collapse
Affiliation(s)
- James W. Nelson
- Department of Anesthesiology, Baylor College of Medicine, Houston, TX, USA
- Integrated Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Sharon C. Phillips
- Department of Anesthesiology, Baylor College of Medicine, Houston, TX, USA
| | - Bhanu P. Ganesh
- Department of Neurology, University of Texas Health Sciences Center, Houston, TX, USA
| | - Joseph F. Petrosino
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- The Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - David J. Durgan
- Department of Anesthesiology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Robert M. Bryan
- Department of Anesthesiology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
116
|
Kim S, Park HE, Park WB, Kim SY, Park HT, Yoo HS. Mycobacterium avium Modulates the Protective Immune Response in Canine Peripheral Blood Mononuclear Cells. Front Cell Infect Microbiol 2021; 10:609712. [PMID: 33520738 PMCID: PMC7840563 DOI: 10.3389/fcimb.2020.609712] [Citation(s) in RCA: 3] [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: 09/24/2020] [Accepted: 11/30/2020] [Indexed: 12/31/2022] Open
Abstract
Mycobacterium avium, an opportunistic intracellular pathogen, is a member of the non-tuberculous mycobacteria species. M. avium causes respiratory disease in immunosuppressed individuals and a wide range of animals, including companion dogs and cats. In particular, the number of infected companion dogs has increased, although the underlying mechanism of M. avium pathogenesis in dogs has not been studied. Therefore, in the present study, the host immune response against M. avium in dogs was investigated by transcriptome analysis of canine peripheral blood mononuclear cells. M. avium was shown to induce different immune responses in canine peripheral blood mononuclear cells at different time points after infection. The expression of Th1-associated genes occurred early during M. avium infection, while that of Th17-associated genes increased after 12 h. In addition, the expression of apoptosis-related genes decreased and the abundance of intracellular M. avium increased in monocyte-derived macrophages after infection for 24 h. These results reveal the M. avium induces Th17 immune response and avoids apoptosis in infected canine cells. As the number of M. avium infection cases increases, the results of the present study will contribute to a better understanding of host immune responses to M. avium infection in companion dogs.
Collapse
Affiliation(s)
- Suji Kim
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
- BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul, South Korea
| | - Hyun-Eui Park
- Department of Microbiology, College of Medicine, Gyeongsang National University, Jinju, South Korea
| | - Woo Bin Park
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Seo Yihl Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hong-Tae Park
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Han Sang Yoo
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
- BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul, South Korea
- Bio-MAX/N-Bio Institute, Seoul National University, Seoul, South Korea
| |
Collapse
|
117
|
Nandi D, Pathak S, Verma T, Singh M, Chattopadhyay A, Thakur S, Raghavan A, Gokhroo A, Vijayamahantesh. T cell costimulation, checkpoint inhibitors and anti-tumor therapy. J Biosci 2021. [PMID: 32345776 DOI: 10.1007/s12038-020-0020-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The hallmarks of the adaptive immune response are specificity and memory. The cellular response is mediated by T cells which express cell surface T cell receptors (TCRs) that recognize peptide antigens in complex with major histocompatibility complex (MHC) molecules on antigen presenting cells (APCs). However, binding of cognate TCRs with MHC-peptide complexes alone (signal 1) does not trigger optimal T cell activation. In addition to signal 1, the binding of positive and negative costimulatory receptors to their ligands modulates T cell activation. This complex signaling network prevents aberrant activation of T cells. CD28 is the main positive costimulatory receptor on naı¨ve T cells; upon activation, CTLA4 is induced but reduces T cell activation. Further studies led to the identification of additional negative costimulatory receptors known as checkpoints, e.g. PD1. This review chronicles the basic studies in T cell costimulation that led to the discovery of checkpoint inhibitors, i.e. antibodies to negative costimulatory receptors (e.g. CTLA4 and PD1) which reduce tumor growth. This discovery has been recognized with the award of the 2018 Nobel prize in Physiology/Medicine. This review highlights the structural and functional roles of costimulatory receptors, the mechanisms by which checkpoint inhibitors work, the challenges encountered and future prospects.
Collapse
Affiliation(s)
- Dipankar Nandi
- Department of Biochemistry, Indian Institute of Science, Bengaluru 560 012, India
| | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Fukuzaki S, Righetti RF, Santos TMD, Camargo LDN, Aristóteles LRCRB, Souza FCR, Garrido AC, Saraiva-Romanholo BM, Leick EA, Prado CM, Martins MDA, Tibério IDFLC. Preventive and therapeutic effect of anti-IL-17 in an experimental model of elastase-induced lung injury in C57Bl6 mice. Am J Physiol Cell Physiol 2020; 320:C341-C354. [PMID: 33326311 DOI: 10.1152/ajpcell.00017.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is an important health care issue, and IL-17 can modulate inflammatory responses. We evaluated preventive and therapeutic effect of anti-interleukin (IL)-17 in a model of lung injury induced by elastase, using 32 male C57Bl6 mice, divided into 4 groups: SAL, ELASTASE CONTROL (EC), ELASTASE + PREVENTIVE ANTI-IL-17 (EP), and ELASTASE + THERAPEUTIC ANTI-IL-17 (ET). On the 29th day, animals were anesthetized with thiopental, tracheotomized, and placed on a ventilator to evaluate lung mechanical, exhaled nitric oxide (eNO), and total cells of bronchoalveolar lavage fluid was collected. We performed histological techniques, and linear mean intercept (Lm) was analyzed. Both treatments with anti-IL-17 decreased respiratory resistance and elastance, airway resistance, elastance of pulmonary parenchyma, eNO, and Lm compared with EC. There was reduction in total cells and macrophages in ET compared with EC. Both treatments decreased nuclear factor-кB, inducible nitric oxide synthase, matrix metalloproteinase (MMP)-9, MMP-12, transforming growth factor-β, tumor necrosis factor-α, neutrophils, IL-1β, isoprostane, and IL-17 in airways and alveolar septa; collagen fibers, decorin and lumican in airways; and elastic fibers and fibronectin in alveolar septa compared with EC. There was reduction of collagen fibers in alveolar septa and biglycan in airways in EP and a reduction of eNO synthase in airways in ET. In conclusion, both treatments with anti-IL-17 contributed to improve most of parameters evaluated in inflammation and extracellular matrix remodeling in this model of lung injury.
Collapse
Affiliation(s)
- Silvia Fukuzaki
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Renato Fraga Righetti
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil.,Hospital Sírio-Libanês, São Paulo, Brazil
| | - Tabata Maruyama Dos Santos
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil.,Hospital Sírio-Libanês, São Paulo, Brazil
| | - Leandro do Nascimento Camargo
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil.,Hospital Sírio-Libanês, São Paulo, Brazil
| | | | - Flavia C R Souza
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Aurelio C Garrido
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Beatriz Mangueira Saraiva-Romanholo
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil.,Department of Medicine (LIM 20), Hospital Public Employee of São Paulo (Instituto de Assistência Médica ao Servidor Público Estadual de São Paulo), University City of São Paulo, São Paulo, Brazil
| | - Edna Aparecida Leick
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Carla Máximo Prado
- School of Medicine-Faculty of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil.,Department of Bioscience, Federal University of São Paulo, Santos, São Paulo, Brazil
| | | | | |
Collapse
|
119
|
Longitudinal Analysis of Peripheral and Colonic CD161 + CD4 + T Cell Dysfunction in Acute HIV-1 Infection and Effects of Early Treatment Initiation. Viruses 2020; 12:v12121426. [PMID: 33322496 PMCID: PMC7764746 DOI: 10.3390/v12121426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/25/2020] [Accepted: 11/27/2020] [Indexed: 01/13/2023] Open
Abstract
CD161 expression on CD4+ T cells is associated with a Th17 functional phenotype, as well as with an innate capacity to respond to interleukin (IL)-12 and IL-18 without T cell receptor (TCR) stimulation. Chronic HIV-1 infection is associated with loss of the CD161+ CD4 T cell population, and non-human primate studies suggest that their depletion is associated with disease progression. However, the dynamics of the CD161+ CD4+ T cell population during acute HIV-1 infection remains unknown. In this study, we characterize peripheral blood CD161+ CD4+ T cells in detail, and examine how they are affected during the earliest stages of HIV-1 infection. Unbiased surface proteome screening and principal component analysis indicated that CD161+ CD4+ T cells are relatively phenotypically homogeneous between donors, and are intermediates between conventional CD4 T cells and innate-like T cells. In acute untreated HIV-1 infection, the circulating CD161+ CD4+ T cell population decreased in frequency, as did absolute cell counts starting from peak viral load, with elevated levels of activation and exhaustion markers expressed throughout acute HIV-1 infection. The capacity of these cells to respond to stimulation with IL-12 and IL-18 was also reduced. Early initiation of anti-retroviral treatment (ART) during acute HIV-1 infection restored the functionality of peripheral blood CD161+ CD4+ T cells, but not their frequency. In contrast, early ART initiation prevented the decline of colonic CD161+ CD4+ T cells that otherwise started during acute infection. Furthermore, loss of peripheral and colonic CD161+ CD4+ T cells in untreated infection was associated with levels of viral load. These results suggest that acute HIV-1 infection has profound effects on the CD161+ CD4+ T cell population that could not be completely prevented by the initiation of ART.
Collapse
|
120
|
Liu NN, Ma Q, Ge Y, Yi CX, Wei LQ, Tan JC, Chu Q, Li JQ, Zhang P, Wang H. Microbiome dysbiosis in lung cancer: from composition to therapy. NPJ Precis Oncol 2020; 4:33. [PMID: 33303906 PMCID: PMC7730185 DOI: 10.1038/s41698-020-00138-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 10/02/2020] [Indexed: 02/07/2023] Open
Abstract
The correlations between microbiota dysbiosis and cancer have gained extensive attention and been widely explored. As a leading cancer diagnosis worldwide, lung cancer poses a great threat to human health. The healthy human lungs are consistently exposed to external environment and harbor a specific pattern of microbiota, sharing many key pathological and physiological characteristics with the intestinal tract. Although previous findings uncovered the critical roles of microbiota in tumorigenesis and response to anticancer therapy, most of them were focused on the intestinal microbiota rather than lung microbiota. Notably, the considerable functions of microbiota in maintaining lung homeostasis should not be neglected as the microbiome dysbiosis may promote tumor development and progression through production of cytokines and toxins and multiple other pathways. Despite the fact that increasing studies have revealed the effect of microbiome on the induction of lung cancer and different disease status, the underlying mechanisms and potential therapeutic strategies remained unclear. Herein, we summarized the recent progresses about microbiome in lung cancer and further discussed the role of microbial communities in promoting lung cancer progression and the current status of therapeutic approaches targeting microbiome to alleviate and even cure lung cancer.
Collapse
Affiliation(s)
- Ning-Ning Liu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Qiang Ma
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital Tongji University, Shanghai, China
| | - Yang Ge
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Cheng-Xiang Yi
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital Tongji University, Shanghai, China
| | - Lu-Qi Wei
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jing-Cong Tan
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Qiao Chu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jing-Quan Li
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital Tongji University, Shanghai, China.
| | - Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| |
Collapse
|
121
|
Ehteshamfar S, Akhbari M, Afshari JT, Seyedi M, Nikfar B, Shapouri‐Moghaddam A, Ghanbarzadeh E, Momtazi‐Borojeni AA. Anti-inflammatory and immune-modulatory impacts of berberine on activation of autoreactive T cells in autoimmune inflammation. J Cell Mol Med 2020; 24:13573-13588. [PMID: 33135395 PMCID: PMC7754052 DOI: 10.1111/jcmm.16049] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/29/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
Autoreactive inflammatory CD4+ T cells, such as T helper (Th)1 and Th17 subtypes, have been found to associate with the pathogenesis of autoimmune disorders. On the other hand, CD4+ Foxp3+ T regulatory (Treg) cells are crucial for the immune tolerance and have a critical role in the suppression of the excessive immune and inflammatory response promoted by these Th cells. In contrast, dendritic cells (DCs) and macrophages are immune cells that through their inflammatory functions promote autoreactive T-cell responses in autoimmune conditions. In recent years, there has been increasing attention to exploring effective immunomodulatory or anti-inflammatory agents from the herbal collection of traditional medicine. Berberine, an isoquinoline alkaloid, is one of the main active ingredients extracted from medicinal herbs and has been shown to exert various biological and pharmacological effects that are suggested to be mainly attributed to its anti-inflammatory and immunomodulatory properties. Several lines of experimental study have recently investigated the therapeutic potential of berberine for treating autoimmune conditions in animal models of human autoimmune diseases. Here, we aimed to seek mechanisms underlying immunomodulatory and anti-inflammatory effects of berberine on autoreactive inflammatory responses in autoimmune conditions. Reported data reveal that berberine can directly suppress functions and differentiation of pro-inflammatory Th1 and Th17 cells, and indirectly decrease Th cell-mediated inflammation through modulating or suppressing other cells assisting autoreactive inflammation, such as Tregs, DCs and macrophages.
Collapse
Affiliation(s)
- Seyed‐Morteza Ehteshamfar
- Department of ImmunologyFaculty of MedicineBuAli Research InstituteMashhad University of Medical SciencesMashhadIran
| | - Masoume Akhbari
- Department of Molecular MedicineSchool of MedicineQazvin University of Medical SciencesQazvinIran
| | - Jalil Tavakol Afshari
- Department of ImmunologyFaculty of MedicineBuAli Research InstituteMashhad University of Medical SciencesMashhadIran
| | | | - Banafsheh Nikfar
- Pars Advanced and Minimally Invasive Medical Manners Research CenterPars HospitalIran University of Medical SciencesTehranIran
| | - Abbas Shapouri‐Moghaddam
- Department of ImmunologyFaculty of MedicineBuAli Research InstituteMashhad University of Medical SciencesMashhadIran
| | | | | |
Collapse
|
122
|
Riva A, Gray EH, Azarian S, Zamalloa A, McPhail MJ, Vincent RP, Williams R, Chokshi S, Patel VC, Edwards LA. Faecal cytokine profiling as a marker of intestinal inflammation in acutely decompensated cirrhosis. JHEP Rep 2020; 2:100151. [PMID: 32838247 PMCID: PMC7391986 DOI: 10.1016/j.jhepr.2020.100151] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/26/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND & AIMS Gut dysbiosis and inflammation perpetuate loss of gut barrier integrity (GBI) and pathological bacterial translocation (BT) in cirrhosis, contributing to infection risk. Little is known about gut inflammation in cirrhosis and how this differs in acute decompensation (AD). We developed a novel approach to characterise intestinal immunopathology by quantifying faecal cytokines (FCs) and GBI markers. METHODS Faeces and plasma were obtained from patients with stable cirrhosis (SC; n = 16), AD (n = 47), and healthy controls (HCs; n = 31). A panel of 15 cytokines and GBI markers, including intestinal fatty-acid-binding protein-2 (FABP2), d-lactate, and faecal calprotectin (FCAL), were quantified by electrochemiluminescence/ELISA. Correlations between analytes and clinical metadata with univariate and multivariate analyses were performed. RESULTS Faecal (F) IL-1β, interferon gamma, tumour necrosis factor alpha, IL-21, IL-17A/F, and IL-22 were significantly elevated in AD vs. SC (q <0.01). F-IL-23 was significantly elevated in AD vs. HC (p = 0.0007). FABP2/d-lactate were significantly increased in faeces in AD vs. SC and AD vs. HC (p <0.0001) and in plasma (p = 0.0004; p = 0.011). F-FABP2 correlated most strongly with disease severity (Spearman's rho: Child-Pugh 0.466; p <0.0001; model for end-stage liver disease 0.488; p <0.0001). FCAL correlated with plasma IL-21, IL-1β, and IL-17F only and none of the faecal analytes. F-cytokines and F-GBI markers were more accurate than plasma in discriminating AD from SC. CONCLUSIONS FC profiling represents an innovative approach to investigating the localised intestinal cytokine micro-environment in cirrhosis. These data reveal that AD is associated with a highly inflamed and permeable gut barrier. FC profiles are very different from the classical innate-like features of systemic inflammation. There is non-specific upregulation of TH1/TH17 effector cytokines and those known to mediate intestinal barrier damage. This prevents mucosal healing in AD and further propagates BT and systemic inflammation. LAY SUMMARY The gut barrier is crucial in cirrhosis in preventing infection-causing bacteria that normally live in the gut from accessing the liver and other organs via the bloodstream. Herein, we characterised gut inflammation by measuring different markers in stool samples from patients at different stages of cirrhosis and comparing this to healthy people. These markers, when compared with equivalent markers usually measured in blood, were found to be very different in pattern and absolute levels, suggesting that there is significant gut inflammation in cirrhosis related to different immune system pathways to that seen outside of the gut. This provides new insights into gut-specific immune disturbances that predispose to complications of cirrhosis, and emphasises that a better understanding of the gut-liver axis is necessary to develop better targeted therapies.
Collapse
Key Words
- ACLF, acute-on-chronic liver failure
- AD, acute decompensation
- AUROC, area under the receiver operating characteristic
- BT, bacterial translocation
- Bacterial translocation
- CLIF-C AD, Chronic Liver Failure Consortium-acute decompensation
- Chronic liver disease
- Cytokines
- DS, discriminant score
- FABP2, fatty-acid-binding protein-2
- FCAL, faecal calprotectin
- FDR, false discovery rate
- FL, faecal lysate
- FWER, family-wise error rate
- GVB, gut vascular barrier
- Gut inflammation
- HC, healthy control
- IBD, inflammatory bowel disease
- IEC, intestinal epithelial cell
- Intestinal barrier function
- MELD, model for end-stage liver disease
- OPLS-DA, orthogonal projection to latent structures discriminant analysis
- PAMP, pathogen-associated molecular pattern
- PCA, principal component analysis
- ROC, receiver operating characteristic
- SC, stable cirrhosis
- UKELD, United Kingdom model for end-stage liver disease
Collapse
Affiliation(s)
- Antonio Riva
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Elizabeth H. Gray
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Sarah Azarian
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Ane Zamalloa
- Institute of Liver Studies, King's College Hospital NHS Foundation Trust, London, UK
| | - Mark J.W. McPhail
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
- Institute of Liver Studies, King's College Hospital NHS Foundation Trust, London, UK
| | - Royce P. Vincent
- Department of Clinical Biochemistry, King's College Hospital NHS Foundation Trust, London, UK
- Department of Nutritional Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Roger Williams
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Shilpa Chokshi
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Vishal C. Patel
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
- Institute of Liver Studies, King's College Hospital NHS Foundation Trust, London, UK
| | - Lindsey A. Edwards
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
123
|
Bilal M, Si W, Barbe F, Chevaux E, Sienkiewicz O, Zhao X. Effects of novel probiotic strains of Bacillus pumilus and Bacillus subtilis on production, gut health, and immunity of broiler chickens raised under suboptimal conditions. Poult Sci 2020; 100:100871. [PMID: 33516480 PMCID: PMC7936155 DOI: 10.1016/j.psj.2020.11.048] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/26/2020] [Accepted: 11/23/2020] [Indexed: 12/29/2022] Open
Abstract
Probiotics are being developed as alternatives to antibiotic growth promoters. The aim of the study was to investigate the effects of 2 novel strains of Bacillus pumilus and Bacillus subtilis on production, intestinal microbiota, gut health, and immunity of broilers raised under suboptimal conditions. Day-old chicks (Cobb 500, n = 2,073) were randomly assigned into 6 groups: Con group (group fed with basal diet), Ab group (group treated with virginiamycin), groups treated with 2 levels of B. pumilus (low dose: 3 × 108 cfu/kg of feed [BPL] and high dose: 1 × 109 cfu/kg [BPH]), and groups treated with 2 levels of B. subtilis (low dose: 3 × 108 cfu/kg [BSL] and high dose: 1 × 109 cfu/kg [BSH]). Production parameters were recorded weekly. Cecal tonsils and content as well as ileum samples were collected on day 14 and day 42. Cecal tonsils were used to sort T-regulatory cells (CD4+CD8–CD25+ and CD4+CD8+CD25+) to study expression of IL-10 and interferon gamma, whereas cecal content was used for bacterial culture. Ileum samples were used to measure gene expression of tight junction proteins, mucin, and cytokines. BW and feed intake increased in the Ab, BPL, BSL, and BSH groups compared with the Con group between day 35 and day 42. The CD4+CD8-CD25+ cells expressed high levels of IL-10 in the BSH group on day 14 and in the BPL, BSL, and BSH groups on day 42 and high levels of interferon gamma in the BPL, BSL, and BSH groups on day 14 and in the BSL and BSH groups on day 42. The expression of IL-10 and interferon gamma in CD4+CD8+CD25+ cells was higher only in the BSH group on day 14 and day 42. Cecal bacterial populations of genera, Lactobacillus (day 14 and day 42) and Clostridium (day 14), were higher in the BSH group. Expression of tight junction protein increased significantly in the ileum on day 14 in the BPL (occludin, zona occludens 1 [ZO-1]), BSL (occludin, ZO-1), and BSH (occludin, ZO-1, junctional adhesion molecule 2 [JAM-2]) groups compared with that in the Con group and declined in all groups except in the BSH group (occludin, ZO-1, JAM-2) on day 42. Expression of MUC2 and IL-17F increased in all groups on day 14 and remained high on day 42 in the BSL and BSH groups. Taken together, both Bacillus probiotics altered the intestinal and immune activities, particularly on day 14, suggesting beneficial influence of probiotics.
Collapse
Affiliation(s)
- Muhammad Bilal
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
| | - Wei Si
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
| | - Florence Barbe
- Research and Development Department, Lallemand Animal Nutrition, Blagnac, France
| | - Eric Chevaux
- Research and Development Department, Lallemand Animal Nutrition, Blagnac, France
| | - Olimpia Sienkiewicz
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
| | - Xin Zhao
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada.
| |
Collapse
|
124
|
Zhao N, Dong W, Kim H, Moallemian R, Lv J, Wang H, Zheng H, Wei F, Ma X. Capping protein regulator and myosin 1 linker 3 regulates transcription of key cytokines in activated phagocytic cells. Cell Signal 2020; 78:109848. [PMID: 33246003 DOI: 10.1016/j.cellsig.2020.109848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 11/19/2020] [Accepted: 11/22/2020] [Indexed: 01/02/2023]
Abstract
We have recently reported that capping protein regulator and myosin 1 linker 3 (CARMIL3), first identified as an oncofetal-like gene, is required for metastasis of breast and prostate cancer cells via regulating the actin cytoskeletal dynamics near the plasma membrane. Here, we demonstrate a novel function of CARMIL3 as an essential regulator of the transcription of several key proinflammatory cytokines in macrophages engulfing apoptotic cells and/or exposed to lipopolysaccharides (LPS). CARMIL3-deficient macrophages expressed strongly abrogated levels of interleukin (IL)-6, TNF-α, IL-1β and IL-23 in response to LPS, whereas IL-10 expression was enhanced. An RNA-seq analysis of CARMIL3-deficient and wild-type (WT) RAW264.7 cells stimulated with LPS revealed many differentially expressed genes, impacting several important inflammatory pathways. At the molecular level, CARMIL3 deficiency caused a strong impairment in LPS-activated nuclear factor-κB (NF-κB) signaling with decreased IKKα/β and IκBα phosphorylation and severely reduced p65 protein levels. This study uncovers a crucial role of CARMIL3 in impacting the balance between inflammation and tissue homeostasis via regulating major cytokines production in phagocytic cells.
Collapse
Affiliation(s)
- Na Zhao
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wenjuan Dong
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Hajeong Kim
- Department of Physiology, Kyungpook National University School of Medicine, Daegu 41944, Republic of Korea
| | - Rezvan Moallemian
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiyang Lv
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Huan Wang
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hua Zheng
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fang Wei
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Xiaojing Ma
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
125
|
Papathemeli D, Patsatsi A, Papanastassiou D, Koletsa T, Papathemelis T, Avgeros C, Pikou O, Lazaridou E, Georgiou E. Protein and mRNA Expression Levels of Interleukin-17A, -17F and -22 in Blood and Skin Samples of Patients with Mycosis Fungoides. Acta Derm Venereol 2020; 100:adv00326. [PMID: 33170303 PMCID: PMC9309846 DOI: 10.2340/00015555-3688] [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] [Accepted: 11/04/2020] [Indexed: 11/16/2022] Open
Abstract
This study investigated the expression of interleukin (IL)-17A, -17F and -22 in mycosis fungoides. Blood samples were collected from 50 patients with mycosis fungoides and 50 healthy controls. Skin samples were obtained from 26 patients with mycosis fungoides and 5 healthy controls. Protein levels of IL-17A, -17F and -22 were measured in serum by multiplex enzyme-linked immunosorbent assay, and mRNA expression levels were measured in blood and skin samples by real-time quantitative reverse transcription PCR. Both IL-17A and IL-17F mRNA expression levels were significantly lower in blood of patients with mycosis fungoides in comparison with healthy controls. IL-22 serum levels and expression levels of IL-22 mRNA in skin tissue, were significantly increased in patients with mycosis fungoides in comparison with healthy controls. These results suggest that low levels of IL-17A and IL-17F in mycosis fungoides may be connected to impaired immune surveillance contributing to tumourigenesis. Upregulation of IL-22 may play a role in the establishment of the tumour microenvironment in mycosis fungoides.
Collapse
Affiliation(s)
- Despoina Papathemeli
- 2nd Department of Dermatology and Venereology, Cutaneous Lymphoma Outpatient Clinic, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
126
|
Cerboni S, Gehrmann U, Preite S, Mitra S. Cytokine-regulated Th17 plasticity in human health and diseases. Immunology 2020; 163:3-18. [PMID: 33064842 DOI: 10.1111/imm.13280] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/30/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023] Open
Abstract
Upon activation, naïve CD4+ T helper (Th) cells differentiate into distinct Th effector cell lineages depending on the local cytokine environment. However, these polarized Th cells can also adapt their function and phenotype depending on the changing cytokine environment, demonstrating functional plasticity. Here, Th17 cells, which play a critical role in host protection from extracellular pathogens and in autoimmune disorders, are of particular interest. While being able to shift phenotype within their lineage, Th17 cells can also acquire characteristics of Th1, Th2, T follicular helper (Tfh) or regulatory T cells. Th17 cell identity is determined by a spectrum of extracellular signals, including cytokines, which are critical orchestrators of cellular immune responses. Cytokine induces changes in epigenetic, transcriptional, translational and metabolomic parameters. How these signals are integrated to determine Th17 plasticity is not well defined, yet this is a crucial point of investigation as it represents a potential target to treat autoimmune and inflammatory diseases. The goal of this review was to discuss how cytokines regulate intracellular networks, focusing on the regulation of lineage-specific transcription factors, chromatin remodelling and metabolism, to control human Th17 cell plasticity. We discuss the importance of Th17 plasticity in autoimmunity and cancer and present current strategies and challenges in targeting pathogenic Th17 cells with cytokine-based approaches, considering human genetic variants associated with altered Th17 differentiation. Finally, we discuss how modulating Th17 plasticity rather than targeting the Th17 lineage as a whole might preserve its essential immune function while purging its adverse effects.
Collapse
Affiliation(s)
- Silvia Cerboni
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology (R&I, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ulf Gehrmann
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology (R&I, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Silvia Preite
- Bioscience, In vivo, Research and Early Development, Respiratory & Immunology (R&I, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Suman Mitra
- CNRS, INSERM, CHU Lille, Institut pour la Recherche contre le Cancer de Lille, UMR9020 - UMR-S 1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| |
Collapse
|
127
|
Kaushik A, Mahajan R, Karim A, Handa S, De D, Saikia B. Successful use of cyclosporine in epidermolysis bullosa pruriginosa. Dermatol Ther 2020; 33:e14489. [DOI: 10.1111/dth.14489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/10/2020] [Accepted: 10/26/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Akanksha Kaushik
- Department of Dermatology, Venereology and Leprology PGIMER Chandigarh India
| | - Rahul Mahajan
- Department of Dermatology, Venereology and Leprology PGIMER Chandigarh India
| | - Adil Karim
- Department of Immunopathology PGIMER Chandigarh India
| | - Sanjeev Handa
- Department of Dermatology, Venereology and Leprology PGIMER Chandigarh India
| | - Dipankar De
- Department of Dermatology, Venereology and Leprology PGIMER Chandigarh India
| | - Biman Saikia
- Department of Immunopathology PGIMER Chandigarh India
| |
Collapse
|
128
|
Zhang H, Yan X, Yang C, Zhan Q, Fu Y, Luo H, Luo H. Intrahepatic T helper 17 cells recruited by hepatitis B virus X antigen-activated hepatic stellate cells exacerbate the progression of chronic hepatitis B virus infection. J Viral Hepat 2020; 27:1138-1149. [PMID: 32559002 DOI: 10.1111/jvh.13352] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/18/2020] [Accepted: 05/18/2020] [Indexed: 02/06/2023]
Abstract
Immunopathological injury induced by persistent hepatitis B virus (HBV) infection contributes to the progression from chronic hepatitis B (CHB) to hepatic cirrhosis and hepatocellular carcinoma (HCC). Regulatory T cells (Tregs), CD4+ T helper (Th) cells, and hepatic stellate cells (HSCs) are considered to be the pivotal factors during this progression. In this study, our aim was to investigate the molecular mechanisms of liver immunopathological injury associated with Tregs, CD4+ Th cells, and HSCs. Liver tissues were collected to assay the cytokines and distribution and frequencies of CD4+ Th cells and Tregs. The chemotaxis of Th17 cells towards the liver and the interactions between IL-22, IL-17A, and HSCs were explored. The data showed the frequencies of Th17 cells, and their effector molecules IL-22 and IL-17A were increased along with the severity of chronic liver diseases. However, the frequencies of Tregs were decreased in HBV-associated cirrhotic tissues compared with those in CHB tissues and HCC tissues. hepatitis B virus X antigen (HBxAg)-activated HSCs recruited more Th17 cells into the liver and conduced to the secretion of IL-17A and IL-22 that could in turn stimulate the proliferation and fibrotic marker secretion of the HSCs. Therefore, we suggest that the interactions between Th17 cells, IL-17A, IL-22, and HSCs form a positive feedback loop that aggravated the progression of chronic liver disease with HBV infection through the phosphoinositide-3-kinase/protein kinase B (PI3K/AKT) signalling pathway. Our findings indicated the IL-17A/IL-22 pathway might become a new treatment target for liver cirrhosis and HCC.
Collapse
Affiliation(s)
- Hongbin Zhang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiong Yan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Cheng Yang
- Department of Infectious Diseases, Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Zhan
- The Center for Clinical Molecular Medical detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yueqiang Fu
- Department of Critical Care Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Huating Luo
- Department of Infectious Diseases, Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongchun Luo
- Department of Infectious Diseases, Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
129
|
Breda LCD, Breda CNDS, de Almeida JRF, Paulo LNM, Jannuzzi GP, Menezes IDG, Albuquerque RC, Câmara NOS, Ferreira KS, de Almeida SR. Fonsecaeapedrosoi Conidia and Hyphae Activate Neutrophils Distinctly: Requirement of TLR-2 and TLR-4 in Neutrophil Effector Functions. Front Immunol 2020; 11:540064. [PMID: 33193308 PMCID: PMC7609859 DOI: 10.3389/fimmu.2020.540064] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 08/31/2020] [Indexed: 12/16/2022] Open
Abstract
Chromoblastomycosis is a chronic and progressive subcutaneous mycosis caused mainly by the fungus Fonsecaea pedrosoi. The infection is characterized by erythematous papules and histological sections demonstrating an external layer of fibrous tissue and an internal layer of thick granulomatous inflammatory tissue containing mainly macrophages and neutrophils. Several groups are studying the roles of the innate and adaptive immune systems in F. pedrosoi infection; however, few studies have focused on the role of neutrophils in this infection. In the current study, we verify the importance of murine neutrophils in the killing of F. pedrosoi conidia and hyphae. We demonstrate that phagocytosis and reactive oxygen species during infection with conidia are TLR-2- and TLR-4-dependent and are essential for conidial killing. Meanwhile, hyphal killing occurs by NET formation in a TLR-2-, TLR-4-, and ROS-independent manner. In vivo experiments show that TLR-2 and TLR-4 are also important in chromoblastomycosis infection. TLR-2KO and TLR-4KO animals had lower levels of CCL3 and CXCL1 chemokines and impaired neutrophil migration to the infected site. These animals also had higher fungal loads during infection with F. pedrosoi conidia, confirming that TLR-2 and TLR-4 are essential receptors for F. pedrosoi recognition and immune system activation. Therefore, this study demonstrates for the first time that neutrophil activation during F. pedrosoi is conidial or hyphal-specific with TLR-2 and TLR-4 being essential during conidial infection but unnecessary for hyphal killing by neutrophils.
Collapse
Affiliation(s)
- Leandro Carvalho Dantas Breda
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | | | - José Roberto Fogaça de Almeida
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Larissa Neves Monteiro Paulo
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Grasielle Pereira Jannuzzi
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Isabela de Godoy Menezes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Renata Chaves Albuquerque
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Niels Olsen Saraiva Câmara
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Karen Spadari Ferreira
- Departamento de Ciências Farmacêuticas, Instituto de Ciências Naturais, Universidade Federal de São Paulo, Diadema, Brazil
| | - Sandro Rogério de Almeida
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
130
|
Weatherhead JE, Gazzinelli-Guimaraes P, Knight JM, Fujiwara R, Hotez PJ, Bottazzi ME, Corry DB. Host Immunity and Inflammation to Pulmonary Helminth Infections. Front Immunol 2020; 11:594520. [PMID: 33193446 PMCID: PMC7606285 DOI: 10.3389/fimmu.2020.594520] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/30/2020] [Indexed: 01/04/2023] Open
Abstract
Helminths, including nematodes, cestodes and trematodes, are complex parasitic organisms that infect at least one billion people globally living in extreme poverty. Helminthic infections are associated with severe morbidity particularly in young children who often harbor the highest burden of disease. While each helminth species completes a distinct life cycle within the host, several helminths incite significant lung disease. This impact on the lungs occurs either directly from larval migration and host immune activation or indirectly from a systemic inflammatory immune response. The impact of helminths on the pulmonary immune response involves a sophisticated orchestration and activation of the host innate and adaptive immune cells. The consequences of activating pulmonary host immune responses are variable with several helminthic infections leading to severe, pulmonary compromise while others providing immune tolerance and protection against the development of pulmonary diseases. Further delineation of the convoluted interface between helminth infection and the pulmonary host immune responses is critical to the development of novel therapeutics that are critically needed to prevent the significant global morbidity caused by these parasites.
Collapse
Affiliation(s)
- Jill E. Weatherhead
- Department of Medicine, Infectious Diseases, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, Pediatric Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | | | - John M. Knight
- Department of Medicine, Pathology and Immunology, and the Biology of Inflammation Center, Baylor College of Medicine, Houston, TX, United States
| | - Ricardo Fujiwara
- Departamento de Parasitologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Peter J. Hotez
- Department of Pediatrics, Pediatric Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Center for Vaccine Development, Houston, TX, United States
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
- Department of Biology, Baylor University, Waco, TX, United States
- Hagler Institute for Advanced Study at Texas A&M University, College State, TX, United States
| | - Maria Elena Bottazzi
- Department of Pediatrics, Pediatric Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Center for Vaccine Development, Houston, TX, United States
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - David B. Corry
- Department of Medicine, Pathology and Immunology, and the Biology of Inflammation Center, Baylor College of Medicine, Houston, TX, United States
- Department of Medicine, Immunology, Allergy, Rheumatology, Baylor College of Medicine, Houston, TX, United States
- Michael E. DeBakey VA Center for Translational Research in Inflammatory Diseases, Houston, TX, United States
| |
Collapse
|
131
|
Azmy Nabeh O, Ishak Attallah M, El-Sayed El-Gawhary N. The pivotal relation between glucagon-like peptides, NFκB and inflammatory bowel disease. Clin Exp Pharmacol Physiol 2020; 47:1641-1648. [PMID: 32511781 DOI: 10.1111/1440-1681.13361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/21/2020] [Accepted: 06/01/2020] [Indexed: 11/30/2022]
Abstract
Glucagon-like peptides (GLPs), GLP-1 and GLP-2, are released from intestinal enteroendocrine cells (L cells) in response to ingested nutrients. GLP-1 plays a crucial role in lowering blood glucose and controlling body weight, through stimulating the islet ß cells of pancreas to secrete insulin, inhibiting gastric emptying, and reducing food ingestion. Therefore, GLP-1 receptor agonists are now used in the treatment of obese patients with type 2 diabetes mellitus (T2DM). GLP-2, on the other hand, is used as a novel therapy for short bowel syndrome (SBS) through its ability to restore intestinal homeostasis and induce epithelial proliferation. GLPs and the inhibitors of their degradation enzymes, dipeptidyl peptidase-IV (DPP-IV) inhibitors, have many anti-inflammatory actions. Many animal-based clinical trials have proved that GLP-based therapy has a pivotal role in the management of inflammatory bowel disease (IBD), possibly through regulating the transcription factor nuclear factor kappa-ligand B (NFκB). NFκB controls the production and secretion of many cytokines and chemokines encountered in the pathophysiology of IBD such as interleukin (IL-1β-IL-12, IL-13, IL-21, IL-22, IL-6) and tumour necrosis factor-alpha (TNF-α) and hence, may provide a promising therapeutic option.
Collapse
Affiliation(s)
- Omnia Azmy Nabeh
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Magdy Ishak Attallah
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | |
Collapse
|
132
|
Type I and Type III Interferons Restrict SARS-CoV-2 Infection of Human Airway Epithelial Cultures. J Virol 2020; 94:JVI.00985-20. [PMID: 32699094 DOI: 10.1128/jvi.00985-20] [Citation(s) in RCA: 229] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/17/2020] [Indexed: 01/08/2023] Open
Abstract
The newly emerged human coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has caused a pandemic of respiratory illness. Current evidence suggests that severe cases of SARS-CoV-2 are associated with a dysregulated immune response. However, little is known about how the innate immune system responds to SARS-CoV-2. In this study, we modeled SARS-CoV-2 infection using primary human airway epithelial (pHAE) cultures, which are maintained in an air-liquid interface. We found that SARS-CoV-2 infects and replicates in pHAE cultures and is directionally released on the apical, but not basolateral, surface. Transcriptional profiling studies found that infected pHAE cultures had a molecular signature dominated by proinflammatory cytokines and chemokine induction, including interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α), and CXCL8, and identified NF-κB and ATF-4 as key drivers of this proinflammatory cytokine response. Surprisingly, we observed a complete lack of a type I or III interferon (IFN) response to SARS-CoV-2 infection. However, pretreatment and posttreatment with type I and III IFNs significantly reduced virus replication in pHAE cultures that correlated with upregulation of antiviral effector genes. Combined, our findings demonstrate that SARS-CoV-2 does not trigger an IFN response but is sensitive to the effects of type I and III IFNs. Our studies demonstrate the utility of pHAE cultures to model SARS-CoV-2 infection and that both type I and III IFNs can serve as therapeutic options to treat COVID-19 patients.IMPORTANCE The current pandemic of respiratory illness, COVID-19, is caused by a recently emerged coronavirus named SARS-CoV-2. This virus infects airway and lung cells causing fever, dry cough, and shortness of breath. Severe cases of COVID-19 can result in lung damage, low blood oxygen levels, and even death. As there are currently no vaccines approved for use in humans, studies of the mechanisms of SARS-CoV-2 infection are urgently needed. Our research identifies an excellent system to model SARS-CoV-2 infection of the human airways that can be used to test various treatments. Analysis of infection in this model system found that human airway epithelial cell cultures induce a strong proinflammatory cytokine response yet block the production of type I and III IFNs to SARS-CoV-2. However, treatment of airway cultures with the immune molecules type I or type III interferon (IFN) was able to inhibit SARS-CoV-2 infection. Thus, our model system identified type I or type III IFN as potential antiviral treatments for COVID-19 patients.
Collapse
|
133
|
Macrophages in pancreatitis: Mechanisms and therapeutic potential. Biomed Pharmacother 2020; 131:110693. [PMID: 32882586 DOI: 10.1016/j.biopha.2020.110693] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023] Open
Abstract
Macrophages play a crucial role in the pathogenesis of pancreatitis that is a common gastrointestinal disease. Particularly, macrophages differentiate into different phenotypes and exert diverse functions in acute pancreatitis (AP) and chronic pancreatitis (CP), respectively. In AP, macrophages in the pancreas and other related organs are mainly activated and differentiated into a pro-inflammatory M1 phenotype, and furthermore secrete inflammatory cytokines and mediators, causing local inflammation of the pancreas, and even intractable systemic inflammatory response or multiple organ failure. In CP, macrophages often exhibit a M2 polarisation and interact with pancreatic stellate cells (PSCs) in an autocrine and paracrine cytokine-dependent manner to promote the progression of pancreatic fibrosis. As the severity of pancreatic fibrosis aggravates, the proportion of M2/M1 macrophage cytokines in the pancreas increases. The discovery of macrophages in the pathogenesis of pancreatitis has promoted the research of targeted drugs, which provides great potential for the effective treatment of pancreatitis. This paper provides an overview of the roles of various macrophages in the pathogenesis of pancreatitis and the current research status of pancreatitis immunotherapy targeting macrophages. The findings addressed in this review are of considerable significance for understanding the pivotal role of macrophages in pancreatitis.
Collapse
|
134
|
Sun YX, Jiang XJ, Lu B, Gao Q, Chen YF, Wu DB, Zeng WY, Yang L, Li HH, Yu B. Roles of Gut Microbiota in Pathogenesis of Alzheimer's Disease and Therapeutic Effects of Chinese Medicine. Chin J Integr Med 2020; 28:1048-1056. [PMID: 32876860 DOI: 10.1007/s11655-020-3274-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2020] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease characterized by progressive cognitive impairment. The pathogenesis of AD is complex, and its susceptibility and development process are affected by age, genetic and epigenetic factors. Recent studies confirmed that gut microbiota (GM) might contribute to AD through a variety of pathways including hypothalamic pituitary adrenal axis and inflflammatory and immune processes. CM formula, herbs, and monomer enjoy unique advantages to treat and prevent AD. Hence, the purpose of this review is to outline the roles of GM and its core metabolites in the pathogenesis of AD. Research progress of CMs regarding the mechanisms of how they regulate GM to improve cognitive impairment of AD is also reviewed. The authors tried to explore new therapeutic strategies to AD based on the regulation of GM using CM.
Collapse
Affiliation(s)
- Ying-Xin Sun
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xi-Juan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Bin Lu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Qing Gao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ye-Fei Chen
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Dan-Bin Wu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Wen-Yun Zeng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lin Yang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Hu-Hu Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Bin Yu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
135
|
Wu J, Wei Z, Cheng P, Qian C, Xu F, Yang Y, Wang A, Chen W, Sun Z, Lu Y. Rhein modulates host purine metabolism in intestine through gut microbiota and ameliorates experimental colitis. Am J Cancer Res 2020; 10:10665-10679. [PMID: 32929373 PMCID: PMC7482825 DOI: 10.7150/thno.43528] [Citation(s) in RCA: 217] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 08/14/2020] [Indexed: 12/21/2022] Open
Abstract
Background: Gut microbiota, which plays a crucial role in inflammatory bowel diseases (IBD), might have therapeutic benefits for ulcerative colitis or Crohn's disease. Targeting gut microbiota represents a new treatment strategy for IBD patients. Rhein is one of the main components of rhubarb and exhibits poor oral bioavailability but still exerts anti-inflammatory effects in some diseases. Therefore, we investigated the effect of rhein on colitis and studied its possible mechanisms. Methods: The chronic mouse colitis model was induced by four rounds of 2% dextran sulfate sodium (DSS) treatment. The mice were treated with 50 mg/kg and 100 mg/kg rhein daily, body weight, colon length, histological score, inflammatory cytokines in serum or intestine, and fecal lipocalin 2 concentration were determined. Th17 cell, Th1 cell and Th2 cell infiltration in the mesenteric lymph node were analyzed by flow cytometry. Metabolic profiles were collected by non-targeted metabolomics and key metabolic pathways were identified using MetaboAnalyst 4.0. We also assessed intestinal barrier permeability and performed 16s rDNA sequencing. Lactobacillus sp. was cultured, and fecal microbiota transplantation (FMT) was employed to evaluate the contribution of gut microbiota. Results: Rhein could significantly alleviate DSS-induced chronic colitis. Uric acid was identified as a crucial modulator of colitis and rhein treatment led to decreased uric acid levels. We determined that rhein changed purine metabolism indirectly, while the probiotic Lactobacillus was involved in the regulation of host metabolism. Uric acid resulted in a worsened intestinal barrier, which could be rescued by rhein. We further confirmed that rhein-treated gut microbiota was sufficient to relieve DSS-induced colitis by FMT. Conclusion: We showed that rhein could modulate gut microbiota, which indirectly changed purine metabolism in the intestine and subsequently alleviated colitis. Our study has identified a new approach to the clinical treatment of colitis.
Collapse
|
136
|
Watanabe Costa R, Batista MF, Meneghelli I, Vidal RO, Nájera CA, Mendes AC, Andrade-Lima IA, da Silveira JF, Lopes LR, Ferreira LRP, Antoneli F, Bahia D. Comparative Analysis of the Secretome and Interactome of Trypanosoma cruzi and Trypanosoma rangeli Reveals Species Specific Immune Response Modulating Proteins. Front Immunol 2020; 11:1774. [PMID: 32973747 PMCID: PMC7481403 DOI: 10.3389/fimmu.2020.01774] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/02/2020] [Indexed: 12/04/2022] Open
Abstract
Chagas disease, a zoonosis caused by the flagellate protozoan Trypanosoma cruzi, is a chronic and systemic parasitic infection that affects ~5–7 million people worldwide, mainly in Latin America. Chagas disease is an emerging public health problem due to the lack of vaccines and effective treatments. According to recent studies, several T. cruzi secreted proteins interact with the human host during cell invasion. Moreover, some comparative studies with T. rangeli, which is non-pathogenic in humans, have been performed to identify proteins directly involved in the pathogenesis of the disease. In this study, we present an integrated analysis of canonical putative secreted proteins (PSPs) from both species. Additionally, we propose an interactome with human host and gene family clusters, and a phylogenetic inference of a selected protein. In total, we identified 322 exclusively PSPs in T. cruzi and 202 in T. rangeli. Among the PSPs identified in T. cruzi, we found several trans-sialidases, mucins, MASPs, proteins with phospholipase 2 domains (PLA2-like), and proteins with Hsp70 domains (Hsp70-like) which have been previously characterized and demonstrated to be related to T. cruzi virulence. PSPs found in T. rangeli were related to protozoan metabolism, specifically carboxylases and phosphatases. Furthermore, we also identified PSPs that may interact with the human immune system, including heat shock and MASP proteins, but in a lower number compared to T. cruzi. Interestingly, we describe a hypothetical hybrid interactome of PSPs which reveals that T. cruzi secreted molecules may be down-regulating IL-17 whilst T. rangeli may enhance the production of IL-15. These results will pave the way for a better understanding of the pathophysiology of Chagas disease and may ultimately lead to the identification of molecular targets, such as key PSPs, that could be used to minimize the health outcomes of Chagas disease by modulating the immune response triggered by T. cruzi infection.
Collapse
Affiliation(s)
- Renata Watanabe Costa
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marina Ferreira Batista
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabela Meneghelli
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ramon Oliveira Vidal
- The Berlin Institute for Medical Systems Biology-Max Delbrück Center for Molecular Medicine in the Helmholtz Association in Berlin, Berlin, Germany.,Laboratorio Nacional de Biociências (LNBio), Campinas, São Paulo, Brazil
| | - Carlos Alcides Nájera
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Clara Mendes
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Izabela Augusta Andrade-Lima
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - José Franco da Silveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Luciano Rodrigo Lopes
- Departamento de Informática em Saúde, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ludmila Rodrigues Pinto Ferreira
- RNA Systems Biology Lab (RSBL), Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernando Antoneli
- Departamento de Informática em Saúde, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Diana Bahia
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil.,Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
137
|
Govers C, Tang Y, Stolte EH, Wichers HJ, Mes JJ. Wheat-derived arabinoxylans reduced M2-macrophage functional activity, but enhanced monocyte-recruitment capacity. Food Funct 2020; 11:7073-7083. [PMID: 32725025 DOI: 10.1039/d0fo00316f] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The immunomodulatory properties of non-digestible polysaccharides (NDPs) have been recognized in in vitro and in vivo studies. The latter mostly demonstrated altered frequencies and inflammatory status of immune cells as clinical parameters. Most of the NDP activity will be exerted in the intestine where they can directly interact with macrophages. The predominant macrophage phenotype in the intestine is M2-like, with M1-like macrophages arising during inflammation. Here, we investigated transcriptional and functional impact on these macrophage phenotypes by NDP-treatment (i.e. yeast-derived soluble β-glucan (yeast-βG), apple-derived RG-I (apple-RGI), shiitake-derived β-glucan (shiitake-βG) or wheat-derived arabinoxylan (wheat-AX)). Wheat-AX, and to a lesser extent shiitake-βG and apple-RGI but not yeast-βG, reduced endocytosis and antigen processing capacity of M1- and M2-like macrophages. Moreover, the NDPs, and most notably wheat-AX, strongly induced transcription and secretion of a unique set of cytokines and chemokines. Conditioned medium from wheat-AX-treated M2-like macrophages subsequently demonstrated strongly increased monocyte recruitment capacity. These findings are in line with clinically observed immunomodulatory aspects of NDPs making it tempting to speculate that clinical activity of some NDPs is mediated through enhanced chemoattraction and modifying activity of intestinal immune cells.
Collapse
Affiliation(s)
- Coen Govers
- Wageningen - Food & Biobased Research, Wageningen University & Research, Wageningen, The Netherlands.
| | - Yongfu Tang
- Wageningen - Food & Biobased Research, Wageningen University & Research, Wageningen, The Netherlands. and Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, The Netherlands and Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, China
| | - Ellen H Stolte
- Host-Microbe Interactomics Group, Animal Sciences Department, Wageningen University & Research, Wageningen, The Netherlands
| | - Harry J Wichers
- Wageningen - Food & Biobased Research, Wageningen University & Research, Wageningen, The Netherlands. and Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, The Netherlands
| | - Jurriaan J Mes
- Wageningen - Food & Biobased Research, Wageningen University & Research, Wageningen, The Netherlands.
| |
Collapse
|
138
|
Kaipe H, Raffetseder J, Ernerudh J, Solders M, Tiblad E. MAIT Cells at the Fetal-Maternal Interface During Pregnancy. Front Immunol 2020; 11:1788. [PMID: 32973750 PMCID: PMC7466580 DOI: 10.3389/fimmu.2020.01788] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
One of the main functions of the human placenta is to provide a barrier between the fetal and maternal blood circulations, where gas exchange and transfer of nutrients to the developing fetus take place. Despite being a barrier, there is a multitude of crosstalk between maternal immune cells and fetally derived semi-allogeneic trophoblast cells. Therefore, the maternal immune system has a difficult task to both tolerate the fetus but at the same time also defend the mother and the fetus from infections. Mucosal-associated invariant T (MAIT) cells are an increasingly recognized subset of T cells with anti-microbial functions that get activated in the context of non-polymorphic MR1 molecules, but also in response to inflammation. MAIT cells accumulate at term pregnancy in the maternal blood that flows into the intervillous space inside the placenta. Chemotactic factors produced by the placenta may be involved in recruiting and retaining particular immune cell subsets, including MAIT cells. In this Mini-Review, we describe what is known about MAIT cells during pregnancy and discuss the potential biological functions of MAIT cells at the fetal-maternal interface. Since MAIT cells have anti-microbial and tissue-repairing functions, but lack alloantigen reactivity, they could play an important role in protecting the fetus from bacterial infections and maintaining tissue homeostasis without risks of mediating harmful responses toward semi-allogenic fetal tissues.
Collapse
Affiliation(s)
- Helen Kaipe
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Johanna Raffetseder
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Jan Ernerudh
- Department of Clinical Immunology and Transfusion Medicine, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Martin Solders
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Eleonor Tiblad
- Center for Fetal Medicine, Karolinska University Hospital, Stockholm, Sweden.,Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
139
|
Terpstra ML, Remmerswaal EBM, van der Bom-Baylon ND, Sinnige MJ, Kers J, van Aalderen MC, Geerlings SE, Bemelman FJ. Tissue-resident mucosal-associated invariant T (MAIT) cells in the human kidney represent a functionally distinct subset. Eur J Immunol 2020; 50:1783-1797. [PMID: 32652598 PMCID: PMC7689767 DOI: 10.1002/eji.202048644] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/24/2020] [Indexed: 12/13/2022]
Abstract
Mucosal‐associated invariant T (MAIT) cells are innate‐like T‐cells that recognize bacterial riboflavin metabolites. They are present in human blood but are abundant at barrier sites, including the liver, lungs, and kidneys, where they possess a CD69+/CD103+/− tissue‐resident phenotype. In renal tissue, MAIT cells likely defend against the ascending uropathogens responsible for urinary tract infections (UTIs), which are common, especially among renal transplant recipients (RTRs). Nevertheless, the functional role for MAIT cells in renal tissue and the influence of renal transplantation on MAIT cells remains unclear. Using multiparameter flow cytometry and the MR1‐tetramer, we characterized MAIT cell phenotype and function in healthy renal tissue (n = 6), renal transplants explanted after allograft failure (n = 14) and in blood from healthy controls (n = 20) and RTRs before and 1‐year after transplantation (n = 21). MAIT cells in renal tissue constitute a distinct CD69+CD103+/− population that displays typical phenotypic features of tissue‐resident T‐cells and is skewed toward IL‐2, GM‐CSF, and IL‐17A production upon stimulation. The circulating MAIT cell population was not decreased in number in RTRs pre‐ or post‐transplantation. Tissue‐resident MAIT cells in the kidney represent a functionally distinct population. This shows how MAIT cells in the kidney may be involved in the protection against microorganisms.
Collapse
Affiliation(s)
- Matty L Terpstra
- Division of Internal Medicine, Department of Nephrology, Renal Transplant Unit, Amsterdam Infection & Immunity Institute (AI&II), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Experimental Immunology, Amsterdam Infection & Immunity Institute (AI&II), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Internal Medicine, Infectious Diseases, Amsterdam Infection & Immunity Institute (AI&II), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ester B M Remmerswaal
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute (AI&II), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nelly D van der Bom-Baylon
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute (AI&II), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marjan J Sinnige
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute (AI&II), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jesper Kers
- Department of Pathology, Amsterdam Infection & Immunity Institute (AI&II), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Center for Analytical Sciences Amsterdam (CASA)-Biomolecular Systems Analytics, Van't Hoff Institute for Molecular Sciences (HIMS), University of Amsterdam, Amsterdam, The Netherlands.,Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Michiel C van Aalderen
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute (AI&II), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Suzanne E Geerlings
- Department of Internal Medicine, Infectious Diseases, Amsterdam Infection & Immunity Institute (AI&II), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Frederike J Bemelman
- Division of Internal Medicine, Department of Nephrology, Renal Transplant Unit, Amsterdam Infection & Immunity Institute (AI&II), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
140
|
Cai Y, Xue F, Qin H, Chen X, Liu N, Fleming C, Hu X, Zhang HG, Chen F, Zheng J, Yan J. Differential Roles of the mTOR-STAT3 Signaling in Dermal γδ T Cell Effector Function in Skin Inflammation. Cell Rep 2020; 27:3034-3048.e5. [PMID: 31167146 PMCID: PMC6617524 DOI: 10.1016/j.celrep.2019.05.019] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/04/2019] [Accepted: 05/02/2019] [Indexed: 02/06/2023] Open
Abstract
Dermal γδT cells play critical roles in skin homeostasis and inflammation. However, the underlying molecular mechanisms by which these cells are activated have not been fully understood. Here, we show that the mechanistic or mammalian target of rapamycin (mTOR) and STAT3 pathways are activated in dermal γδT cells in response to innate stimuli such as interleukin-1β (IL-1β) and IL-23. Although both mTOR complex 1 (mTORC1) and mTORC2 are essential for dermal γδT cell proliferation, mTORC2 deficiency leads to decreased dermal γδT17 cells. It appears that mitochondria-mediated oxidative phosphorylation is critical in this process. Notably, although the STAT3 pathway is critical for dermal Vγ4T17 effector function, it is not required for γδ6T17 cells. Transcription factor IRF-4 activation promotes dermal γδT cell IL-17 production by linking IL-1β and IL-23 signaling. The absence of mTORC2 in dermal γδT cells, but not STAT3, ameliorates skin inflammation. Taken together, our results demonstrate that the mTOR-STAT3 signaling differentially regulates dermal γδT cell effector function in skin inflammation. Cai et al. demonstrate that the mTOR and STAT3 signaling pathways differentially regulate dermal Vγ4 and Vγ6 T cell effector function, leading to distinct outcomes in skin inflammation.
Collapse
Affiliation(s)
- Yihua Cai
- Department of Medicine and Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Feng Xue
- Department of Dermatology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P.R. China
| | - Hui Qin
- Department of Medicine and Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA; Department of Dermatology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P.R. China
| | - Xu Chen
- Department of Medicine and Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA; Department of Laboratory Medicine, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Na Liu
- Department of Dermatology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P.R. China
| | - Chris Fleming
- Department of Medicine and Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Xiaoling Hu
- Department of Medicine and Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Huang-Ge Zhang
- Department of Medicine and Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Fuxiang Chen
- Department of Laboratory Medicine, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Jie Zheng
- Department of Dermatology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P.R. China
| | - Jun Yan
- Department of Medicine and Department of Microbiology and Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
141
|
Barrea L, Muscogiuri G, Frias-Toral E, Laudisio D, Pugliese G, Castellucci B, Garcia-Velasquez E, Savastano S, Colao A. Nutrition and immune system: from the Mediterranean diet to dietary supplementary through the microbiota. Crit Rev Food Sci Nutr 2020; 61:3066-3090. [PMID: 32691606 DOI: 10.1080/10408398.2020.1792826] [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] [Indexed: 12/16/2022]
Abstract
The interaction between nutrition and the immune system is very complex. In particular, at every stage of the immune response, specific micronutrients, including vitamins and minerals play a key role and often synergistic, and the deficiency of only one essential nutrient may impair immunity. An individual's overall nutrition status and pattern of dietary intake (comprised of nutrients and non-nutritive bioactive compounds and food) and any supplementation with nutraceuticals including vitamins and minerals, can influence positively or negatively the function of the immune system. This influence can occur at various levels from the innate immune system and adaptive immune system to the microbiome. Although there are conflicting evidence, the current results point out that dietary supplementation with some nutrients such as vitamin D and zinc may modulate immune function. An update on the complex relationship between nutrition, diet, and the immune system through gut microbiota is the aim of this current review. Indeed, we will provide the overview of the link among immune function, nutrition and gut microbiota, paying particular attention at the effect of the Mediterranean diet on the immune system, and finally we will speculate the possible role of the main one functional supplements on immune function.
Collapse
Affiliation(s)
- Luigi Barrea
- Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Naples, Italy.,Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy
| | - Giovanna Muscogiuri
- Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Naples, Italy.,Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy
| | | | - Daniela Laudisio
- Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Naples, Italy.,Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy
| | - Gabriella Pugliese
- Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Naples, Italy.,Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy
| | - Bianca Castellucci
- Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Naples, Italy.,Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy
| | | | - Silvia Savastano
- Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Naples, Italy.,Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Naples, Italy.,Centro Italiano per la cura e il Benessere del paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Naples, Italy.,Cattedra Unesco "Educazione alla salute e allo sviluppo sostenibile," University Federico II, Naples, Italy
| |
Collapse
|
142
|
Ahlawat S, Asha, Sharma KK. Gut-organ axis: a microbial outreach and networking. Lett Appl Microbiol 2020; 72:636-668. [PMID: 32472555 DOI: 10.1111/lam.13333] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 05/05/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022]
Abstract
Human gut microbiota (GM) includes a complex and dynamic population of microorganisms that are crucial for well-being and survival of the organism. It has been reported as diverse and relatively stable with shared core microbiota, including Bacteroidetes and Firmicutes as the major dominants. They are the key regulators of body homeostasis, involving both intestinal and extra-intestinal effects by influencing many physiological functions such as metabolism, maintenance of barrier homeostasis, inflammation and hematopoiesis. Any alteration in GM community structures not only trigger gut disorders but also influence other organs and cause associated diseases. In recent past, the GM has been defined as a 'vital organ' with its involvement with other organs; thus, establishing a link or a bi- or multidirectional communication axis between the organs via neural, endocrine, immune, humoral and metabolic pathways. Alterations in GM have been linked to several diseases known to humans; although the exact interaction mechanism between the gut and the organs is yet to be defined. In this review, the bidirectional relationship between the gut and the vital human organs was envisaged and discussed under several headings. Furthermore, several disease symptoms were also revisited to redefine the communication network between the gut microbes and the associated organs.
Collapse
Affiliation(s)
- S Ahlawat
- Laboratory of Enzymology and Recombinant DNA Technology, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Asha
- Laboratory of Enzymology and Recombinant DNA Technology, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, India
| | - K K Sharma
- Laboratory of Enzymology and Recombinant DNA Technology, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, India
| |
Collapse
|
143
|
Xu H, Agalioti T, Zhao J, Steglich B, Wahib R, Vesely MCA, Bielecki P, Bailis W, Jackson R, Perez D, Izbicki J, Licona-Limón P, Kaartinen V, Geginat J, Esplugues E, Tolosa E, Huber S, Flavell RA, Gagliani N. The induction and function of the anti-inflammatory fate of T H17 cells. Nat Commun 2020; 11:3334. [PMID: 32620760 PMCID: PMC7335205 DOI: 10.1038/s41467-020-17097-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 06/11/2020] [Indexed: 01/19/2023] Open
Abstract
TH17 cells exemplify environmental immune adaptation: they can acquire both a pathogenic and an anti-inflammatory fate. However, it is not known whether the anti-inflammatory fate is merely a vestigial trait, or whether it serves to preserve the integrity of the host tissues. Here we show that the capacity of TH17 cells to acquire an anti-inflammatory fate is necessary to sustain immunological tolerance, yet it impairs immune protection against S. aureus. Additionally, we find that TGF-β signalling via Smad3/Smad4 is sufficient for the expression of the anti-inflammatory cytokine, IL-10, in TH17 cells. Our data thus indicate a key function of TH17 cell plasticity in maintaining immune homeostasis, and dissect the molecular mechanisms explaining the functional flexibility of TH17 cells with regard to environmental changes. CD4+ T helper cells producing IL-17A (TH17 cells) can take on pathogenic or anti-inflammatory functions in context-specific manners. Here the authors show that the anti-inflammatory fate of TH17 cells contributes, via TGF-β signaling and induction of IL-10, to host immune tolerance, but also simultaneously dampens protective immunity against S. aureus.
Collapse
Affiliation(s)
- Hao Xu
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, 06520, USA
| | - Theodora Agalioti
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Jun Zhao
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, 06520, USA
| | - Babett Steglich
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Ramez Wahib
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | | | - Piotr Bielecki
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, 06520, USA
| | - Will Bailis
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Ruaidhri Jackson
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, 06520, USA
| | - Daniel Perez
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Jakob Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Paula Licona-Limón
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, D.F, México
| | - Vesa Kaartinen
- Biologic and Material Sciences, University of Michigan, 1011N. University Ave, Ann Arbor, MI, 48109, USA
| | - Jens Geginat
- INGM-National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy.,Department of Clinical Sciences and Community Health, Università degli studi di Milano, Milan, Italy
| | - Enric Esplugues
- Laboratory of Molecular and Cellular Immunology, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - Eva Tolosa
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Richard A Flavell
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, 06520, USA. .,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA.
| | - Nicola Gagliani
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany. .,I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany. .,Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, Stockholm, Sweden.
| |
Collapse
|
144
|
Tonomura S, Ihara M, Friedland RP. Microbiota in cerebrovascular disease: A key player and future therapeutic target. J Cereb Blood Flow Metab 2020; 40:1368-1380. [PMID: 32312168 PMCID: PMC7308516 DOI: 10.1177/0271678x20918031] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Stroke is the second leading cause of death and a significant cause of disability worldwide. Recent advances in DNA sequencing, proteomics, metabolomics, and computational tools are dramatically increasing access to the identification of host-microbiota interactions in systemic diseases. In this review, we describe the accumulating evidence showing how human microbiota plays an essential role in cerebrovascular diseases. We introduce the symbiotic relationships between microbiota and the mucosal immune system, focusing on differences by anatomical sites. Microbiota directly or indirectly contributes to the pathogenesis of traditional vascular risk factors including age, obesity, diabetes mellitus, dyslipidemia, and hypertension. Moreover, recent studies proposed independent effects of the microbiome on the progression of various subtypes of stroke through direct microbial invasion, exotoxins, functional amyloids, inflammation, and microbe-derived metabolites. We propose the critical concept of gene-microbial interaction to elucidate the heterogeneity of stroke and provide possible therapeutic avenues. We suggest ways to resolve the vast inter-individual diversity of cerebrovascular disease and mechanisms for personalized prevention and treatment.
Collapse
Affiliation(s)
- Shuichi Tonomura
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan.,Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Robert P Friedland
- Department of Neurology, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
145
|
Inamura K. Gut microbiota contributes towards immunomodulation against cancer: New frontiers in precision cancer therapeutics. Semin Cancer Biol 2020; 70:11-23. [PMID: 32580023 DOI: 10.1016/j.semcancer.2020.06.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 02/08/2023]
Abstract
The microbiota influences human health and the development of diverse diseases, including cancer. Microbes can influence tumor initiation and development in either a positive or negative manner. In addition, the composition of the gut microbiota affects the efficacy and toxicity of cancer therapeutics as well as therapeutic resistance. The striking impact of microbiota on oncogenesis and cancer therapy provides compelling evidence to support the notion that manipulating microbial networks represents a promising strategy for treating and preventing cancer. Specific microbes or the microbial ecosystem can be modified via a multiplicity of processes, and therapeutic methods and approaches have been evolving. Microbial manipulation can be applied as an adjunct to traditional cancer therapies such as chemotherapy and immunotherapy. Furthermore, this approach displays great promise as a stand-alone therapy following the failure of standard therapy. Moreover, such strategies may also benefit patients by avoiding the emergence of toxic side effects that result in treatment discontinuation. A better understanding of the host-microbial ecosystem in patients with cancer, together with the development of methodologies for manipulating the microbiome, will help expand the frontiers of precision cancer therapeutics, thereby improving patient care. This review discusses the roles of the microbiota in oncogenesis and cancer therapy, with a focus on efforts to harness the microbiota to fight cancer.
Collapse
Affiliation(s)
- Kentaro Inamura
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan.
| |
Collapse
|
146
|
Abstract
The ketogenic diet is used to treat neurological and metabolic symptoms of disease, but the extent of its influences across organ systems remains unclear. Ang et al., 2020 reveal that ketone bodies induced by the diet inhibit specific bacteria of the gut microbiota and suppress pro-inflammatory T cells in the intestine.
Collapse
Affiliation(s)
- Christine A Olson
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gregory R Lum
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Elaine Y Hsiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
147
|
Kang J. Vitamin intervention for cytokine storm in the patients with coronavirus disease 2019. MedComm (Beijing) 2020; 1:81-83. [PMID: 34173637 PMCID: PMC7323139 DOI: 10.1002/mco2.7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/19/2020] [Accepted: 04/21/2020] [Indexed: 01/14/2023] Open
Affiliation(s)
- Jian‐Sheng Kang
- Clinical Systems Biology LaboratoriesThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
148
|
Iddir M, Brito A, Dingeo G, Fernandez Del Campo SS, Samouda H, La Frano MR, Bohn T. Strengthening the Immune System and Reducing Inflammation and Oxidative Stress through Diet and Nutrition: Considerations during the COVID-19 Crisis. Nutrients 2020; 12:E1562. [PMID: 32471251 PMCID: PMC7352291 DOI: 10.3390/nu12061562] [Citation(s) in RCA: 422] [Impact Index Per Article: 84.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/21/2020] [Accepted: 05/25/2020] [Indexed: 12/13/2022] Open
Abstract
The coronavirus-disease 2019 (COVID-19) was announced as a global pandemic by the World Health Organization. Challenges arise concerning how to optimally support the immune system in the general population, especially under self-confinement. An optimal immune response depends on an adequate diet and nutrition in order to keep infection at bay. For example, sufficient protein intake is crucial for optimal antibody production. Low micronutrient status, such as of vitamin A or zinc, has been associated with increased infection risk. Frequently, poor nutrient status is associated with inflammation and oxidative stress, which in turn can impact the immune system. Dietary constituents with especially high anti-inflammatory and antioxidant capacity include vitamin C, vitamin E, and phytochemicals such as carotenoids and polyphenols. Several of these can interact with transcription factors such as NF-kB and Nrf-2, related to anti-inflammatory and antioxidant effects, respectively. Vitamin D in particular may perturb viral cellular infection via interacting with cell entry receptors (angiotensin converting enzyme 2), ACE2. Dietary fiber, fermented by the gut microbiota into short-chain fatty acids, has also been shown to produce anti-inflammatory effects. In this review, we highlight the importance of an optimal status of relevant nutrients to effectively reduce inflammation and oxidative stress, thereby strengthening the immune system during the COVID-19 crisis.
Collapse
Affiliation(s)
- Mohammed Iddir
- Nutrition and Health Research Group, Population Health Department, Luxembourg Institute of Health, 1A-B, rue Thomas Edison, L-1445 Strassen, Luxembourg; (M.I.); or (A.B.); (S.S.F.D.C.); (H.S.)
| | - Alex Brito
- Nutrition and Health Research Group, Population Health Department, Luxembourg Institute of Health, 1A-B, rue Thomas Edison, L-1445 Strassen, Luxembourg; (M.I.); or (A.B.); (S.S.F.D.C.); (H.S.)
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology. I.M. Sechenov First Moscow Medical University, Trubetskay Str. 8, 119991 Moscow, Russia
| | - Giulia Dingeo
- Independent Researcher, Val de Marne, 94999 Paris, France;
| | - Sofia Sosa Fernandez Del Campo
- Nutrition and Health Research Group, Population Health Department, Luxembourg Institute of Health, 1A-B, rue Thomas Edison, L-1445 Strassen, Luxembourg; (M.I.); or (A.B.); (S.S.F.D.C.); (H.S.)
| | - Hanen Samouda
- Nutrition and Health Research Group, Population Health Department, Luxembourg Institute of Health, 1A-B, rue Thomas Edison, L-1445 Strassen, Luxembourg; (M.I.); or (A.B.); (S.S.F.D.C.); (H.S.)
| | - Michael R. La Frano
- Department of Food Science and Nutrition, California Polytechnic State University, 1 Grand Avenue, San Luis Obispo, CA 93407, USA;
- Center for Health Research, California Polytechnic State University, 1 Grand Avenue, San Luis Obispo, CA 93407, USA
| | - Torsten Bohn
- Nutrition and Health Research Group, Population Health Department, Luxembourg Institute of Health, 1A-B, rue Thomas Edison, L-1445 Strassen, Luxembourg; (M.I.); or (A.B.); (S.S.F.D.C.); (H.S.)
| |
Collapse
|
149
|
Troncone E, Marafini I, Del Vecchio Blanco G, Di Grazia A, Monteleone G. Novel Therapeutic Options for People with Ulcerative Colitis: An Update on Recent Developments with Janus Kinase (JAK) Inhibitors. Clin Exp Gastroenterol 2020; 13:131-139. [PMID: 32440190 PMCID: PMC7211304 DOI: 10.2147/ceg.s208020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/13/2020] [Indexed: 12/13/2022] Open
Abstract
Crohn’s disease (CD) and ulcerative colitis (UC), the main forms of inflammatory bowel disease (IBD) in human beings, are chronic relapsing-remitting disorders of the gastrointestinal tract, which usually require lifelong therapies. For many years, IBD have been managed with corticosteroids, aminosalicylates and immunosuppressants (ie, thiopurines). The advent of biologic therapies (anti-TNF-α agents) has significantly improved the outcome of IBD patients in terms of prolonged clinical remission, corticosteroid sparing, achievement of mucosal healing and prevention of disease-related complications. Nevertheless, primary failure or loss of response to biologics occur in about 50% of patients treated with these drugs. Therefore, the need for new effective treatments for such patients has critically emerged as an urgent priority. With this regard, several small-molecule drugs (SMDs) targeting lymphocyte trafficking (ie, sphingosine-1-phosphate receptor modulators) and the JAK/STAT pathway (eg, tofacitinib) have been recently developed and tested in IBD. In particular, JAK inhibitors are oral compounds characterized by short half-life, low antigenicity and the ability to dampen several pro-inflammatory pathways simultaneously. Tofacitinib, a pan-JAK inhibitor, has shown good efficacy and safety in UC clinical trials and has been recently approved for the treatment of UC patients. In this review, we analyze the main evidence supporting the use of JAK inhibitors in UC and explore the unanswered questions about the use of this class of drug in UC.
Collapse
Affiliation(s)
- Edoardo Troncone
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Irene Marafini
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | | | - Antonio Di Grazia
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
150
|
Lee S, Kim J, Min H, Seong RH. RORγt-driven T H17 Cell Differentiation Requires Epigenetic Control by the Swi/Snf Chromatin Remodeling Complex. iScience 2020; 23:101106. [PMID: 32434140 PMCID: PMC7235640 DOI: 10.1016/j.isci.2020.101106] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 02/24/2020] [Accepted: 04/22/2020] [Indexed: 12/11/2022] Open
Abstract
Epigenetic regulation, including chromatin accessibility and posttranslational modifications of histones, is of importance for T cell lineage decision. TH17 cells play a critical role in protective mucosal immunity and pathogenic multiple autoimmune diseases. The differentiation of TH17 cells is dictated by a master transcription factor, RORγt. However, the epigenetic mechanism that controls TH17 cell differentiation remains poorly understood. Here we show that the Swi/Snf complex is required for TH17-mediated cytokine production both in vitro and in vivo. We demonstrate that RORγt recruits and forms a complex with the Swi/Snf complex to cooperate for the RORγt-mediated epigenetic modifications of target genes, including both permissive and repressive ones for TH17 cell differentiation. Our findings thus highlight the Swi/Snf complex as an essential epigenetic regulator of TH17 cell differentiation and provide a basis for the understanding of how a master transcription factor RORγt collaborates with the Swi/Snf complex to govern epigenetic regulation. The Swi/Snf complex plays essential roles for TH17 differentiation SRG3/mBAF155 deficiency abrogates the expression of major target genes of RORγt RORγt-dependent TH17 transcriptional program requires the Swi/Snf complex The Swi/Snf complex is required for RORγt-driven histone modifications
Collapse
Affiliation(s)
- Sungkyu Lee
- Departement of Biological Sciences, Institute of Molecular Biology and Genetics (Bldg 105), Seoul National University, Gwanak-gu, Gwanak-ro 1, Seoul 151-742-08826, Korea
| | - Jieun Kim
- Departement of Biological Sciences, Institute of Molecular Biology and Genetics (Bldg 105), Seoul National University, Gwanak-gu, Gwanak-ro 1, Seoul 151-742-08826, Korea
| | - Hyungyu Min
- Departement of Biological Sciences, Institute of Molecular Biology and Genetics (Bldg 105), Seoul National University, Gwanak-gu, Gwanak-ro 1, Seoul 151-742-08826, Korea
| | - Rho H Seong
- Departement of Biological Sciences, Institute of Molecular Biology and Genetics (Bldg 105), Seoul National University, Gwanak-gu, Gwanak-ro 1, Seoul 151-742-08826, Korea.
| |
Collapse
|