1
|
Mihalić A, Železnjak J, Lisnić B, Jonjić S, Juranić Lisnić V, Brizić I. Immune surveillance of cytomegalovirus in tissues. Cell Mol Immunol 2024; 21:959-981. [PMID: 39134803 PMCID: PMC11364667 DOI: 10.1038/s41423-024-01186-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/14/2024] [Indexed: 09/01/2024] Open
Abstract
Cytomegalovirus (CMV), a representative member of the Betaherpesvirinae subfamily of herpesviruses, is common in the human population, but immunocompetent individuals are generally asymptomatic when infected with this virus. However, in immunocompromised individuals and immunologically immature fetuses and newborns, CMV can cause a wide range of often long-lasting morbidities and even death. CMV is not only widespread throughout the population but it is also widespread in its hosts, infecting and establishing latency in nearly all tissues and organs. Thus, understanding the pathogenesis of and immune responses to this virus is a prerequisite for developing effective prevention and treatment strategies. Multiple arms of the immune system are engaged to contain the infection, and general concepts of immune control of CMV are now reasonably well understood. Nonetheless, in recent years, tissue-specific immune responses have emerged as an essential factor for resolving CMV infection. As tissues differ in biology and function, so do immune responses to CMV and pathological processes during infection. This review discusses state-of-the-art knowledge of the immune response to CMV infection in tissues, with particular emphasis on several well-studied and most commonly affected organs.
Collapse
Affiliation(s)
- Andrea Mihalić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Jelena Železnjak
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Berislav Lisnić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Stipan Jonjić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Department of Biomedical Sciences, Croatian Academy of Sciences and Arts, Rijeka, Croatia
| | - Vanda Juranić Lisnić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.
| | - Ilija Brizić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.
| |
Collapse
|
2
|
Liu W, Zeng X, Wang X, Hu Y, Chen L, Luo N, Ouyang D, Rao T. 2,3,5,4'- tetrahydroxystilbene-2-O-β-D- glucopyranoside (TSG)-Driven immune response in the hepatotoxicity of Polygonum multiflorum. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117865. [PMID: 38369066 DOI: 10.1016/j.jep.2024.117865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 01/31/2024] [Accepted: 02/03/2024] [Indexed: 02/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE 2,3,5,4'-tetrahydroxystilbene-2-O-β-D-glucopyranoside (TSG) as the primary constituent of Polygonum multiflorum Thumb. (PM) possesses anti-oxidative, antihypercholesterolemic, anti-tumor and many more biological activities. The root of PM has been used as a tonic medicine for thousands of years. However, cases of PM-induced liver injury are occasionally reported, and considered to be related to the host immune status. AIM OF THE STUDY The primary toxic elements and specific mechanisms PM causing liver damage are still not thoroughly clear. Our study aimed to investigate the influences of TSG on the immune response in idiosyncratic hepatotoxicity of PM. MATERIALS AND METHODS The male C57BL/6 mice were treated with different doses of TSG and the alterations in liver histology, serum liver enzyme levels, proportions of T cells and cytokines secretion were evaluated by hematoxylin and eosin (HE), RNA sequencing, quantitative real time polymerase chain reaction (qRT-PCR), Flow cytometry (FCM), and enzyme-linked immunosorbent assay (ELISA), respectively. Then, primary spleen cells from drug-naive mice were isolated and cultured with TSG in vitro. T cell subsets proliferation and cytokines secretion after treated with TSG were assessed by CCK8, FCM and ELISA. In addition, mice were pre-treated with anti-CD25 for depleting regulatory T cells (Tregs), and then administered with TSG. Liver functions and immunological alterations were analyzed to evaluate liver injury. RESULTS Data showed that TSG induced liver damage, and immune cells infiltration in the liver tissues. FCM results showed that TSG could activate CD4+T and CD8+T in the liver. Results further confirmed that TSG notably up-regulated the levels of inflammatory cytokines including TNF-α, IFN-γ, IL-18, perforin and granzyme B in the liver tissues. Furthermore, based on transcriptomics profiles, some immune system-related pathways including leukocyte activation involved in inflammatory response, leukocyte cell-cell adhesion, regulation of interleukin-1 beta production, mononuclear cell migration, antigen processing and presentation were altered in TSG treated mice. CD8+T/CD4+T cells were also stimulated by TSG in vitro. Interestingly, increased proportion of Tregs was observed after TSG treatment in vitro and in vivo. Foxp3 and TGF-β1 mRNA expressions were up-regulated in the liver tissues. Depletion of Tregs moderately enhanced TSG induced the secretion of inflammatory cytokines in serum. CONCLUSIONS Our findings showed that TSG could trigger CD4+T and CD8+T cells proliferation, promote cytokines secretion, which revealed that adaptive immune response associated with the mild liver injury cause by TSG administration. Regulatory T cells (Tregs) mainly sustain immunological tolerance, and in this study, the progression of TSG induced liver injury was limited by Tregs. The results of our investigations allow us to preliminarily understand the mechanisms of PM related idiosyncratic hepatotoxicity.
Collapse
Affiliation(s)
- Wenhui Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan Province, 410078, China; Department of Clinical Laboratory, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi Province, 541001, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan Province, 410078, China
| | - Xiangchang Zeng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan Province, 410078, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan Province, 410078, China; Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, Hunan Province, 410221, China
| | - Xinfeng Wang
- Department of Human Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, Guangxi Province, 541199, China
| | - Yuwei Hu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan Province, 410078, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan Province, 410078, China
| | - Lulu Chen
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, Hunan Province, 410221, China
| | - Naixiang Luo
- Department of Immunology, College of Basic Medicine, Guilin Medical University, Guilin, Guangxi Province, 541199, China.
| | - Dongsheng Ouyang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan Province, 410078, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan Province, 410078, China; Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, Hunan Province, 410221, China.
| | - Tai Rao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan Province, 410078, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan Province, 410078, China.
| |
Collapse
|
3
|
Nakonechnaya TO, Moltedo B, Putintseva EV, Leyn S, Bolotin DA, Britanova OV, Shugay M, Chudakov DM. Convergence, plasticity, and tissue residence of regulatory T cell response via TCR repertoire prism. eLife 2024; 12:RP89382. [PMID: 38591522 PMCID: PMC11003740 DOI: 10.7554/elife.89382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024] Open
Abstract
Suppressive function of regulatory T cells (Treg) is dependent on signaling of their antigen receptors triggered by cognate self, dietary, or microbial peptides presented on MHC II. However, it remains largely unknown whether distinct or shared repertoires of Treg TCRs are mobilized in response to different challenges in the same tissue or the same challenge in different tissues. Here we use a fixed TCRβ chain FoxP3-GFP mouse model to analyze conventional (eCD4) and regulatory (eTreg) effector TCRα repertoires in response to six distinct antigenic challenges to the lung and skin. This model shows highly 'digital' repertoire behavior with easy-to-track challenge-specific TCRα CDR3 clusters. For both eCD4 and eTreg subsets, we observe challenge-specific clonal expansions yielding homologous TCRα clusters within and across animals and exposure sites, which are also reflected in the draining lymph nodes but not systemically. Some CDR3 clusters are shared across cancer challenges, suggesting a response to common tumor-associated antigens. For most challenges, eCD4 and eTreg clonal response does not overlap. Such overlap is exclusively observed at the sites of certain tumor challenges, and not systematically, suggesting transient and local tumor-induced eCD4=>eTreg plasticity. This transition includes a dominant tumor-responding eCD4 CDR3 motif, as well as characteristic iNKT TCRα CDR3. In addition, we examine the homeostatic tissue residency of clonal eTreg populations by excluding the site of challenge from our analysis. We demonstrate that distinct CDR3 motifs are characteristic of eTreg cells residing in particular lymphatic tissues, regardless of the challenge. This observation reveals the tissue-resident, antigen-specific clonal Treg populations.
Collapse
Affiliation(s)
- Tatyana O Nakonechnaya
- Institute of Translational Medicine, Pirogov Russian National Research Medical UniversityMoscowRussian Federation
- Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
| | - Bruno Moltedo
- Howard Hughes Medical Institute and Immunology Program, Sloan Kettering Institute and Ludwig Center at Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Ekaterina V Putintseva
- Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
| | - Sofya Leyn
- Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
| | - Dmitry A Bolotin
- Institute of Translational Medicine, Pirogov Russian National Research Medical UniversityMoscowRussian Federation
- Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
| | - Olga V Britanova
- Institute of Translational Medicine, Pirogov Russian National Research Medical UniversityMoscowRussian Federation
- Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
| | - Mikhail Shugay
- Institute of Translational Medicine, Pirogov Russian National Research Medical UniversityMoscowRussian Federation
- Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
| | - Dmitriy M Chudakov
- Institute of Translational Medicine, Pirogov Russian National Research Medical UniversityMoscowRussian Federation
- Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
- Central European Institute of TechnologyBrnoCzech Republic
- Abu Dhabi Stem Cells CenterAbu DhabiUnited Arab Emirates
| |
Collapse
|
4
|
Tong M, Yang X, Qiao Y, Liu G, Ge H, Huang G, Wang Y, Yang Y, Fan W. Serine protease inhibitor from the muscle larval Trichinella spiralis ameliorates non-alcoholic fatty liver disease in mice via anti-inflammatory properties and gut-liver crosstalk. Biomed Pharmacother 2024; 172:116223. [PMID: 38325266 DOI: 10.1016/j.biopha.2024.116223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/09/2024] Open
Abstract
Trichinella spiralis is recognized for its ability to regulate host immune responses. The serine protease inhibitor of T. spiralis (Ts-SPI) participates in T. spiralis-mediated immunoregulatory effects. Studies have shown that helminth therapy exhibits therapeutic effects on metabolic diseases. In addition, we previously found that T. spiralis-derived crude antigens could alleviate diet-induced obesity. Thus, Ts-SPI was hypothesized to alleviate non-alcoholic fatty liver disease (NAFLD). Herein, recombinant Ts-SPI (rTs-SPI) was prepared from the muscle larvae T. spiralis. The relative molecular mass of rTs-SPI was approximately 35,000 Da, and western blot analysis indicated good immunoreactivity. rTs-SPI ameliorated hepatic steatosis, inflammation, and pyroptosis in NAFLD mice, which validated the hypothesis. rTs-SPI also reduced macrophage infiltration, significantly expanded Foxp3+ Treg population, and inactivated TLR4/NF-κB/NLRP3 signaling in the liver. Furthermore, rTs-SPI treatment significantly shifted the gut microbiome structure, with a remarkable increase in beneficial bacteria and reduction in harmful bacteria to improve gut barrier integrity. Finally, Abx-treated mice and FMT confirmed that gut-liver crosstalk contributed to NAFLD improvement after rTs-SPI treatment. Taken together, Taken together, these findings suggest that rTs-SPI exerts therapeutic effects in NAFLD via anti-inflammatory activity and gut-liver crosstalk.
Collapse
Affiliation(s)
- Mingwei Tong
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and Shanxi Key Laboratory of Cellular Physiology, Taiyuan 030001, China.
| | - Xiaodan Yang
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China
| | - Yuyu Qiao
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China
| | - Ge Liu
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China
| | - Huihui Ge
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China
| | - Guangrong Huang
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China
| | - Yanhong Wang
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and Shanxi Key Laboratory of Cellular Physiology, Taiyuan 030001, China
| | - Yong Yang
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and Shanxi Key Laboratory of Cellular Physiology, Taiyuan 030001, China.
| | - Weiping Fan
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and Shanxi Key Laboratory of Cellular Physiology, Taiyuan 030001, China.
| |
Collapse
|
5
|
Karner D, Kvestak D, Lisnic B, Cokaric Brdovcak M, Juranic Lisnic V, Kucan Brlic P, Hasan M, Lenac Rovis T. Comprehensive Analysis of Soluble Mediator Profiles in Congenital CMV Infection Using an MCMV Model. Viruses 2024; 16:208. [PMID: 38399983 PMCID: PMC10891658 DOI: 10.3390/v16020208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/25/2024] Open
Abstract
Congenital human cytomegalovirus (HCMV) infection may cause life-threatening disease and permanent damage to the central nervous system. The mouse model of CMV infection is most commonly used to study mechanisms of infection and pathogenesis. While essential to limit mouse CMV (MCMV) replication, the inflammatory responses, particularly IFNγ and TNFα, cause neurodevelopmental abnormalities. Other soluble mediators of the immune response in most tissues remain largely unexplored. To address this gap, we quantified 48 soluble mediators of the immune response, including 32 cytokines, 10 chemokines, 3 growth factors/regulators, and 3 soluble receptors in the spleen, liver, lungs, and brain at 9 and 14 days postinfection (dpi). Our analysis found 25 induced molecules in the brain at 9 dpi, with an additional 8 showing statistically elevated responses at 14 dpi. Specifically, all analyzed CCL group cytokines (CCL2, CCL3, CCL4, CCL5, CCL7, and CCL11) were upregulated at 14 dpi in the brain. Furthermore, data revealed differentially regulated analytes across tissues, such as CCL11, CXCL5, and IL-10 in the brain, IL-33/IL-33R in the liver, and VEGF-a and IL-5 in the lungs. Overall, this study provides an overview of the immune dynamics of soluble mediators in congenital CMV.
Collapse
Affiliation(s)
- Dubravka Karner
- Center for Proteomics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (D.K.); (D.K.); (B.L.); (M.C.B.); (V.J.L.); (P.K.B.)
| | - Daria Kvestak
- Center for Proteomics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (D.K.); (D.K.); (B.L.); (M.C.B.); (V.J.L.); (P.K.B.)
| | - Berislav Lisnic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (D.K.); (D.K.); (B.L.); (M.C.B.); (V.J.L.); (P.K.B.)
| | - Maja Cokaric Brdovcak
- Center for Proteomics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (D.K.); (D.K.); (B.L.); (M.C.B.); (V.J.L.); (P.K.B.)
| | - Vanda Juranic Lisnic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (D.K.); (D.K.); (B.L.); (M.C.B.); (V.J.L.); (P.K.B.)
| | - Paola Kucan Brlic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (D.K.); (D.K.); (B.L.); (M.C.B.); (V.J.L.); (P.K.B.)
| | - Milena Hasan
- Cytometry and Biomarkers Unit of Technology and Service (CB TechS), Institut Pasteur, Université Paris Cité, 75015 Paris, France;
| | - Tihana Lenac Rovis
- Center for Proteomics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia; (D.K.); (D.K.); (B.L.); (M.C.B.); (V.J.L.); (P.K.B.)
| |
Collapse
|
6
|
Norris GT, Ames JM, Ziegler SF, Oberst A. Oligodendrocyte-derived IL-33 functions as a microglial survival factor during neuroinvasive flavivirus infection. PLoS Pathog 2023; 19:e1011350. [PMID: 37983247 PMCID: PMC10695366 DOI: 10.1371/journal.ppat.1011350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 12/04/2023] [Accepted: 11/05/2023] [Indexed: 11/22/2023] Open
Abstract
In order to recover from infection, organisms must balance robust immune responses to pathogens with the tolerance of immune-mediated pathology. This balance is particularly critical within the central nervous system, whose complex architecture, essential function, and limited capacity for self-renewal render it susceptible to both pathogen- and immune-mediated pathology. Here, we identify the alarmin IL-33 and its receptor ST2 as critical for host survival to neuroinvasive flavivirus infection. We identify oligodendrocytes as the critical source of IL-33, and microglia as the key cellular responders. Notably, we find that the IL-33/ST2 axis does not impact viral control or adaptive immune responses; rather, it is required to promote the activation and survival of microglia. In the absence of intact IL-33/ST2 signaling in the brain, neuroinvasive flavivirus infection triggered aberrant recruitment of monocyte-derived peripheral immune cells, increased neuronal stress, and neuronal cell death, effects that compromised organismal survival. These findings identify IL-33 as a critical mediator of CNS tolerance to pathogen-initiated immunity and inflammation.
Collapse
Affiliation(s)
- Geoffrey T. Norris
- Department of Immunology, University of Washington, Seattle Washington, United States of America
| | - Joshua M. Ames
- Department of Immunology, University of Washington, Seattle Washington, United States of America
| | - Steven F. Ziegler
- Department of Immunology, University of Washington, Seattle Washington, United States of America
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle Washington, United States of America
| | - Andrew Oberst
- Department of Immunology, University of Washington, Seattle Washington, United States of America
| |
Collapse
|
7
|
Raugh A, Jing Y, Bettini ML, Bettini M. The amphiregulin/EGFR axis has limited contribution in controlling autoimmune diabetes. Sci Rep 2023; 13:18653. [PMID: 37903947 PMCID: PMC10616065 DOI: 10.1038/s41598-023-45738-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/23/2023] [Indexed: 11/01/2023] Open
Abstract
Conventional immunosuppressive functions of CD4+Foxp3+ regulatory T cells (Tregs) in type 1 diabetes (T1D) pathogenesis have been well described, but whether Tregs have additional non-immunological functions supporting tissue homeostasis in pancreatic islets is unknown. Within the last decade novel tissue repair functions have been ascribed to Tregs. One function is production of the epidermal growth factor receptor (EGFR) ligand, amphiregulin, which promotes tissue repair in response to inflammatory or mechanical tissue injury. However, whether such pathways are engaged during autoimmune diabetes and promote tissue repair is undetermined. Previously, we observed that upregulation of amphiregulin at the transcriptional level was associated with functional Treg populations in the non-obese diabetic (NOD) mouse model of T1D. From this we postulated that amphiregulin promoted islet tissue repair and slowed the progression of diabetes in NOD mice. Here, we report that islet-infiltrating Tregs have increased capacity to produce amphiregulin, and that both Tregs and beta cells express EGFR. Moreover, we show that amphiregulin can directly modulate mediators of endoplasmic reticulum stress in beta cells. Despite this, NOD amphiregulin deficient mice showed no acceleration of spontaneous autoimmune diabetes. Taken together, the data suggest that the ability for amphiregulin to affect the progression of autoimmune diabetes is limited.
Collapse
Affiliation(s)
- Arielle Raugh
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Pathology, University of Utah, Salt Lake City, UT, 84112, USA
| | - Yi Jing
- Microbiology and Immunology Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Pathology, University of Utah, Salt Lake City, UT, 84112, USA
| | - Matthew L Bettini
- Department of Pathology, University of Utah, Salt Lake City, UT, 84112, USA
| | - Maria Bettini
- Department of Pathology, University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
8
|
Raugh A, Jing Y, Bettini ML, Bettini M. The Amphiregulin/EGFR axis has limited contribution in controlling autoimmune diabetes. RESEARCH SQUARE 2023:rs.3.rs-3204139. [PMID: 37577652 PMCID: PMC10418547 DOI: 10.21203/rs.3.rs-3204139/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Conventional immunosuppressive functions of CD4+Foxp3+ regulatory T cells (Tregs) in type 1 diabetes (T1D) pathogenesis have been well described, but whether Tregs have additional non-immunological functions supporting tissue homeostasis in pancreatic islets is unknown. Within the last decade novel tissue repair functions have been ascribed to Tregs. One function is production of the epidermal growth factor receptor (EGFR) ligand, amphiregulin, which promotes tissue repair in response to inflammatory or mechanical tissue injury. Whether such pathways are engaged during autoimmune diabetes and promote tissue repair is undetermined. Previously, we observed upregulation of amphiregulin at the transcriptional level was associated with functional Treg populations in the non-obese diabetic (NOD) mouse model of T1D. We postulated that amphiregulin promoted islet tissue repair and slowed the progression of diabetes in NOD mice. Here, we report that islet-infiltrating Tregs have increased capacity to produce amphiregulin and both Tregs and beta cells express EGFR. Moreover, we show that amphiregulin can directly modulate mediators of endoplasmic reticulum (ER) stress in beta cells. Despite this, NOD amphiregulin deficient mice showed no acceleration of spontaneous autoimmune diabetes. Taken together, the data suggest that the ability for amphiregulin to affect the progression of autoimmune diabetes is limited.
Collapse
Affiliation(s)
- Arielle Raugh
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, Texas, 77030, USA
- Department of Pathology, University of Utah, Salt Lake City, Utah, 84112, USA
| | - Yi Jing
- Department of Pediatric Endocrinology, Texas Children’s Hospital, Houston, Texas, 77030, USA
- Department of Pathology, University of Utah, Salt Lake City, Utah, 84112, USA
| | - Matthew L. Bettini
- Department of Pathology, University of Utah, Salt Lake City, Utah, 84112, USA
| | - Maria Bettini
- Department of Pathology, University of Utah, Salt Lake City, Utah, 84112, USA
| |
Collapse
|
9
|
Ruan Y, Wen Z, Chen K, Xi J, Wu B, Xu Z, Jiang M, Zhang J, Chen Y, Liu Q. Exogenous Interleukin-37 Alleviates Hepatitis with Reduced Dendritic Cells and Induced Regulatory T Cells in Acute Murine Cytomegalovirus Infection. J Immunol Res 2023; 2023:1462048. [PMID: 37215069 PMCID: PMC10198762 DOI: 10.1155/2023/1462048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 04/23/2023] [Accepted: 04/25/2023] [Indexed: 05/24/2023] Open
Abstract
Human cytomegalovirus (HCMV) infection is globally distributed, and the liver is one of the major targeting organs. So far, the mechanisms for cell and organ damage have not fully been elucidated and the treatments for the infection are mainly at symptoms. IL-37 has shown a protective role in certain inflammatory diseases. In the present study, potential protective effect of exogenous IL-37 on murine cytomegalovirus- (MCMV-) infected hepatitis was evaluated through analyses of serum transaminases, the liver histopathology and cytokine expression, and functional state of dendritic cells (DCs) and regulatory T cells (Tregs). These analyses showed a significant decrease in serum transaminase levels and a lower Ishak histopathologic score at the early stage of MCMV-infected mice with exogenous IL-37 pretreatment. The frequencies of MHC-Ⅱ, CD40, CD80, and CD86 positive DCs in the liver and spleen were decreased significantly at 7 days postinfection (dpi) in MCMV-infected mice with IL-37 pretreatment when compared with those without the pretreatment, while the total number of DCs in the liver was reduced in IL-37-pretreated mice. The induction of Tregs in the spleen was enhanced at dpi 3 with IL-37 pretreatment in MCMV-infected mice. The mRNA expression levels of cytokines in the liver were decreased significantly (IL-1β, IL-6, IL-10, IL-4) or to some extent (TGF-β and TNF-α). The present study suggested that exogenous IL-37 can alleviate MCMV-infected hepatitis, likely through reduced DCs and induced Tregs with a weaker cytokine storm, demonstrating its potential value in clinical management for HCMV-infected hepatitis.
Collapse
Affiliation(s)
- Yufei Ruan
- Department of Pediatric Infectious Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325027, China
- Department of Emergency, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhengwang Wen
- Department of Pediatric Infectious Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325027, China
| | - Ke Chen
- Department of Pediatric Infectious Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325027, China
| | - Jianan Xi
- Department of Pediatric Infectious Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325027, China
| | - Bo Wu
- School of Basic Medicine, Anhui Medical University, Hefei 230000, China
| | - Zhiyong Xu
- School of Basic Medicine, Anhui Medical University, Hefei 230000, China
| | - Minzhi Jiang
- School of Basic Medicine, Anhui Medical University, Hefei 230000, China
| | - Junling Zhang
- School of Basic Medicine, Anhui Medical University, Hefei 230000, China
| | - Yiping Chen
- Department of Pediatric Infectious Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325027, China
| | - Qi Liu
- Department of Pediatric Infectious Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325027, China
| |
Collapse
|
10
|
A Novel Synbiotic Alleviates Autoimmune Hepatitis by Modulating the Gut Microbiota-Liver Axis and Inhibiting the Hepatic TLR4/NF-κB/NLRP3 Signaling Pathway. mSystems 2023; 8:e0112722. [PMID: 36794950 PMCID: PMC10134874 DOI: 10.1128/msystems.01127-22] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
Autoimmune hepatitis (AIH) is a liver disease characterized by chronic liver inflammation. The intestinal barrier and microbiome play critical roles in AIH progression. AIH treatment remains challenging because first-line drugs have limited efficacy and many side effects. Thus, there is growing interest in developing synbiotic therapies. This study investigated the effects of a novel synbiotic in an AIH mouse model. We found that this synbiotic (Syn) ameliorated liver injury and improved liver function by reducing hepatic inflammation and pyroptosis. The Syn reversed gut dysbiosis, as indicated by an increase in beneficial bacteria (e.g., Rikenella and Alistipes) and a decrease in potentially harmful bacteria (e.g., Escherichia-Shigella) and lipopolysaccharide (LPS)-bearing Gram-negative bacterial levels. The Syn maintained intestinal barrier integrity, reduced LPS, and inhibited the TLR4/NF-κB and NLRP3/Caspase-1 signaling pathway. In addition, microbiome phenotype prediction by BugBase and bacterial functional potential prediction using Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) showed that Syn improved gut microbiota function involving inflammatory injury, metabolism, immune response, and pathopoiesia. Furthermore, the new Syn was as effective as prednisone against AIH. Therefore, this novel Syn could be a candidate drug for alleviating AIH through its anti-inflammatory and antipyroptosis properties that relieve endothelial dysfunction and gut dysbiosis. IMPORTANCE Synbiotics can ameliorate liver injury and improve liver function by reducing hepatic inflammation and pyroptosis. Our data indicate that our new Syn not only reverses gut dysbiosis by increasing beneficial bacteria and decreasing lipopolysaccharide (LPS)-bearing Gram-negative bacteria but also maintains intestinal barrier integrity. Thus, its mechanism might be associated with modulating gut microbiota composition and intestinal barrier function by inhibiting the TLR4/NF-κB/NLRP3/pyroptosis signaling pathway in the liver. This Syn is as effective as prednisone in treating AIH without side effects. Based on these findings, this novel Syn represents a potential therapeutic agent for AIH in clinical practice.
Collapse
|
11
|
The translational approach to liver transplantation. Ann Hepatol 2022; 27:100747. [PMID: 36057435 DOI: 10.1016/j.aohep.2022.100747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/11/2022] [Indexed: 02/04/2023]
|
12
|
Riet T, Chmielewski M. Regulatory CAR-T cells in autoimmune diseases: Progress and current challenges. Front Immunol 2022; 13:934343. [PMID: 36032080 PMCID: PMC9399761 DOI: 10.3389/fimmu.2022.934343] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
CAR (Chimeric Antigen Receptor) T-cell therapy has revolutionized the field of oncology in recent years. This innovative shift in cancer treatment also provides the opportunity to improve therapies for many patients suffering from various autoimmune diseases. Recent studies have confirmed the therapeutic suppressive potential of regulatory T cells (Tregs) to modulate immune response in autoimmune diseases. However, the polyclonal character of regulatory T cells and their unknown TCR specificity impaired their therapeutic potency in clinical implementation. Genetical engineering of these immune modulating cells to express antigen-specific receptors and using them therapeutically is a logical step on the way to overcome present limitations of the Treg strategy for the treatment of autoimmune diseases. Encouraging preclinical studies successfully demonstrated immune modulating properties of CAR Tregs in various mouse models. Still, there are many concerns about targeted Treg therapies relating to CAR target selectivity, suppressive functions, phenotype stability and safety aspects. Here, we summarize recent developments in CAR design, Treg biology and future strategies and perspectives in CAR Treg immunotherapy aiming at clinical translation.
Collapse
|
13
|
Contreras A, Wiesner DL, Kingstad-Bakke B, Lee W, Svaren JP, Klein BS, Suresh M. BACH2 in TRegs Limits the Number of Adipose Tissue Regulatory T Cells and Restrains Type 2 Immunity to Fungal Allergens. J Immunol Res 2022; 2022:6789055. [PMID: 36033397 PMCID: PMC9410868 DOI: 10.1155/2022/6789055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 07/19/2022] [Indexed: 01/28/2023] Open
Abstract
FoxP3+ regulatory T cells (Tregs) are essential for self-tolerance and moderating tissue-damaging inflammation. Tregs that develop and mature in the thymus are classified as central Tregs or effector Tregs based on whether Tregs predominately inhabit secondary lymphoid organs (central Tregs) or tissues (effector Tregs). By generating mice that are conditionally deficient for Bach2 in peripheral Tregs, we have examined the role of Bach2 in regulating Treg homeostasis and effector functions. Unlike global and T cell-specific Bach2-deficient mice, Treg-specific Bach2 ablation did not result in unprovoked TH2 inflammation in the lungs. However, Bach2 deficiency in Tregs led to augmented expressions of IRF4, BATF, and GATA3 and a significant increase in the accumulation of ST2 (IL-33R)+ve effector Tregs in the spleen and visceral adipose tissue (VAT) but not in the lungs. Enhanced Bach2-deficient Treg numbers in VAT was not linked to hyperresponsiveness to exogenous IL-33 in vivo. Most strikingly, Treg-specific Bach2 deficiency resulted in enhanced fungal protease-induced Type 2 allergic inflammation in the lungs, with no detectable effects on Type 1 responses to systemic or respiratory viral infections. In summary, we ascribe vital roles for Bach2 in peripheral Tregs: as a transcriptional checkpoint to limit precocious differentiation into effector Tregs in lymphoid tissues and as a regulator of the functional program that restrains Type 2 but not Type 1 inflammation in lungs. Results presented in this manuscript implicate dysregulated Tregs in the pathogenesis of airway hypersensitivities, asthma, and other allergic disorders.
Collapse
Affiliation(s)
- Amanda Contreras
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, 53706 WI, USA
| | - Darin L. Wiesner
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, 53706 WI, USA
| | - Brock Kingstad-Bakke
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, 53706 WI, USA
| | - Woojong Lee
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, 53706 WI, USA
| | - John P. Svaren
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, 53706 WI, USA
| | - Bruce S. Klein
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, 53706 WI, USA
| | - M. Suresh
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, 53706 WI, USA
| |
Collapse
|
14
|
Copsel SN, Wolf D, Pfeiffer B, Barreras H, Perez VL, Levy RB. Recipient Tregs: Can They Be Exploited for Successful Hematopoietic Stem Cell Transplant Outcomes? Front Immunol 2022; 13:932527. [PMID: 35799783 PMCID: PMC9253768 DOI: 10.3389/fimmu.2022.932527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/18/2022] [Indexed: 02/03/2023] Open
Abstract
Human and mouse CD4+FoxP3+ T cells (Tregs) comprise non-redundant regulatory compartments which maintain self-tolerance and have been found to be of potential therapeutic usefulness in autoimmune disorders and transplants including allogeneic hematopoietic stem cell transplantation (allo-HSCT). There is substantial literature interrogating the application of donor derived Tregs for the prevention of graft versus host disease (GVHD). This Mini-Review will focus on the recipient's Tregs which persist post-transplant. Although treatment in patients with low dose IL-2 months post-HSCT are encouraging, manipulating Tregs in recipients early post-transplant is challenging, in part likely an indirect consequence of damage to the microenvironment required to support Treg expansion of which little is understood. This review will discuss the potential for manipulating recipient Tregs in vivo prior to and after HSCT (fusion proteins, mAbs). Strategies that would circumvent donor/recipient peripheral blood harvest, cell culture and ex-vivo Treg expansion will be considered for the translational application of Tregs to improve HSCT outcomes.
Collapse
Affiliation(s)
- Sabrina N. Copsel
- Department of Microbiology and Immunology, University of Miami School of Medicine, Miami, FL, United States
| | - Dietlinde Wolf
- Sylvester Comprehensive Cancer Center, University of Miami School of Medicine, Miami, FL, United States
| | - Brent Pfeiffer
- Department of Pediatrics, University of Miami School of Medicine, Miami, FL, United States
| | - Henry Barreras
- Department of Microbiology and Immunology, University of Miami School of Medicine, Miami, FL, United States
| | - Victor L. Perez
- Foster Center for Ocular Immunology, Duke Eye Center, Duke University, Durham, NC, United States
| | - Robert B. Levy
- Department of Microbiology and Immunology, University of Miami School of Medicine, Miami, FL, United States,Sylvester Comprehensive Cancer Center, University of Miami School of Medicine, Miami, FL, United States,Department of Ophthalmology, University of Miami School of Medicine, Miami, FL, United States,*Correspondence: Robert B. Levy,
| |
Collapse
|
15
|
Kaufmann B, Reca A, Kim AD, Feldstein AE. Novel Mechanisms for Resolution of Liver Inflammation: Therapeutic Implications. Semin Liver Dis 2021; 41:150-162. [PMID: 34107544 DOI: 10.1055/s-0041-1723031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Traditional concepts have classically viewed resolution of inflammation as a passive process yet insight into the pathways by which inflammation is resolved has challenged this idea. Resolution has been revealed as a highly dynamic and active event that is essential to counteract the dysregulated inflammatory response that drives diverse disease states. Abrogation of the hepatic inflammatory response through the stimulation of proresolving mechanisms represents a new paradigm in the setting of chronic inflammatory-driven liver diseases. Elucidation of the role of different cells of the innate and adaptive immune system has highlighted the interplay between them as an important orchestrator of liver repair. A finely tuned interaction between neutrophils and macrophages has risen as revolutionary mechanism that drives the restoration of hepatic function and architecture. Specialized proresolving mediators have also been shown to act as stop signals of the inflammatory response and promote resolution as well as tissue regeneration. In this review, we discuss the discovery and understanding of the mechanisms by which inflammation is resolved and highlight novel proresolving pathways that represent promising therapeutic strategies.
Collapse
Affiliation(s)
- Benedikt Kaufmann
- Department of Pediatrics, University of California, San Diego (UCSD), California and Rady Children's Hospital, San Diego, California.,Department of Surgery, TUM School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Agustina Reca
- Department of Pediatrics, University of California, San Diego (UCSD), California and Rady Children's Hospital, San Diego, California
| | - Andrea D Kim
- Department of Pediatrics, University of California, San Diego (UCSD), California and Rady Children's Hospital, San Diego, California
| | - Ariel E Feldstein
- Department of Pediatrics, University of California, San Diego (UCSD), California and Rady Children's Hospital, San Diego, California
| |
Collapse
|
16
|
Wang Y, Yang Y, Wang M, Wang S, Jeong JM, Xu L, Wen Y, Emontzpohl C, Atkins CL, Duong K, Moreno NF, Yuan X, Hall DR, Dar W, Feng D, Gao B, Xu Y, Czigany Z, Colgan SP, Bynon JS, Akira S, Brown JM, Eltzschig HK, Jacobsen EA, Ju C. Eosinophils attenuate hepatic ischemia-reperfusion injury in mice through ST2-dependent IL-13 production. Sci Transl Med 2021; 13:eabb6576. [PMID: 33536281 PMCID: PMC8167890 DOI: 10.1126/scitranslmed.abb6576] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 09/28/2020] [Accepted: 01/12/2021] [Indexed: 12/14/2022]
Abstract
Eosinophils are a myeloid cell subpopulation that mediates type 2 T helper cell immune responses. Unexpectedly, we identified a rapid accumulation of eosinophils in 22 human liver grafts after hepatic transplantation. In contrast, no eosinophils were detectable in healthy liver tissues before transplantation. Studies with two genetic mouse models of eosinophil deficiency and a mouse model of antibody-mediated eosinophil depletion revealed exacerbated liver injury after hepatic ischemia and reperfusion. Adoptive transfer of bone marrow-derived eosinophils normalized liver injury of eosinophil-deficient mice and reduced hepatic ischemia and reperfusion injury in wild-type mice. Mechanistic studies combining genetic and adoptive transfer approaches identified a critical role of suppression of tumorigenicity (ST2)-dependent production of interleukin-13 by eosinophils in the hepatoprotection against ischemia-reperfusion-induced injury. Together, these data provide insight into a mechanism of eosinophil-mediated liver protection that could serve as a therapeutic target to improve outcomes of patients undergoing liver transplantation.
Collapse
Affiliation(s)
- Yaochun Wang
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Center for Translational Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yang Yang
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Meng Wang
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Shuhong Wang
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jong-Min Jeong
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Long Xu
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yankai Wen
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Christoph Emontzpohl
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Constance Lynn Atkins
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Kevin Duong
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Nicolas F Moreno
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - David R Hall
- Department of Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Wasim Dar
- Department of Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Dechun Feng
- Laboratory of Liver Disease, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Bin Gao
- Laboratory of Liver Disease, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zoltan Czigany
- Department of Surgery and Transplantation, Faculty of Medicine, University Hospital RWTH Aachen, Aachen 52074, Germany
| | - Sean P Colgan
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - J Steve Bynon
- Department of Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Shizuo Akira
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Jared M Brown
- School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Holger K Eltzschig
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Elizabeth A Jacobsen
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Cynthia Ju
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
17
|
Khan HA, Munir T, Khan JA, Shafia Tehseen Gul AH, Ahmad MZ, Aslam MA, Umar MN, Arshad MI. IL-33 ameliorates liver injury and inflammation in Poly I:C and Concanavalin-A induced acute hepatitis. Microb Pathog 2020; 150:104716. [PMID: 33383149 DOI: 10.1016/j.micpath.2020.104716] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 01/04/2023]
Abstract
The IL-33/ST2 axis is known to be involved in liver pathologies and IL-33 is over-expressed in mouse hepatitis models. We aimed to investigate the proposed protective effect of IL-33 in murine fulminant hepatitis induced by a Toll like receptor 3 (TLR3) viral mimetic, Poly I:C or by Concanavalin-A (ConA). The Balb/C mice were administered intravenously with ConA (15 mg/kg) or Poly I:C (30 μg/mouse) to induce acute hepatitis along with vehicle control. The recombinant mouse IL-33 (rIL-33) was injected (0.2 μg/mouse) to mice 2 h prior to ConA or Poly I:C injection to check its hepato-protective effects. The gross lesions, level of serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), histopathology (H&E staining) and levels of IFNγ and TNFα were measured by ELISA. The gross pathological liver injury induced by Poly I:C or ConA was reduced by rIL-33 administration in mice. The levels of AST and ALT were significantly (P ≤ 0.05) higher in mice challenged with Poly I:C or ConA in comparison to control mice. The rIL-33 pre-treated mice in both Poly I:C and ConA challenge groups showed significantly (P ≤ 0.05) lower levels of AST and ALT, and decreased liver injury (parenchymal and per-vascular necrotic areas) in histological liver sections. The soluble levels of TNFα and IFNγ were significantly (P ≤ 0.05) raised in Poly I:C or ConA challenged mice than control mice. The levels of TNFα and IFNγ were significantly reduced (P ≤ 0.05) in rIL-33 pre-treated mice. In conclusion, the exogenous IL-33 administration mitigated liver injury and inflammation (decreased levels of IFNγ and TNFα) in Poly I:C and ConA-induced acute hepatitis in mice.
Collapse
Affiliation(s)
- Hilal Ahmad Khan
- Institute of Microbiology, University of Agriculture, Faisalabad, Pakistan
| | - Tariq Munir
- Institute of Microbiology, University of Agriculture, Faisalabad, Pakistan
| | - Junaid Ali Khan
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | | | - Muhammad Zishan Ahmad
- Department of Veterinary Pathology, Faculty of Veterinary and Animal Science, PMAS Arid Agriculture University, Rawalpindi, Pakistan
| | | | | | | |
Collapse
|
18
|
Estrada Brull A, Rost F, Oderbolz J, Kirchner FR, Leibundgut-Landmann S, Oxenius A, Joller N. CD85k Contributes to Regulatory T Cell Function in Chronic Viral Infections. Int J Mol Sci 2020; 22:E31. [PMID: 33375121 PMCID: PMC7792974 DOI: 10.3390/ijms22010031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/20/2022] Open
Abstract
Regulatory T cells (Tregs) prevent excessive immune responses and limit immune pathology upon infections. To fulfill this role in different immune environments elicited by different types of pathogens, Tregs undergo functional specialization into distinct subsets. During acute type 1 immune responses, type 1 Tregs are induced and recruited to the site of ongoing Th1 responses to efficiently control Th1 responses. However, whether a similar specialization process also takes place following chronic infections is still unknown. In this study, we investigated Treg specialization in persistent viral infections using lymphocytic choriomeningitis virus (LCMV) and murine cytomegalovirus (MCMV) infection as models for chronic and latent infections, respectively. We identify CD85k as a Th1-specific co-inhibitory receptor with sustained expression in persistent viral infections and show that recombinant CD85k inhibits LCMV-specific effector T cells. Furthermore, expression of the CD85k ligand ALCAM is induced on LCMV-specific and exhausted T cells during chronic LCMV infection. Finally, we demonstrate that type 1 Tregs arising during chronic LCMV infection suppress Th1 effector cells in an ALCAM-dependent manner. These results extend the current knowledge of Treg specialization from acute to persistent viral infections and reveal an important functional role of CD85k in Treg-mediated suppression of type 1 immunity.
Collapse
MESH Headings
- Animals
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Cell Adhesion Molecules, Neuronal/immunology
- Cell Adhesion Molecules, Neuronal/metabolism
- Cell Line
- Cells, Cultured
- Herpesviridae Infections/immunology
- Herpesviridae Infections/metabolism
- Herpesviridae Infections/virology
- Lymphocytic Choriomeningitis/immunology
- Lymphocytic Choriomeningitis/metabolism
- Lymphocytic Choriomeningitis/virology
- Lymphocytic choriomeningitis virus/immunology
- Lymphocytic choriomeningitis virus/physiology
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- Mice, Inbred C57BL
- Muromegalovirus/immunology
- Muromegalovirus/physiology
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/virology
- Th1 Cells/immunology
- Th1 Cells/metabolism
Collapse
Affiliation(s)
- Anna Estrada Brull
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland; (A.E.B.); (F.R.); (F.R.K.); (S.L.-L.)
| | - Felix Rost
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland; (A.E.B.); (F.R.); (F.R.K.); (S.L.-L.)
| | - Josua Oderbolz
- ETH Zurich, Institute of Microbiology, 8093 Zurich, Switzerland; (J.O.); (A.O.)
| | - Florian R. Kirchner
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland; (A.E.B.); (F.R.); (F.R.K.); (S.L.-L.)
- Section of Immunology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Salomé Leibundgut-Landmann
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland; (A.E.B.); (F.R.); (F.R.K.); (S.L.-L.)
- Section of Immunology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Annette Oxenius
- ETH Zurich, Institute of Microbiology, 8093 Zurich, Switzerland; (J.O.); (A.O.)
| | - Nicole Joller
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland; (A.E.B.); (F.R.); (F.R.K.); (S.L.-L.)
| |
Collapse
|
19
|
Ikeno Y, Ohara D, Takeuchi Y, Watanabe H, Kondoh G, Taura K, Uemoto S, Hirota K. Foxp3+ Regulatory T Cells Inhibit CCl 4-Induced Liver Inflammation and Fibrosis by Regulating Tissue Cellular Immunity. Front Immunol 2020; 11:584048. [PMID: 33178216 PMCID: PMC7593684 DOI: 10.3389/fimmu.2020.584048] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022] Open
Abstract
Foxp3+ regulatory T (Treg) cells are pivotal in maintaining immunological self-tolerance and tissue homeostasis; however, it remains unclear how tissue Treg cells respond to liver injury and regulate chronic inflammation, which can cause liver fibrosis. We report here that hepatic Treg cells play a critical role in preventing liver pathology by suppressing inflammatory cellular immunity that can promote liver damage and fibrosis. Chronic liver inflammation induced by injections of carbon tetrachloride (CCl4) led to preferential expansion of hepatic Treg cells that prevented liver fibrosis. In contrast, depletion of Treg cells in the CCl4-induced liver fibrosis model exacerbated the severity of liver pathology. Treg depletion unleashed tissue cellular immunity and drove the activation and expansion of the pro-fibrotic IL-4-producing T helper 2 cells, as well as CCR2high Ly-6Chigh inflammatory monocytes/macrophages in the inflamed liver. Although Treg expression of amphiregulin plays a key role in tissue remodeling and repair in various inflammation models, amphiregulin from hepatic Treg cells, the largest producer among liver immune cells, was dispensable for maintaining liver homeostasis and preventing liver fibrosis during CCl4-induced chronic inflammation. Our results indicate that Treg cells control chronic liver inflammation and fibrosis by regulating the aberrant activation and functions of immune effector cells. Harnessing Treg functions, which effectively regulate tissue cellular immunity, may be a therapeutic strategy for preventing and treating liver fibrosis.
Collapse
Affiliation(s)
- Yoshinobu Ikeno
- Department of Hepatobiliary, Pancreas and Transplant Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Daiya Ohara
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yusuke Takeuchi
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hitomi Watanabe
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Gen Kondoh
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Kojiro Taura
- Department of Hepatobiliary, Pancreas and Transplant Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinji Uemoto
- Department of Hepatobiliary, Pancreas and Transplant Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keiji Hirota
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
20
|
Olson KE, Namminga KL, Schwab AD, Thurston MJ, Lu Y, Woods A, Lei L, Shen W, Wang F, Joseph SB, Gendelman HE, Mosley RL. Neuroprotective Activities of Long-Acting Granulocyte-Macrophage Colony-Stimulating Factor (mPDM608) in 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine-Intoxicated Mice. Neurotherapeutics 2020; 17:1861-1877. [PMID: 32638217 PMCID: PMC7851309 DOI: 10.1007/s13311-020-00877-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Loss of dopaminergic neurons along the nigrostriatal axis, neuroinflammation, and peripheral immune dysfunction are the pathobiological hallmarks of Parkinson's disease (PD). Granulocyte-macrophage colony-stimulating factor (GM-CSF) has been successfully tested for PD treatment. GM-CSF is a known immune modulator that induces regulatory T cells (Tregs) and serves as a neuronal protectant in a broad range of neurodegenerative diseases. Due to its short half-life, limited biodistribution, and potential adverse effects, alternative long-acting treatment schemes are of immediate need. A long-acting mouse GM-CSF (mPDM608) was developed through Calibr, a Division of Scripps Research. Following mPDM608 treatment, complete hematologic and chemistry profiles and T-cell phenotypes and functions were determined. Neuroprotective and anti-inflammatory capacities of mPDM608 were assessed in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated mice that included transcriptomic immune profiles. Treatment with a single dose of mPDM608 resulted in dose-dependent spleen and white blood cell increases with parallel enhancements in Treg numbers and immunosuppressive function. A shift in CD4+ T-cell gene expression towards an anti-inflammatory phenotype corresponded with decreased microgliosis and increased dopaminergic neuronal cell survival. mPDM608 elicited a neuroprotective peripheral immune transformation. The observed phenotypic shift and neuroprotective response was greater than observed with recombinant GM-CSF (rGM-CSF) suggesting human PDM608 as a candidate for PD treatment.
Collapse
Affiliation(s)
- Katherine E. Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Krista L. Namminga
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Aaron D. Schwab
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Mackenzie J. Thurston
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Ashley Woods
- Calibr, a Division of Scripps Research, La Jolla, CA 92037 USA
| | - Lei Lei
- Calibr, a Division of Scripps Research, La Jolla, CA 92037 USA
| | - Weijun Shen
- Calibr, a Division of Scripps Research, La Jolla, CA 92037 USA
| | - Feng Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Sean B. Joseph
- Calibr, a Division of Scripps Research, La Jolla, CA 92037 USA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 6898-5880 USA
| | - R. Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 6898-5880 USA
| |
Collapse
|
21
|
Brown CY, Sadlon T, Hope CM, Wong YY, Wong S, Liu N, Withers H, Brown K, Bandara V, Gundsambuu B, Pederson S, Breen J, Robertson SA, Forrest A, Beyer M, Barry SC. Molecular Insights Into Regulatory T-Cell Adaptation to Self, Environment, and Host Tissues: Plasticity or Loss of Function in Autoimmune Disease. Front Immunol 2020; 11:1269. [PMID: 33072063 PMCID: PMC7533603 DOI: 10.3389/fimmu.2020.01269] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 05/19/2020] [Indexed: 12/19/2022] Open
Abstract
There has been much interest in the ability of regulatory T cells (Treg) to switch function in vivo, either as a result of genetic risk of disease or in response to environmental and metabolic cues. The relationship between levels of FOXP3 and functional fitness plays a significant part in this plasticity. There is an emerging role for Treg in tissue repair that may be less dependent on FOXP3, and the molecular mechanisms underpinning this are not fully understood. As a result of detailed, high-resolution functional genomics, the gene regulatory networks and key functional mediators of Treg phenotype downstream of FOXP3 have been mapped, enabling a mechanistic insight into Treg function. This transcription factor-driven programming of T-cell function to generate Treg requires the switching on and off of key genes that form part of the Treg gene regulatory network and raises the possibility that this is reversible. It is plausible that subtle shifts in expression levels of specific genes, including transcription factors and non-coding RNAs, change the regulation of the Treg gene network. The subtle skewing of gene expression initiates changes in function, with the potential to promote chronic disease and/or to license appropriate inflammatory responses. In the case of autoimmunity, there is an underlying genetic risk, and the interplay of genetic and environmental cues is complex and impacts gene regulation networks frequently involving promoters and enhancers, the regulatory elements that control gene expression levels and responsiveness. These promoter–enhancer interactions can operate over long distances and are highly cell type specific. In autoimmunity, the genetic risk can result in changes in these enhancer/promoter interactions, and this mainly impacts genes which are expressed in T cells and hence impacts Treg/conventional T-cell (Tconv) function. Genetic risk may cause the subtle alterations to the responsiveness of gene regulatory networks which are controlled by or control FOXP3 and its target genes, and the application of assays of the 3D organization of chromatin, enabling the connection of non-coding regulatory regions to the genes they control, is revealing the direct impact of environmental/metabolic/genetic risk on T-cell function and is providing mechanistic insight into susceptibility to inflammatory and autoimmune conditions.
Collapse
Affiliation(s)
- Cheryl Y Brown
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Timothy Sadlon
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia.,Women's and Children's Health Network, North Adelaide, SA, Australia
| | | | - Ying Y Wong
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Soon Wong
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Ning Liu
- Bioinformatics Hub, University of Adelaide, Adelaide, SA, Australia
| | - Holly Withers
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Katherine Brown
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Veronika Bandara
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Batjargal Gundsambuu
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Stephen Pederson
- Bioinformatics Hub, University of Adelaide, Adelaide, SA, Australia
| | - James Breen
- Bioinformatics Hub, University of Adelaide, Adelaide, SA, Australia
| | - Sarah Anne Robertson
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Alistair Forrest
- QEII Medical Centre and Centre for Medical Research, Harry Perkins Institute of Medical Research, Murdoch, WA, Australia
| | - Marc Beyer
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Simon Charles Barry
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia.,Women's and Children's Health Network, North Adelaide, SA, Australia
| |
Collapse
|
22
|
Nakamura R, Yoshizawa A, Moriyasu T, Deloer S, Senba M, Kikuchi M, Koyasu S, Moro K, Hamano S. Group 2 Innate Lymphoid Cells Exacerbate Amebic Liver Abscess in Mice. iScience 2020; 23:101544. [PMID: 33083770 PMCID: PMC7509001 DOI: 10.1016/j.isci.2020.101544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 07/24/2020] [Accepted: 09/04/2020] [Indexed: 01/21/2023] Open
Abstract
Entamoeba histolytica, a protozoan parasite in the lumen of the human large intestine, occasionally spreads to the liver and induces amebic liver abscesses (ALAs). Upon infection with E. histolytica, high levels of type 2 cytokines are induced in the liver early after infection. However, the sources and functions of these initial type 2 cytokines in ALA formation remain unclear. In this study, we examined the roles of group 2 innate lymphoid cells (ILC2s) in ALA formation. Hepatic ILC2 numbers were significantly increased and they produced robust levels of IL-5. The in vivo transfer of ILC2s into Rag2−/−common γ chain (γc)−/− KO mice aggravated ALA formation accompanied by eosinophilia and neutrophilia. Furthermore, IL-33-deficient mice and IL-5-neutralized mice had less ALA formations. These results suggest that ILC2s contribute to exacerbating the pathogenesis of ALA by producing early type 2 cytokines and promoting the accumulation of eosinophils and neutrophils in the liver. ILC2s exacerbate ALA by promoting the accumulation of eosinophils and neutrophils Hepatic ILC2s are increased and the main source of IL-5 in the early phase of ALA Hepatic ILC2s localize with IL-33+ cells in the inflammatory areas of ALA IL-33 is a trigger of ILC2-mediated ALA formation
Collapse
Affiliation(s)
- Risa Nakamura
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,Graduate School of Biomedical Sciences Doctoral Leadership Program, Nagasaki University, Nagasaki, Japan.,The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan
| | - Akihiro Yoshizawa
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.,Department of Cardiovascular Medicine, International University of Health and Welfare (IUHW), School of Medicine, Chiba, Japan
| | - Taeko Moriyasu
- Graduate School of Biomedical Sciences Doctoral Leadership Program, Nagasaki University, Nagasaki, Japan.,The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan.,Kenya Research Station, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Sharmina Deloer
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,Graduate School of Biomedical Sciences Doctoral Leadership Program, Nagasaki University, Nagasaki, Japan.,The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan.,Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and infectious Diseases (NIAID), NIH, Maryland, USA
| | - Masachika Senba
- The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan.,Department of Pathology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Mihoko Kikuchi
- The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan.,Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Shigeo Koyasu
- Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.,Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuyo Moro
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.,Laboratory for Innate Immune Systems, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinjiro Hamano
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,Graduate School of Biomedical Sciences Doctoral Leadership Program, Nagasaki University, Nagasaki, Japan.,The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan
| |
Collapse
|
23
|
Cui G, Yuan A, Li Z, Goll R, Florholmen J. ST2 and regulatory T cells in the colorectal adenoma/carcinoma microenvironment: implications for diseases progression and prognosis. Sci Rep 2020; 10:5892. [PMID: 32246094 PMCID: PMC7125220 DOI: 10.1038/s41598-020-62502-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 03/13/2020] [Indexed: 02/07/2023] Open
Abstract
ST2 (also known as IL1RL1) is the critical functional receptor for interleukin (IL)-33 in stimulating regulatory T cell (Treg) expansion and function in physiological and pathological conditions. We examined the correlation between ST2 cell expression and FoxP3 positive Tregs in both colorectal adenoma and cancer (CRC) microenvironment by real-time PCR, immunohistochemistry (IHC) and double immunofluorescences. The clinicopathological and prognostic significance of cellular ST2-positive cells and FoxP3-positive Tregs in patients with adenoma and CRC were evaluated. Real-time PCR results revealed increased expression levels of ST2 and FoxP3 mRNAs in both adenoma and CRC tissues as compared with control tissues. IHC analysis confirmed increased densities of ST2-positive cells in both the adenoma/CRC epithelium and stroma, which show a close positive linear association with the densities of FoxP3-positive Tregs in respective compartments. Pathological feature analysis showed that densities of ST2-positive cells in the tumor stroma were notably associated with degree of dysplastic grading in patients with adenoma, and disease stages and lymph node metastasis in patients with CRC. Kaplan-Meier survival curves suggested that CRC patients with high densities of ST2-positive cells in the stroma tend to have a shorter overall survival. We therefore concluded that increased densities of ST2-postive cells relate to Treg accumulation within the adenoma/CRC microenvironment, suggesting the IL-33/ST2 pathway as a potential contributor for immunosuppressive milieu formation that impact disease stage and prognosis in patients with CRC.
Collapse
Affiliation(s)
- Guanglin Cui
- Department of Gastroenterology, the Second Affiliated Hospital of Zhengzhou University, Henan, China. .,Faculty of Health Science, Nord University at Campus Levanger, Levanger, Norway.
| | - Aping Yuan
- Department of Gastroenterology, the Second Affiliated Hospital of Zhengzhou University, Henan, China
| | - Zhenfeng Li
- Department of Gastroenterology, the Second Affiliated Hospital of Zhengzhou University, Henan, China
| | - Rasmus Goll
- Department of Gastroenterology & Nutrition, University Hospital of North Norway, Tromsø, University of Tromsø, Tromsø, Norway
| | - Jon Florholmen
- Department of Gastroenterology & Nutrition, University Hospital of North Norway, Tromsø, University of Tromsø, Tromsø, Norway
| |
Collapse
|
24
|
Campbell C, Rudensky A. Roles of Regulatory T Cells in Tissue Pathophysiology and Metabolism. Cell Metab 2020; 31:18-25. [PMID: 31607562 PMCID: PMC7657366 DOI: 10.1016/j.cmet.2019.09.010] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/12/2019] [Accepted: 09/13/2019] [Indexed: 12/12/2022]
Abstract
Regulatory T (Treg) cells expressing the X-chromosome-encoded transcription factor Foxp3 represent a specialized immunosuppressive lineage with a well-recognized, essential function in preventing fatal autoimmunity and inflammation. Recent studies revealed that Treg cells can also exert systemic effects on metabolism and partake in tissue repair, suggesting a dual role for these cells in serving and protecting tissues. Here, we review multiple means by which Treg cells support tissue function and organismal homeostasis.
Collapse
Affiliation(s)
- Clarissa Campbell
- Howard Hughes Medical Institute and Immunology Program, Sloan Kettering Institute, New York, NY 10065, USA; Ludwig Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Alexander Rudensky
- Howard Hughes Medical Institute and Immunology Program, Sloan Kettering Institute, New York, NY 10065, USA; Ludwig Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10021, USA.
| |
Collapse
|
25
|
Lu YY, Liu XL, Huang Y, Liao Y, Xi T, Zhang YN, Zhang LL, Shu SN, Fang F. Short-lived AIM2 Inflammasome Activation Relates to Chronic MCMV Infection in BALB/c Mice. Curr Med Sci 2019; 39:899-905. [PMID: 31845220 DOI: 10.1007/s11596-019-2121-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 10/20/2019] [Indexed: 12/19/2022]
Abstract
Absent in melanoma 2 (AIM2) inflammasome is a crucial link bridging the innate host defense and the subsequent adaptive immunity when activated by exogenous double stranded DNA (dsDNA). Through establishing models of disseminated murine cytomegalovirus (MCMV) infection in BALB/c and C57BL/6 mice, we evaluated dynamic expression of AIM2 inflammasome components and its relationship with pathological damage and viral replication, trying to figure out whether AIM2 inflammasome is related to the chronic mechanism of MCMV. BALB/c and C57BL/6 mice were sacrificed on day 0, 1, 3, 7, 14 and 28 post infection. Expression levels of AIM2, pro-caspase-1, caspase-1 p20, pro-IL1β and mature IL1β in primary peritoneal macrophages (PMs) and spleens were detected by Western blotting. Contents of IL18 in the serum were detected by ELISA. Pathological examinations of livers were performed, and mRNA levels of MCMV glycoprotein B (gB) in salivary glands also assessed. Results showed that expression levels of AIM2 in PMs and spleens of C57BL/6 mice increased on day 3, even continued to day 28; caspase-1 p20 and mature IL1β increased on day 7, 14 and 28; the persistently high expression of IL18 in the serum started on day 1, showing a double peak curve. As for BALB/c mice, expression of AIM2 in PMs increased on day 1 and day 7, while contents of AIM2 in spleens increased on day 1 and day 3; caspase-1 p20 and mature IL1β merely increased 7 days fter infection. Thereafter, expression levels of AIM2, caspase-1 p20, mature IL1β and IL18 were limited; the duration of AIM2 inflammasome activation in BALB/c mice was much shorter than that in C57BL/6 mice. The severer pathological damage and more viral replications in BALB/c mice further proved the deficient antiviral immunity to MCMV. In conclusion, the activation of AIM2 inflammasome in BALB/c mice was short-lived, which is quite possibly related to the chronicity of MCMV infection.
Collapse
Affiliation(s)
- Yuan-Yuan Lu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xing-Lou Liu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuan Huang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yi Liao
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ting Xi
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ya-Nan Zhang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lin-Lin Zhang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sai-Nan Shu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Feng Fang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
26
|
Lung T, Sakem B, Risch L, Würzner R, Colucci G, Cerny A, Nydegger U. The complement system in liver diseases: Evidence-based approach and therapeutic options. J Transl Autoimmun 2019; 2:100017. [PMID: 32743505 PMCID: PMC7388403 DOI: 10.1016/j.jtauto.2019.100017] [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: 07/21/2019] [Revised: 09/05/2019] [Accepted: 09/10/2019] [Indexed: 12/12/2022] Open
Abstract
Complement is usually seen to largely originate from the liver to accomplish its tasks systemically - its return to the production site has long been underestimated. Recent progress in genomics, therapeutic effects on complement, standardised possibilities in medical laboratory tests and involvement of complosome brings the complement system with its three major functions of opsonization, cytolysis and phagocytosis back to liver biology and pathology. The LOINC™ system features 20 entries for the C3 component of complement to anticipate the application of artificial intelligence data banks algorythms of which are fed with patient-specific data connected to standard lab assays for liver function. These advancements now lead to increased vigilance by clinicians. This reassessment article will further elucidate the distribution of synthesis sites to the three germ layer-derived cell systems and the role complement now known to play in embryogenesis, senescence, allotransplantation and autoimmune disease. This establishes the liver as part of the gastro-intestinal system in connection with nosological entities never thought of, such as the microbiota-liver-brain axis. In neurological disease etiology infectious and autoimmune hepatitis play an important role in the context of causative viz reactive complement activation. The mosaic of autoimmunity, i.e. multiple combinations of the many factors producing varying clinical pictures, leads to the manifold facets of liver autoimmunity.
Collapse
Affiliation(s)
- Thomas Lung
- Labormedizinisches Zentrum Dr. Risch, Lagerstrasse 30, CH-9470, Buchs, Switzerland
| | - Benjamin Sakem
- Labormedizinisches Zentrum Dr. Risch, Waldeggstrasse 37, CH-3097, Liebefeld bei Bern, Switzerland
| | - Lorenz Risch
- Labormedizinisches Zentrum Dr. Risch, Waldeggstrasse 37, CH-3097, Liebefeld bei Bern, Switzerland
| | - Reinhard Würzner
- Medical University Innsbruck, Division of Hygiene & Medical Microbiology, Department of Hygiene, Microbiology and Public Health, Schöpfstrasse 41, A-6020, Innsbruck, Austria
| | - Giuseppe Colucci
- Clinica Luganese Moncucco, Lugano, Via Moncucco, CH-6900, Lugano, Switzerland
- Faculty of Medicine, University of Basel, Basel, Switzerland
| | - Andreas Cerny
- Epatocentro Ticino, Via Soldino 5, CH-6900, Lugano, Switzerland
| | - Urs Nydegger
- Labormedizinisches Zentrum Dr. Risch, Waldeggstrasse 37, CH-3097, Liebefeld bei Bern, Switzerland
| |
Collapse
|
27
|
Jergović M, Contreras NA, Nikolich-Žugich J. Impact of CMV upon immune aging: facts and fiction. Med Microbiol Immunol 2019; 208:263-269. [PMID: 31004198 PMCID: PMC6635032 DOI: 10.1007/s00430-019-00605-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 03/30/2019] [Indexed: 12/28/2022]
Abstract
Aging is accompanied by significant defects in immunity and compromised responses to new, previously unencountered microbial pathogens. Most humans carry several persistent or latent viruses as they age, interacting with the host immune systems for years. In that context maybe the most studied persistent virus is Cytomegalovirus, infamous for its ability to recruit very large T cell responses which increase with age and to simultaneously evade elimination by the immune system. Here we will address how lifelong CMV infection and the immunological burden of its control might affect immune reactivity and health of the host over time.
Collapse
Affiliation(s)
- Mladen Jergović
- Department of Immunobiology and the University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85718, USA
| | - Nico A Contreras
- Department of Immunobiology and the University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85718, USA
| | - Janko Nikolich-Žugich
- Department of Immunobiology and the University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85718, USA.
- University of Arizona College of Medicine-Tucson, 1501 N Campbell Ave, P.O. Box 221245, Tucson, AZ, 85724, USA.
| |
Collapse
|
28
|
Copsel S, Wolf D, Komanduri KV, Levy RB. The promise of CD4 +FoxP3 + regulatory T-cell manipulation in vivo: applications for allogeneic hematopoietic stem cell transplantation. Haematologica 2019; 104:1309-1321. [PMID: 31221786 PMCID: PMC6601084 DOI: 10.3324/haematol.2018.198838] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022] Open
Abstract
CD4+FoxP3+ regulatory T cells (Tregs) are a non-redundant population critical for the maintenance of self-tolerance. Over the past decade, the use of these cells for therapeutic purposes in transplantation and autoimmune disease has emerged based on their capacity to inhibit immune activation. Basic science discoveries have led to identifying key receptors on Tregs that can regulate their proliferation and function. Notably, the understanding that IL-2 signaling is crucial for Treg homeostasis promoted the hypothesis that in vivo IL-2 treatment could provide a strategy to control the compartment. The use of low-dose IL-2 in vivo was shown to selectively expand Tregs versus other immune cells. Interestingly, a number of other Treg cell surface proteins, including CD28, CD45, IL-33R and TNFRSF members, have been identified which can also induce activation and proliferation of this population. Pre-clinical studies have exploited these observations to prevent and treat mice developing autoimmune diseases and graft-versus-host disease post-allogeneic hematopoietic stem cell transplantation. These findings support the development of translational strategies to expand Tregs in patients. Excitingly, the use of low-dose IL-2 for patients suffering from graft-versus-host disease and autoimmune disease has demonstrated increased Treg levels together with beneficial outcomes. To date, promising pre-clinical and clinical studies have directly targeted Tregs and clearly established the ability to increase their levels and augment their function in vivo. Here we review the evolving field of in vivo Treg manipulation and its application to allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
| | | | - Krishna V Komanduri
- Department of Microbiology and Immunology.,Sylvester Comprehensive Cancer Center.,Division of Transplantation and Cellular Therapy, Department of Medicine
| | - Robert B Levy
- Department of Microbiology and Immunology .,Division of Transplantation and Cellular Therapy, Department of Medicine.,Department of Ophthalmology, Miller School of Medicine, University of Miami, FL, USA
| |
Collapse
|
29
|
Caspase-8-dependent control of NK- and T cell responses during cytomegalovirus infection. Med Microbiol Immunol 2019; 208:555-571. [PMID: 31098689 DOI: 10.1007/s00430-019-00616-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 04/17/2019] [Indexed: 12/26/2022]
Abstract
Caspase-8 (CASP8) impacts antiviral immunity in expected as well as unexpected ways. Mice with combined deficiency in CASP8 and RIPK3 cannot support extrinsic apoptosis or RIPK3-dependent programmed necrosis, enabling studies of CASP8 function without complications of unleashed necroptosis. These extrinsic cell death pathways are naturally targeted by murine cytomegalovirus (MCMV)-encoded cell death suppressors, showing they are key to cell-autonomous host defense. Remarkably, Casp8-/-Ripk3-/-, Ripk1-/-Casp8-/-Ripk3-/- and Casp8-/-Ripk3K51A/K51A mice mount robust antiviral T cell responses to control MCMV infection. Studies in Casp8-/-Ripk3-/- mice show that CASP8 restrains expansion of MCMV-specific natural killer (NK) and CD8 T cells without compromising contraction or immune memory. Infected Casp8-/-Ripk3-/- or Casp8-/-Ripk3K51A/K51A mice have higher levels of virus-specific NK cells and CD8 T cells compared to matched RIPK3-deficient littermates or WT mice. CASP8, likely acting downstream of Fas death receptor, dampens proliferation of CD8 T cells during expansion. Importantly, contraction proceeds unimpaired in the absence of extrinsic death pathways owing to intact Bim-dependent (intrinsic) apoptosis. CD8 T cell memory develops in Casp8-/-Ripk3-/- mice, but memory inflation characteristic of MCMV infection is not sustained in the absence of CASP8 function. Despite this, Casp8-/-Ripk3-/- mice are immune to secondary challenge. Interferon (IFN)γ is recognized as a key cytokine for adaptive immune control of MCMV. Ifngr-/-Casp8-/-Ripk3-/- mice exhibit increased lifelong persistence in salivary glands as well as lungs compared to Ifngr-/- and Casp8-/-Ripk3-/- mice. Thus, mice deficient in CASP8 and RIPK3 are more dependent on IFNγ mechanisms for sustained T cell immune control of MCMV. Overall, appropriate NK- and T cell immunity to MCMV is dependent on host CASP8 function independent of RIPK3-regulated pathways.
Collapse
|
30
|
Cui G, Li Z, Ren J, Yuan A. IL-33 in the tumor microenvironment is associated with the accumulation of FoxP3-positive regulatory T cells in human esophageal carcinomas. Virchows Arch 2019; 475:579-586. [DOI: 10.1007/s00428-019-02579-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/07/2019] [Accepted: 04/18/2019] [Indexed: 12/13/2022]
|
31
|
Mantovani A, Dinarello CA, Molgora M, Garlanda C. Interleukin-1 and Related Cytokines in the Regulation of Inflammation and Immunity. Immunity 2019; 50:778-795. [PMID: 30995499 PMCID: PMC7174020 DOI: 10.1016/j.immuni.2019.03.012] [Citation(s) in RCA: 624] [Impact Index Per Article: 124.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/08/2019] [Accepted: 03/14/2019] [Indexed: 02/06/2023]
Abstract
Forty years after its naming, interleukin-1 (IL-1) is experiencing a renaissance brought on by the growing understanding of its context-dependent roles and advances in the clinic. Recent studies have identified important roles for members of the IL-1 family-IL-18, IL-33, IL-36, IL-37, and IL-38-in inflammation and immunity. Here, we review the complex functions of IL-1 family members in the orchestration of innate and adaptive immune responses and their diversity and plasticity. We discuss the varied roles of IL-1 family members in immune homeostasis and their contribution to pathologies, including autoimmunity and auto-inflammation, dysmetabolism, cardiovascular disorders, and cancer. The trans-disease therapeutic activity of anti-IL-1 strategies argues for immunity and inflammation as a metanarrative of modern medicine.
Collapse
Affiliation(s)
- Alberto Mantovani
- IRCCS Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano Milan, Italy; Humanitas University, via Rita Levi Montalcini, 20090 Pieve Emanuele Milan, Italy; William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA; Department of Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Martina Molgora
- IRCCS Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano Milan, Italy
| | - Cecilia Garlanda
- IRCCS Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano Milan, Italy; Humanitas University, via Rita Levi Montalcini, 20090 Pieve Emanuele Milan, Italy.
| |
Collapse
|
32
|
Zaiss DM, Minutti CM, Knipper JA. Immune- and non-immune-mediated roles of regulatory T-cells during wound healing. Immunology 2019; 157:190-197. [PMID: 30866049 DOI: 10.1111/imm.13057] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 01/29/2019] [Indexed: 12/16/2022] Open
Abstract
The immune system has a well-established contribution to tissue homeostasis and wound healing. However, in many cases immune responses themselves can cause severe tissue damage. Thus, the question arose to which extent cells of the immune system directly contribute to the process of wound healing and to which extent the resolution of excessive immune responses may indirectly contribute to wound healing. FoxP3-expressing CD4 T-cells, so-called regulatory T-cells (Tregs ), have an important contribution in the regulation of immune responses; and, in recent years, it has been suggested that Tregs next to an immune-regulatory, 'damage-limiting' function may also have an immune-independent 'damage-resolving' direct role in wound healing. In particular, the release of the epidermal growth factor-like growth factor Amphiregulin by tissue-resident Tregs during wound repair suggested such a function. Our recent findings have now revealed that Amphiregulin induces the local release of bio-active transforming growth factor (TGF)β, a cytokine involved both in immune regulation as well as in the process of wound repair. In light of these findings, we discuss whether, by locally activating TGFβ, Treg -derived Amphiregulin may contribute to both wound repair and immune suppression. Furthermore, we propose that Treg -derived Amphiregulin in an autocrine way may enable an IL-33-mediated survival and expansion of tissue-resident Tregs upon injury. Furthermore, Treg -derived Amphiregulin may contribute to a constitutive, low-level release of bio-active TGFβ within tissues, leading to continuous tissue regeneration and to an immune-suppressive environment, which may keep inflammation-prone tissues in an homeostatic state.
Collapse
Affiliation(s)
- Dietmar M Zaiss
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Carlos M Minutti
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK.,Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Johanna A Knipper
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
33
|
Alvarez F, Fritz JH, Piccirillo CA. Pleiotropic Effects of IL-33 on CD4 + T Cell Differentiation and Effector Functions. Front Immunol 2019; 10:522. [PMID: 30949175 PMCID: PMC6435597 DOI: 10.3389/fimmu.2019.00522] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/26/2019] [Indexed: 12/16/2022] Open
Abstract
IL-33, a member of the IL-1 family of cytokines, was originally described in 2005 as a promoter of type 2 immune responses. However, recent evidence reveals a more complex picture. This cytokine is released locally as an alarmin upon cellular damage where innate cell types respond to IL-33 by modulating their differentiation and influencing the polarizing signals they provide to T cells at the time of antigen presentation. Moreover, the prominent expression of the IL-33 receptor, ST2, on GATA3+ T helper 2 cells (TH2) demonstrated that IL-33 could have a direct impact on T cells. Recent observations reveal that T-bet+ TH1 cells and Foxp3+ regulatory T (TREG) cells can also express the ST2 receptor, either transiently or permanently. As such, IL-33 can have a direct effect on the dynamics of T cell populations. As IL-33 release was shown to play both an inflammatory and a suppressive role, understanding the complex effect of this cytokine on T cell homeostasis is paramount. In this review, we will focus on the factors that modulate ST2 expression on T cells, the effect of IL-33 on helper T cell responses and the role of IL-33 on TREG cell function.
Collapse
Affiliation(s)
- Fernando Alvarez
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, The Research Institute of the McGill University Health Center, Montréal, QC, Canada
- Centre of Excellence in Translational Immunology, Montréal, QC, Canada
| | - Jörg H. Fritz
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Centre of Excellence in Translational Immunology, Montréal, QC, Canada
- McGill University Research Center on Complex Traits, McGill University, Montréal, QC, Canada
| | - Ciriaco A. Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, The Research Institute of the McGill University Health Center, Montréal, QC, Canada
- Centre of Excellence in Translational Immunology, Montréal, QC, Canada
- McGill University Research Center on Complex Traits, McGill University, Montréal, QC, Canada
| |
Collapse
|
34
|
Lam AJ, MacDonald KN, Pesenacker AM, Juvet SC, Morishita KA, Bressler B, Pan JG, Sidhu SS, Rioux JD, Levings MK. Innate Control of Tissue-Reparative Human Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2019; 202:2195-2209. [PMID: 30850479 DOI: 10.4049/jimmunol.1801330] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/23/2019] [Indexed: 12/17/2022]
Abstract
Regulatory T cell (Treg) therapy is a potential curative approach for a variety of immune-mediated conditions, including autoimmunity and transplantation, in which there is pathological tissue damage. In mice, IL-33R (ST2)-expressing Tregs mediate tissue repair by producing the growth factor amphiregulin, but whether similar tissue-reparative Tregs exist in humans remains unclear. We show that human Tregs in blood and multiple tissue types produced amphiregulin, but this was neither a unique feature of Tregs nor selectively upregulated in tissues. Human Tregs in blood, tonsil, synovial fluid, colon, and lung tissues did not express ST2, so ST2+ Tregs were engineered via lentiviral-mediated overexpression, and their therapeutic potential for cell therapy was examined. Engineered ST2+ Tregs exhibited TCR-independent, IL-33-stimulated amphiregulin expression and a heightened ability to induce M2-like macrophages. The finding that amphiregulin-producing Tregs have a noneffector phenotype and are progressively lost upon TCR-induced proliferation and differentiation suggests that the tissue repair capacity of human Tregs may be an innate function that operates independently from their classical suppressive function.
Collapse
Affiliation(s)
- Avery J Lam
- Department of Surgery, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada.,BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Katherine N MacDonald
- BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.,Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Anne M Pesenacker
- Department of Surgery, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada.,BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Stephen C Juvet
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, Ontario M5G 2C4, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Kimberly A Morishita
- BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada.,Division of Rheumatology, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Brian Bressler
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, British Columbia V6Z 1Y6, Canada
| | | | - James G Pan
- Toronto Recombinant Antibody Centre, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Sachdev S Sidhu
- Toronto Recombinant Antibody Centre, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - John D Rioux
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec H3T 1J4, Canada; and.,Montreal Heart Institute, Montreal, Quebec H1T 1C8, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada; .,BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| |
Collapse
|
35
|
Sun P, Yu F, Lu J, Zhang M, Wang H, Xu D, Lu L. In vivo effects of neomycin sulfate on non-specific immunity, oxidative damage and replication of cyprinid herpesvirus 2 in crucian carp (Carassius auratus gibelio). AQUACULTURE AND FISHERIES 2019. [DOI: 10.1016/j.aaf.2018.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
36
|
McLaren JE, Clement M, Marsden M, Miners KL, Llewellyn-Lacey S, Grant EJ, Rubina A, Gimeno Brias S, Gostick E, Stacey MA, Orr SJ, Stanton RJ, Ladell K, Price DA, Humphreys IR. IL-33 Augments Virus-Specific Memory T Cell Inflation and Potentiates the Efficacy of an Attenuated Cytomegalovirus-Based Vaccine. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 202:943-955. [PMID: 30635396 PMCID: PMC6341181 DOI: 10.4049/jimmunol.1701757] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 11/21/2018] [Indexed: 01/01/2023]
Abstract
Candidate vaccines designed to generate T cell-based immunity are typically vectored by nonpersistent viruses, which largely fail to elicit durable effector memory T cell responses. This limitation can be overcome using recombinant strains of CMV. Proof-of-principle studies have demonstrated the potential benefits of this approach, most notably in the SIV model, but safety concerns require the development of nonreplicating alternatives with comparable immunogenicity. In this study, we show that IL-33 promotes the accumulation and recall kinetics of circulating and tissue-resident memory T cells in mice infected with murine CMV. Using a replication-deficient murine CMV vector, we further show that exogenous IL-33 boosts vaccine-induced memory T cell responses, which protect against subsequent heterologous viral challenge. These data suggest that IL-33 could serve as a useful adjuvant to improve the efficacy of vaccines based on attenuated derivatives of CMV.
Collapse
Affiliation(s)
- James E McLaren
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom;
| | - Mathew Clement
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Morgan Marsden
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Kelly L Miners
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Sian Llewellyn-Lacey
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Emma J Grant
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; and
| | - Anzelika Rubina
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Silvia Gimeno Brias
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Emma Gostick
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Maria A Stacey
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Selinda J Orr
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Richard J Stanton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Ian R Humphreys
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| |
Collapse
|
37
|
Clement M, Humphreys IR. Cytokine-Mediated Induction and Regulation of Tissue Damage During Cytomegalovirus Infection. Front Immunol 2019; 10:78. [PMID: 30761144 PMCID: PMC6362858 DOI: 10.3389/fimmu.2019.00078] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 01/11/2019] [Indexed: 12/25/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a β-herpesvirus with high sero-prevalence within the human population. Primary HCMV infection and life-long carriage are typically asymptomatic. However, HCMV is implicated in exacerbation of chronic conditions and associated damage in individuals with intact immune systems. Furthermore, HCMV is a significant cause of morbidity and mortality in the immunologically immature and immune-compromised where disease is associated with tissue damage. Infection-induced inflammation, including robust cytokine responses, is a key component of pathologies associated with many viruses. Despite encoding a large number of immune-evasion genes, HCMV also triggers the induction of inflammatory cytokine responses during infection. Thus, understanding how cytokines contribute to CMV-induced pathologies and the mechanisms through which they are regulated may inform clinical management of disease. Herein, we discuss our current understanding based on clinical observation and in vivo modeling of disease of the role that cytokines play in CMV pathogenesis. Specifically, in the context of the different tissues and organs in which CMV replicates, we give a broad overview of the beneficial and adverse effects that cytokines have during infection and describe how cytokine-mediated tissue damage is regulated. We discuss the implications of findings derived from mice and humans for therapeutic intervention strategies and our understanding of how host genetics may influence the outcome of CMV infections.
Collapse
Affiliation(s)
- Mathew Clement
- Division of Infection and Immunity/Systems Immunity University Research Institute, Cardiff, United Kingdom
| | - Ian R Humphreys
- Division of Infection and Immunity/Systems Immunity University Research Institute, Cardiff, United Kingdom
| |
Collapse
|
38
|
Pavlovic S, Petrovic I, Jovicic N, Ljujic B, Miletic Kovacevic M, Arsenijevic N, Lukic ML. IL-33 Prevents MLD-STZ Induction of Diabetes and Attenuate Insulitis in Prediabetic NOD Mice. Front Immunol 2018; 9:2646. [PMID: 30498495 PMCID: PMC6249384 DOI: 10.3389/fimmu.2018.02646] [Citation(s) in RCA: 13] [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: 05/28/2018] [Accepted: 10/26/2018] [Indexed: 12/13/2022] Open
Abstract
Type 1 diabetes is an autoimmune disease caused by the immune-mediated destruction of pancreatic β-cells. Prevention of type 1 diabetes requires early intervention in the autoimmune process against beta-cells of the pancreatic islets of Langerhans, which is believed to result from disordered immunoregulation. CD4+Foxp3+ regulatory T cells (Tregs) participate as one of the most important cell types in limiting the autoimmune process. The aim of this study was to investigate the effect of exogenous IL-33 in multiple low dose streptozotocin (MLD-STZ) induced diabetes and to delineate its role in the induction of protective Tregs in an autoimmune attack. C57BL/6 mice were treated i. p. with five doses of 40 mg/kg STZ and 0.4 μg rIL-33 four times, starting from day 0, 6, or 12 every second day from the day of disease induction. 16 weeks old NOD mice were treated with 6 injections of 0.4 μg/mouse IL-33 (every second day). Glycemia and glycosuria were measured and histological parameters in pancreatic islets were evaluated at the end of experiments. Cellular make up of the pancreatic lymph nodes and islets were evaluated by flow cytometry. IL-33 given simultaneously with the application of STZ completely prevented the development of hyperglycemia, glycosuria and profoundly attenuated mononuclear cell infiltration. IL-33 treatment was accompanied by higher number of IL-13 and IL-5 producing CD4+ T cells and increased presence of ST2+Foxp3+ regulatory T cells in pancreatic lymph nodes and islets. Elimination of Tregs abrogated protective effect of IL-33. We provide evidence that exogenous IL-33 completely prevents the development of T cell mediated inflammation in pancreatic islets and consecutive development of diabetes in C57BL/6 mice by facilitating the induction Treg cells. To extend this finding for possible relevance in spontaneous diabetes, we showed that IL-33 attenuate insulitis in prediabetic NOD mice.
Collapse
Affiliation(s)
- Sladjana Pavlovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Ivica Petrovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nemanja Jovicic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Biljana Ljujic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Marina Miletic Kovacevic
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nebojsa Arsenijevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Miodrag L. Lukic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
39
|
Laboratory diagnostics of murine blood for detection of mouse cytomegalovirus (MCMV)-induced hepatitis. Sci Rep 2018; 8:14823. [PMID: 30287927 PMCID: PMC6172243 DOI: 10.1038/s41598-018-33167-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/24/2018] [Indexed: 12/16/2022] Open
Abstract
Mouse models are important and versatile tools to study mechanisms and novel therapies of human disease in vivo. Both, the number and the complexity of murine models are constantly increasing and modification of genes of interest as well as any exogenous challenge may lead to unanticipated biological effects. Laboratory diagnostics of blood samples provide a comprehensive and rapid screening for multiple organ function and are fundamental to detect human disease. Here, we adapt an array of laboratory medicine-based tests commonly used in humans to establish a platform for standardized, multi-parametric, and quality-controlled diagnostics of murine blood samples. We determined sex-dependent reference intervals of 51 commonly used laboratory medicine tests for samples obtained from the C57BL/6J mouse strain. As a proof of principle, we applied these diagnostic tests in a mouse cytomegalovirus (MCMV) infection model to screen for organ damage. Consistent with histopathological findings, plasma concentrations of liver-specific enzymes were elevated, supporting the diagnosis of a virus-induced hepatitis. Plasma activities of aminotransferases correlated with viral loads in livers at various days after MCMV infection and discriminated infected from non-infected animals. This study provides murine blood reference intervals of common laboratory medicine parameters and illustrates the use of these tests for diagnosis of infectious disease in experimental animals.
Collapse
|
40
|
Xu L, Li W, Wang X, Zhang L, Qi Q, Dong L, Wei C, Pu Y, Li Y, Zhu J, Zhou S, Liu F, Chen X, Su C. The IL-33-ST2-MyD88 axis promotes regulatory T cell proliferation in the murine liver. Eur J Immunol 2018; 48:1302-1307. [DOI: 10.1002/eji.201747402] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 03/19/2018] [Accepted: 04/20/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Lei Xu
- Department of Pathogen Biology and Immunology; Jiangsu Key Laboratory of Pathogen Biology; Nanjing Medical University; Nanjing Jiangsu P. R. China
| | - Wei Li
- Department of Pathogen Biology and Immunology; Jiangsu Key Laboratory of Pathogen Biology; Nanjing Medical University; Nanjing Jiangsu P. R. China
| | - Xiaofan Wang
- Department of Pathogen Biology and Immunology; Jiangsu Key Laboratory of Pathogen Biology; Nanjing Medical University; Nanjing Jiangsu P. R. China
| | - Lina Zhang
- Department of Pathogen Biology and Immunology; Jiangsu Key Laboratory of Pathogen Biology; Nanjing Medical University; Nanjing Jiangsu P. R. China
| | - Qianqian Qi
- Department of Pathogen Biology and Immunology; Jiangsu Key Laboratory of Pathogen Biology; Nanjing Medical University; Nanjing Jiangsu P. R. China
| | - Liyang Dong
- Department of Pathogen Biology and Immunology; Jiangsu Key Laboratory of Pathogen Biology; Nanjing Medical University; Nanjing Jiangsu P. R. China
| | - Chuan Wei
- Department of Pathogen Biology and Immunology; Jiangsu Key Laboratory of Pathogen Biology; Nanjing Medical University; Nanjing Jiangsu P. R. China
| | - Yanan Pu
- Department of Pathogen Biology and Immunology; Jiangsu Key Laboratory of Pathogen Biology; Nanjing Medical University; Nanjing Jiangsu P. R. China
| | - Yalin Li
- Department of Pathogen Biology and Immunology; Jiangsu Key Laboratory of Pathogen Biology; Nanjing Medical University; Nanjing Jiangsu P. R. China
| | - Jifeng Zhu
- Department of Pathogen Biology and Immunology; Jiangsu Key Laboratory of Pathogen Biology; Nanjing Medical University; Nanjing Jiangsu P. R. China
| | - Sha Zhou
- Department of Pathogen Biology and Immunology; Jiangsu Key Laboratory of Pathogen Biology; Nanjing Medical University; Nanjing Jiangsu P. R. China
| | - Feng Liu
- Department of Pathogen Biology and Immunology; Jiangsu Key Laboratory of Pathogen Biology; Nanjing Medical University; Nanjing Jiangsu P. R. China
| | - Xiaojun Chen
- Department of Pathogen Biology and Immunology; Jiangsu Key Laboratory of Pathogen Biology; Nanjing Medical University; Nanjing Jiangsu P. R. China
| | - Chuan Su
- Department of Pathogen Biology and Immunology; Jiangsu Key Laboratory of Pathogen Biology; Nanjing Medical University; Nanjing Jiangsu P. R. China
| |
Collapse
|
41
|
Chen Y, Qian J. Increased serum levels of IL-33 and soluble ST2 in neonates with human cytomegalovirus infection. J Med Virol 2018; 90:1383-1388. [PMID: 29663450 DOI: 10.1002/jmv.25187] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 03/31/2018] [Indexed: 01/15/2023]
Affiliation(s)
- Yanru Chen
- Department of Neonatology; Xinhua Hospital; Shanghai Jiaotong University School of Medicine; Shanghai China
| | - Jihong Qian
- Department of Neonatology; Xinhua Hospital; Shanghai Jiaotong University School of Medicine; Shanghai China
| |
Collapse
|
42
|
Fauteux-Daniel S, Viel S, Besson L, Zhang J, Marotel M, Mathieu AL, Pescarmona R, Charrier E, Henry T, Belot A, Walzer T. Deletion of Inflammasome Components Is Not Sufficient To Prevent Fatal Inflammation in Models of Familial Hemophagocytic Lymphohistiocytosis. THE JOURNAL OF IMMUNOLOGY 2018; 200:3769-3776. [PMID: 29695416 DOI: 10.4049/jimmunol.1701628] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/27/2018] [Indexed: 11/19/2022]
Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a severe inflammatory condition that occurs in patients with genetic defects of cytotoxicity (familial HLH [FHL]) or secondary to other immunological disorders such as juvenile idiopathic arthritis. HLH is characterized by elevated levels of serum IL-18 and other cytokines. Moreover, a novel clinical entity has been recently identified in which constitutive NLRC4 inflammasome activation leads to severe HLH. Altogether, these clinical observations suggest that inflammasome activation is a central event in the development of all HLH forms and that inflammasome blockade could alleviate inflammation in FHL patients. To formally address this question, we invalidated genes encoding for Caspase-1 or the inflammasome adapter ASC in perforin-deficient mice that were subsequently infected with lymphocytic or mouse choriomeningitis virus as models of FHL. These deletions nearly abrogated IL-18 production occurring during HLH in all models. However, they did not reduce serum IFN-γ levels at the peak of the inflammatory reaction nor did they modulate inflammatory parameters at mid and late stages or fatal outcome. These data show that inflammasome blockade is not sufficient to prevent cytokine storm and lethality in mouse models of FHL and suggest that different pathophysiological mechanisms underlie HLH in genetic defects of cytotoxicity and genetic forms of inflammasome activation.
Collapse
Affiliation(s)
- Sébastien Fauteux-Daniel
- Centre International de Recherche en Infectiologie, Lyon 69007, France.,Inserm, U1111, Lyon 69007, France.,École Normale Supérieure de Lyon, Lyon 69007, France.,Université Lyon 1, Lyon 69100, France.,CNRS, UMR5308, Centre International de Recherche en Infectiologie, Lyon 69007, France
| | - Sébastien Viel
- Centre International de Recherche en Infectiologie, Lyon 69007, France.,Inserm, U1111, Lyon 69007, France.,École Normale Supérieure de Lyon, Lyon 69007, France.,Université Lyon 1, Lyon 69100, France.,CNRS, UMR5308, Centre International de Recherche en Infectiologie, Lyon 69007, France.,Service de Biologie, Hospices Civils de Lyon, Université Claude-Bernard Lyon 1, Lyon 69310, France; and
| | - Laurie Besson
- Centre International de Recherche en Infectiologie, Lyon 69007, France.,Inserm, U1111, Lyon 69007, France.,École Normale Supérieure de Lyon, Lyon 69007, France.,Université Lyon 1, Lyon 69100, France.,CNRS, UMR5308, Centre International de Recherche en Infectiologie, Lyon 69007, France
| | - Jiang Zhang
- Centre International de Recherche en Infectiologie, Lyon 69007, France.,Inserm, U1111, Lyon 69007, France.,École Normale Supérieure de Lyon, Lyon 69007, France.,Université Lyon 1, Lyon 69100, France.,CNRS, UMR5308, Centre International de Recherche en Infectiologie, Lyon 69007, France
| | - Marie Marotel
- Centre International de Recherche en Infectiologie, Lyon 69007, France.,Inserm, U1111, Lyon 69007, France.,École Normale Supérieure de Lyon, Lyon 69007, France.,Université Lyon 1, Lyon 69100, France.,CNRS, UMR5308, Centre International de Recherche en Infectiologie, Lyon 69007, France
| | - Anne-Laure Mathieu
- Centre International de Recherche en Infectiologie, Lyon 69007, France.,Inserm, U1111, Lyon 69007, France.,École Normale Supérieure de Lyon, Lyon 69007, France.,Université Lyon 1, Lyon 69100, France.,CNRS, UMR5308, Centre International de Recherche en Infectiologie, Lyon 69007, France
| | - Rémi Pescarmona
- Centre International de Recherche en Infectiologie, Lyon 69007, France.,Inserm, U1111, Lyon 69007, France.,École Normale Supérieure de Lyon, Lyon 69007, France.,Université Lyon 1, Lyon 69100, France.,CNRS, UMR5308, Centre International de Recherche en Infectiologie, Lyon 69007, France.,Service de Biologie, Hospices Civils de Lyon, Université Claude-Bernard Lyon 1, Lyon 69310, France; and
| | - Emily Charrier
- Centre International de Recherche en Infectiologie, Lyon 69007, France.,Inserm, U1111, Lyon 69007, France.,École Normale Supérieure de Lyon, Lyon 69007, France.,Université Lyon 1, Lyon 69100, France.,CNRS, UMR5308, Centre International de Recherche en Infectiologie, Lyon 69007, France
| | - Thomas Henry
- Centre International de Recherche en Infectiologie, Lyon 69007, France.,Inserm, U1111, Lyon 69007, France.,École Normale Supérieure de Lyon, Lyon 69007, France.,Université Lyon 1, Lyon 69100, France.,CNRS, UMR5308, Centre International de Recherche en Infectiologie, Lyon 69007, France
| | - Alexandre Belot
- Centre International de Recherche en Infectiologie, Lyon 69007, France.,Inserm, U1111, Lyon 69007, France.,École Normale Supérieure de Lyon, Lyon 69007, France.,Université Lyon 1, Lyon 69100, France.,CNRS, UMR5308, Centre International de Recherche en Infectiologie, Lyon 69007, France.,Service de Néphrologie-Rhumatologie-Dermatologie Pédiatriques, Hospices Civils de Lyon, Université Claude-Bernard Lyon 1, Lyon 69677, France
| | - Thierry Walzer
- Centre International de Recherche en Infectiologie, Lyon 69007, France; .,Inserm, U1111, Lyon 69007, France.,École Normale Supérieure de Lyon, Lyon 69007, France.,Université Lyon 1, Lyon 69100, France.,CNRS, UMR5308, Centre International de Recherche en Infectiologie, Lyon 69007, France
| |
Collapse
|
43
|
DaSilva-Arnold SC, Thyagarajan A, Seymour LJ, Yi Q, Bradish JR, Al-Hassani M, Zhou H, Perdue NJ, Nemeth V, Krbanjevic A, Serezani APM, Olson MR, Spandau DF, Travers JB, Kaplan MH, Turner MJ. Phenotyping acute and chronic atopic dermatitis-like lesions in Stat6VT mice identifies a role for IL-33 in disease pathogenesis. Arch Dermatol Res 2018; 310:197-207. [PMID: 29368135 PMCID: PMC6198812 DOI: 10.1007/s00403-018-1807-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 12/18/2017] [Accepted: 01/04/2018] [Indexed: 12/27/2022]
Abstract
The Stat6VT mouse model of atopic dermatitis (AD) is induced by T-cell-specific expression of a constitutively active form of the protein signal transducer and activator of transcription 6 (STAT6). Although AD-like lesions are known to develop in Stat6VT mice, this study was designed to determine if these mice develop acute and chronic phases of disease similar to humans. To address this, AD-like lesions from Stat6VT mice were harvested at two different timepoints relative to their onset. Lesions harvested within 1 week after development were defined as acute lesions, and those present for 1 month or more were defined as chronic lesions. Acute and chronic AD-like lesions from Stat6VT mice exhibited histologic findings and cytokine expression patterns similar to acute and chronic AD lesions in humans. Further analysis revealed increased levels of interleukin (IL)-33 transcripts in AD-like lesions compared to Stat6VT nonlesional and wild-type skin controls. Immunofluorescence also revealed increased numbers of IL-33+ keratinocytes in Stat6VT lesional skin and localized IL-33+ keratinocytes to a keratin 5+ subset. Furthermore, AD-like disease was more severe in IL-33-deficient Stat6VT mice compared to IL-33-sufficient Stat6VT mice. These studies suggest that Stat6VT mice can serve as a model of acute and chronic AD and that IL-33 may attenuate inflammation in this system.
Collapse
Affiliation(s)
- Sonia C DaSilva-Arnold
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Anita Thyagarajan
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Leroy J Seymour
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Qiaofang Yi
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Joshua R Bradish
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Mohammed Al-Hassani
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hongming Zhou
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Nikolajs J Perdue
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Val Nemeth
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Aleksandar Krbanjevic
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Ana P M Serezani
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Matthew R Olson
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Dan F Spandau
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jeffrey B Travers
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA
| | - Mark H Kaplan
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Matthew J Turner
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA.
| |
Collapse
|
44
|
Dichotomous function of IL-33 in health and disease: From biology to clinical implications. Biochem Pharmacol 2018; 148:238-252. [PMID: 29309756 DOI: 10.1016/j.bcp.2018.01.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/03/2018] [Indexed: 12/13/2022]
Abstract
Interleukin (IL)-33 is a cytokine that is released from epithelial and endothelial cells at barrier surfaces upon tissue stress or damage to operate as an alarmin. IL-33 has been primarily implicated in the induction of T helper (Th) 2 type immune responses. Therefore, IL-33 has attracted a lot of interest as a potential therapeutic target in asthma and other allergic diseases. Over the years, it has become clear that IL-33 has a much broader activity and also contributes to Th1 immunity, expanding the possibilities for therapeutic modulation of IL-33 activity to multiple inflammatory diseases. However, more recently IL-33 has also been shown to mediate immunosuppression and tissue repair by activating regulatory T cells (Treg) and promoting M2 macrophage polarization. These pleiotropic activities of IL-33 illustrate the need for a tight molecular regulation of IL-33 activity, and have to be taken into account when IL-33 or its receptor is targeted for therapeutic modulation. Here we review the multiple molecular mechanisms that regulate IL-33 activity and describe how IL-33 can shape innate and adaptive immune responses by promoting Th1, Th2 and Treg function. Finally, we will discuss the possibilities for therapeutic modulation of IL-33 signaling as well as possible safety issues.
Collapse
|
45
|
Abstract
Human cytomegalovirus (HCMV) encodes numerous proteins and microRNAs that function to evade the immune response and allow the virus to replicate and disseminate in the face of a competent innate and acquired immune system. The establishment of a latent infection by CMV, which if completely quiescent at the level of viral gene expression would represent an ultimate in immune evasion strategies, is not sufficient for lifelong persistence and dissemination of the virus. CMV needs to reactivate and replicate in a lytic cycle of infection in order to disseminate further, which occurs in the face of a fully primed secondary immune response. Without reactivation, latency itself would be redundant for the virus. It is also becoming clear that latency is not a totally quiescent state, but is characterized by limited viral gene expression. Therefore, the virus also needs immune evasion strategies during latency. An effective immune response to CMV is required or viral replication will cause morbidity and ultimately mortality in the host. There is clearly a complex balance between virus immune evasion and host immune recognition over a lifetime. This poses the important question of whether long-term evasion or manipulation of the immune response driven by CMV is detrimental to health. In this meeting report, three groups used the murine model of CMV (MCMV) to examine if the contribution of the virus to immune senescence is set by the (i) initial viral inoculum, (ii) inflation of T cell responses, (iii) or the balance between functionally distinct effector CD4+ T cells. The work of other groups studying the CMV response in humans is discussed. Their work asks whether the ability to make immune responses to new antigens is compromised by (i) age and HCMV carriage, (ii) long-term exposure to HCMV giving rise to an overall immunosuppressive environment and increased levels of latent virus, or (iii) adapted virus mutants (used as potential vaccines) that have the capacity to elicit conventional and unconventional T cell responses.
Collapse
|