1
|
Ren X, Wei Y, Zhao H, Shao J, Zeng F, Wang Z, Li L. A comprehensive review and comparison of L-tryptophan biosynthesis in Saccharomyces cerevisiae and Escherichia coli. Front Bioeng Biotechnol 2023; 11:1261832. [PMID: 38116200 PMCID: PMC10729320 DOI: 10.3389/fbioe.2023.1261832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/22/2023] [Indexed: 12/21/2023] Open
Abstract
L-tryptophan and its derivatives are widely used in the chemical, pharmaceutical, food, and feed industries. Microbial fermentation is the most commonly used method to produce L-tryptophan, which calls for an effective cell factory. The mechanism of L-tryptophan biosynthesis in Escherichia coli, the widely used producer of L-tryptophan, is well understood. Saccharomyces cerevisiae also plays a significant role in the industrial production of biochemicals. Because of its robustness and safety, S. cerevisiae is favored for producing pharmaceuticals and food-grade biochemicals. However, the biosynthesis of L-tryptophan in S. cerevisiae has been rarely summarized. The synthetic pathways and engineering strategies of L-tryptophan in E. coli and S. cerevisiae have been reviewed and compared in this review. Furthermore, the information presented in this review pertains to the existing understanding of how L-tryptophan affects S. cerevisiae's stress fitness, which could aid in developing a novel plan to produce more resilient industrial yeast and E. coli cell factories.
Collapse
Affiliation(s)
- Xinru Ren
- College of Science and Technology, Hebei Agricultural University, Cangzhou, China
| | - Yue Wei
- College of Science and Technology, Hebei Agricultural University, Cangzhou, China
| | - Honglu Zhao
- College of Science and Technology, Hebei Agricultural University, Cangzhou, China
| | - Juanjuan Shao
- College of Science and Technology, Hebei Agricultural University, Cangzhou, China
| | - Fanli Zeng
- College of Life Sciences, Hebei Agricultural University, Baoding, China
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Baoding, China
| | - Zhen Wang
- College of Science and Technology, Hebei Agricultural University, Cangzhou, China
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Baoding, China
| | - Li Li
- College of Science and Technology, Hebei Agricultural University, Cangzhou, China
| |
Collapse
|
2
|
Dou M, Ding C, Zheng B, Deng G, Zhu K, Xu C, Xue W, Ding X, Zheng J, Tian P. Immune-Related Genes for Predicting Future Kidney Graft Loss: A Study Based on GEO Database. Front Immunol 2022; 13:859693. [PMID: 35281025 PMCID: PMC8913884 DOI: 10.3389/fimmu.2022.859693] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/08/2022] [Indexed: 11/22/2022] Open
Abstract
Objective We aimed to identify feature immune-related genes that correlated with graft rejection and to develop a prognostic model based on immune-related genes in kidney transplantation. Methods Gene expression profiles were obtained from the GEO database. The GSE36059 dataset was used as a discovery cohort. Then, differential expression analysis and a machine learning method were performed to select feature immune-related genes. After that, univariate and multivariate Cox regression analyses were used to identify prognosis-related genes. A novel Riskscore model was built based on the results of multivariate regression. The levels of these feature genes were also confirmed in an independent single-cell dataset and other GEO datasets. Results 15 immune-related genes were expressed differently between non-rejection and rejection kidney allografts. Those differentially expressed immune-related genes (DE-IRGs) were mainly associated with immune-related biological processes and pathways. Subsequently, a 5-immune-gene signature was constructed and showed favorable predictive results in the GSE21374 dataset. Recipients were divided into the high-risk and low-risk groups according to the median value of RiskScore. The GO and KEGG analysis indicated that the differentially expressed genes (DEGs) between high-risk and low-risk groups were mainly involved in inflammatory pathways, chemokine-related pathways, and rejection-related pathways. Immune infiltration analysis demonstrated that RiskScore was potentially related to immune infiltration. Kaplan-Meier survival analysis suggested that recipients in the high-risk group had poor graft survival. AUC values of 1- and 3-year graft survival were 0.804 and 0.793, respectively. Conclusion Our data suggest that this immune-related prognostic model had good sensitivity and specificity in predicting the 1- and 3-year kidney graft survival and might act as a useful tool for predicting kidney graft loss.
Collapse
Affiliation(s)
- Meng Dou
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Chenguang Ding
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Bingxuan Zheng
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ge Deng
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Kun Zhu
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Cuixiang Xu
- Center of Shaanxi Provincial Clinical Laboratory, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Wujun Xue
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaoming Ding
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jin Zheng
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Puxun Tian
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Puxun Tian,
| |
Collapse
|
3
|
Heidari F, Razmkhah M, Razban V, Erfani N. Effects of indoleamine 2, 3-dioxygenase (IDO) silencing on immunomodulatory function and cancer-promoting characteristic of adipose-derived mesenchymal stem cells (ASCs). Cell Biol Int 2021; 45:2544-2556. [PMID: 34498786 DOI: 10.1002/cbin.11698] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 08/25/2021] [Accepted: 09/05/2021] [Indexed: 12/28/2022]
Abstract
Indoleamine 2, 3-dioxygenase (IDO) catabolizes tryptophan, mediates immunomodulatory functions, and is released by stromal cells such as mesenchymal stem cells. The aims of this study were to investigate the effects of IDO silencing on immunosuppressive function of adipose-derived mesenchymal stem cells (ASCs), T cells phenotype, and the proliferation/migration of tumor cells. ASCs isolated from adipose tissues of healthy women were transfected with IDO-siRNA. Galectin-3, transforming growth factor-β1, hepatocyte growth factor, and interleukin-10 as immunomodulators were measured in ASCs using qRT-PCR. T cells phenotype, interferon-γ, and interleukin-17 expression were evaluated in peripheral blood lymphocytes (PBLs) cocultured with IDO silenced-ASCs by flow cytometry and qRT-PCR, respectively. Scratch assay was applied to assess the proliferation/migration of MDA-MB-231 cell line. Galectin-3 was upregulated (p ˂ 0.05) while hepatocyte growth factor was downregulated (p ˂ 0.05) in IDO-silenced ASCs compared to control groups. Regulatory T cells were inhibited in PBLs cocultured with IDO-silenced ASCs; also T helper2 was decreased in PBLs cocultured with IDO-silenced ASCs relative to the scramble group. IDO-silenced ASCs caused interferon-γ overexpression but interleukin-17 downregulation in PBLs. The proliferation/migration of MDA-MB-231 was suppressed after exposing to condition media of IDO-silenced ASCs compared with condition media of untransfected (p < 0.01) and scramble-transfected ASCs (p < 0.05). The results exhibited the weakened capacity of IDO-silenced ASCs for suppressing the immune cells and promoting the tumor cells' proliferation/migration. IDO suppression may be utilized as a strategy for cancer treatment. Simultaneous blocking of immunomodulators along with IDO inhibitors may show more effects on boosting the efficiency of immune-based cancer therapies.
Collapse
Affiliation(s)
- Fahimeh Heidari
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboobeh Razmkhah
- School of Medicine, Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasrollah Erfani
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.,School of Medicine, Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
4
|
Increased Expression of Indoleamine 2,3-Dioxygenase (IDO) in Vogt-Koyanagi-Harada (VKH) Disease May Lead to a Shift of T Cell Responses Toward a Treg Population. Inflammation 2021; 43:1780-1788. [PMID: 32435912 DOI: 10.1007/s10753-020-01252-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Previous studies have pointed out that indoleamine 2,3-dioxygenase (IDO), the rate-limiting enzyme initiating tryptophan metabolism, plays a role in the regulation of the immune system. This project was designed to investigate the potential role of IDO in monocyte-derived dendritic cells (moDCs) obtained from active Vogt-Koyanagi-Harada (VKH) disease patients. In this study, we found that the IDO mRNA expression and enzyme activity were increased in active VKH patients as compared with healthy controls and patients in remission. To investigate the role of IDO in immune regulation, an effective inhibitor 1-methyl-L-tryptophan (1-MT) was used to suppress its activity in DCs. The results showed that inhibition of IDO with 1-MT significantly decreased the expression of DC marker CD86. IDO inhibition did not affect the cytokine production of IL-6, IL-1β, TNF-α, IL-10, and TGF-β in DCs. Downregulation of IDO in DCs also led to the reduction of regulatory T (Treg) cells and an increased CD4+ T cell proliferation. Treatment with 1-MT did not affect the phosphorylation of the MAPK pathway in DCs. In general, our study suggests that IDO may play an important role in the pathogenesis of VKH disease by regulating DC and CD4+ T cell function. Tryptophan deficiency and kynurenine accumulation may account for the complicated effects of IDO. Further research is needed to study the precise tryptophan metabolites that may limit immune responses in VKH disease.
Collapse
|
5
|
The Uniqueness of Tryptophan in Biology: Properties, Metabolism, Interactions and Localization in Proteins. Int J Mol Sci 2020; 21:ijms21228776. [PMID: 33233627 PMCID: PMC7699789 DOI: 10.3390/ijms21228776] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Tryptophan (Trp) holds a unique place in biology for a multitude of reasons. It is the largest of all twenty amino acids in the translational toolbox. Its side chain is indole, which is aromatic with a binuclear ring structure, whereas those of Phe, Tyr, and His are single-ring aromatics. In part due to these elaborate structural features, the biosynthetic pathway of Trp is the most complex and the most energy-consuming among all amino acids. Essential in the animal diet, Trp is also the least abundant amino acid in the cell, and one of the rarest in the proteome. In most eukaryotes, Trp is the only amino acid besides Met, which is coded for by a single codon, namely UGG. Due to the large and hydrophobic π-electron surface area, its aromatic side chain interacts with multiple other side chains in the protein, befitting its strategic locations in the protein structure. Finally, several Trp derivatives, namely tryptophylquinone, oxitriptan, serotonin, melatonin, and tryptophol, have specialized functions. Overall, Trp is a scarce and precious amino acid in the cell, such that nature uses it parsimoniously, for multiple but selective functions. Here, the various aspects of the uniqueness of Trp are presented in molecular terms.
Collapse
|
6
|
Kaul NC, Mohapatra SR, Adam I, Tucher C, Tretter T, Opitz CA, Lorenz HM, Tykocinski LO. Hypoxia decreases the T helper cell-suppressive capacity of synovial fibroblasts by downregulating IDO1-mediated tryptophan metabolism. Rheumatology (Oxford) 2020; 59:1148-1158. [PMID: 31846032 DOI: 10.1093/rheumatology/kez587] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/07/2019] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE The development of RA is linked to local infiltration of immune cells and to changes in the phenotype of synovial fibroblasts. Synovial fibroblasts possess the capacity to suppress T cell responses through indoleamine 2, 3-dioxygenase 1 (IDO1)-mediated tryptophan metabolism. However, synovial fibroblasts from RA patients are restricted in this immune-modulatory function. Moreover, hypoxic conditions are detected within synovial tissues of RA patients, with oxygen tensions of only 3.2% O2. This study aims at investigating the effects of hypoxia on the interaction between T cells and synovial fibroblasts, particularly on the T cell-suppressive capacities of synovial fibroblasts. METHODS Synovial fibroblasts were cultured with Th cells under normoxic and hypoxic conditions (3% O2). Th cell proliferation was detected by flow cytometry. Tryptophan and kynurenine amounts were measured by HPLC. IDO1 expression and signal transducer and activator of transcription 1 (STAT1) phosphorylation were quantified by real-time PCR or western blot, and cytokine secretion by ELISA. RESULTS Hypoxic conditions strongly diminished the Th cell-suppressive capacities of both OA synovial fibroblasts and RA synovial fibroblasts. Accordingly, IDO1 mRNA and protein expression, STAT1 phosphorylation and tryptophan metabolism were greatly reduced in OA synovial fibroblasts by hypoxia. MMP-3, IL-6, IL-10 and IFNγ secretion were significantly decreased under hypoxia in synovial fibroblast-Th cell co-cultures, while IL-17A levels were elevated. Supplementation with IFNγ, a well-known inducer of IDO1 expression, could rescue neither IDO1 expression nor Th cell suppression under hypoxic conditions. CONCLUSION Hypoxia strongly affected the crosstalk between synovial fibroblasts and Th cells. By reducing the efficiency of synovial fibroblasts to restrict Th cell proliferation and by increasing the expression of IL-17A, hypoxia might have implications on the pathophysiology of RA.
Collapse
Affiliation(s)
- Nathalie-Christin Kaul
- Division of Rheumatology, Department of Internal Medicine V, Heidelberg University Hospital
| | - Soumya R Mohapatra
- Brain Cancer Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Isabell Adam
- Brain Cancer Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christine Tucher
- Division of Rheumatology, Department of Internal Medicine V, Heidelberg University Hospital
| | - Theresa Tretter
- Division of Rheumatology, Department of Internal Medicine V, Heidelberg University Hospital
| | - Christiane A Opitz
- Brain Cancer Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Division of Rheumatology, Department of Internal Medicine V, Heidelberg University Hospital
| | - Lars-Oliver Tykocinski
- Division of Rheumatology, Department of Internal Medicine V, Heidelberg University Hospital
| |
Collapse
|
7
|
Jiang S, Li H, Piao L, Jin Z, Liu J, Chen S, Liu LL, Shao Y, Zhong S, Wu B, Li W, Ren J, Zhang Y, Wang H, Jin R. Computational study on new natural compound inhibitors of indoleamine 2,3-dioxygenase 1. Aging (Albany NY) 2020; 12:11349-11363. [PMID: 32568737 PMCID: PMC7343476 DOI: 10.18632/aging.103113] [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: 07/31/2019] [Accepted: 04/07/2020] [Indexed: 12/30/2022]
Abstract
Indoleamine 2,3-Dioxygenase (IDO), is a speed limiting enzyme that catalyzes the decomposition and metabolism of Tryptophan along Tryptophan-IDO-Kynurenine pathway [1]. Tryptophan is a necessary amino acid for activating cell growth and metabolism. Additionally, the insufficiency of Tryptophan can lead to immune system dysfunction. Raising the level of Indoleamine 2,3-Dioxygenase protein can promote stagnation and apoptosis of effector T cells [2]. In contrast, the decline in the number of effect T cells naturally protects cancer cells from attack. Therefore, Indoleamine 2,3-Dioxygenase is a potential target for tumour immunotherapy, such as melanoma, ovarian cancer, lung cancer, leukaemia, and so on, especially in solid tumours [3]. In the study, we have done sets of virtual screening aided by computer techniques in order to find potentially effective inhibitors of Indoleamine 2,3-Dioxygenase. Firstly, screening based on structure was carried out by Libdock. Then, ADME (adsorption, distribution, metabolism, excretion) and toxicity prediction were also analyzed. Molecular docking and 3D-QSAR pharmacophore generation were used to study the mechanism of these compounds and Indoleamine 2,3-Dioxygenase’s binding. A molecular dynamic analysis was carried out to assess if these potential compound’s binding is stable enough. According to the results of the analysis above, two potential compounds (ZINC000012495022 and ZINC000003791817) from the ZINC database were discovered to interact with Indoleamine 2,3-Dioxygenase with appropriate energy and proved to be none toxic. The study offered valuable information of Indoleamine 2,3-Dioxygenase inhibitor-based drug discovery in cancer therapy by increasing the activity of T cells and releasing immunity suppression [4, 5].
Collapse
Affiliation(s)
- Shanshan Jiang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China.,Institute of Zoology, Chinese Academy of Sciences, Chaoyang, China
| | - Hui Li
- Clinical College, Jilin University, Changchun, China.,Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Lianhua Piao
- College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Zheng Jin
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Jingyi Liu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Sitong Chen
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Luwei Lucy Liu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Yujie Shao
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Sheng Zhong
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China.,Clinical College, Jilin University, Changchun, China.,Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Bo Wu
- Clinical College, Jilin University, Changchun, China.,Department of Orthopaedic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Weihang Li
- Clinical College, Jilin University, Changchun, China.,Department of Orthopaedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jiaxin Ren
- Clinical College, Jilin University, Changchun, China
| | - Yu Zhang
- Clinical College, Jilin University, Changchun, China
| | - Hao Wang
- Clinical College, Jilin University, Changchun, China
| | - Rihua Jin
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
8
|
Shen F, Feng L, Zhu Y, Tao D, Xu J, Peng R, Liu Z. Oxaliplatin-/NLG919 prodrugs-constructed liposomes for effective chemo-immunotherapy of colorectal cancer. Biomaterials 2020; 255:120190. [PMID: 32563943 DOI: 10.1016/j.biomaterials.2020.120190] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/25/2020] [Accepted: 06/09/2020] [Indexed: 12/12/2022]
Abstract
High expression of indoleamine 2,3-dioxygenase 1 (IDO1) is a major cause of tumor induced immunosuppression, and appears to be associated with poor prognosis in human colorectal cancer and some others. In this study, we construct a bifunctional liposome by self-assembly of oxaliplatin-prodrug (Oxa(IV)) conjugated phospholipid and alkylated NLG919 (aNLG), an IDO1 inhibitor, together with other commercial lipids. The obtained aNLG/Oxa(IV)-Lip can not only release cytotoxic oxaliplatin inside the reductive cytosol to trigger immunogenic cell death (ICD) of cancer cells, but also efficiently retard the degradation of tryptophan to immunosuppressive kynurenine via the NLG919 mediated inhibition of IDO1. Moreover, in vivo pharmacokinetic studies indicate that such aNLG/Oxa(IV)-Lip has a long blood circulation time, thereby enables highly-efficient passive tumor homing. Upon tumor accumulation, such aNLG/Oxa(IV)-Lip presents superior synergistic antitumor efficacies to both subcutaneous and orthotopic CT26 tumors, ascribing to significantly primed anti-tumor immunity of enhanced intratumoral infiltration of CD8+ T cells, scretion of cytotoxic cytokines and downregulation of immunosuppressive regulatory T cells. This work highlights that such bifunctional aNLG/Oxa(IV)-Lip is a potent candidate for future clinical translation owing to its excellent biocompatibility and high therapeutic efficacy.
Collapse
Affiliation(s)
- Fengyun Shen
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Liangzhu Feng
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Yujie Zhu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Danlei Tao
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jun Xu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Rui Peng
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Zhuang Liu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
9
|
Desbois AC, Cacoub P, Leroyer AS, Tellier E, Garrido M, Maciejewski-Duval A, Comarmond C, Barete S, Arock M, Bruneval P, Launay JM, Fouret P, Blank U, Rosenzwajg M, Klatzmann D, Jarraya M, Cluzel P, Koskas F, Kaplanski G, Saadoun D. Immunomodulatory role of Interleukin-33 in large vessel vasculitis. Sci Rep 2020; 10:6405. [PMID: 32286393 PMCID: PMC7156501 DOI: 10.1038/s41598-020-63042-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 02/25/2020] [Indexed: 01/19/2023] Open
Abstract
The mechanisms regulating inflammation in large vessels vasculitis (LVV) are poorly understood. Interleukin 33 (IL-33) has been shown to license innate and adaptive immunity by enhancing Th2 cytokines production. We aimed to examine the role of IL-33 in the immunomodulation of T cell activation in LVV. T cell homeostasis and cytokines production were determined in peripheral blood from 52 patients with giant cell arteritis (GCA) and 50 healthy donors (HD), using Luminex assay, flow cytometry, quantitative RT-PCR and by immunofluorescence analysis in inflammatory aorta lesions. We found increased level of IL-33 and its receptor ST2/IL-1R4 in the serum of patient with LVV. Endothelial cells were the main source of IL-33, whereas Th2 cells, Tregs and mast cells (MC) express ST2 in LVV vessels. IL-33 had a direct immunomodulatory impact by increasing Th2 and Tregs. IL-33 and MC further enhanced Th2 and regulatory responses by inducing a 6.1 fold increased proportion of Tregs (p = 0.008). Stimulation of MC by IL-33 increased indoleamine 2 3-dioxygenase (IDO) activity and IL-2 secretion. IL-33 mRNA expression was significantly correlated with the expression of IL-10 and TGF-β within aorta inflammatory lesions. To conclude, our findings suggest that IL-33 may exert a critical immunoregulatory role in promoting Tregs and Th2 cells in LVV.
Collapse
Affiliation(s)
- Anne-Claire Desbois
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, UMR S 959, Immunology-Immunopathology- Immunotherapy (I3), F-75005, Paris, France
- Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Hôpital Pitié-Salpêtrière, AP-HP, F-75651, Paris, France
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, F-75013, Paris, France
- Centre national de références Maladies Autoimmunes et systémiques rares et Maladies Autoinflammatoires rares, Paris, France
| | - Patrice Cacoub
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, UMR S 959, Immunology-Immunopathology- Immunotherapy (I3), F-75005, Paris, France
- Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Hôpital Pitié-Salpêtrière, AP-HP, F-75651, Paris, France
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, F-75013, Paris, France
- Centre national de références Maladies Autoimmunes et systémiques rares et Maladies Autoinflammatoires rares, Paris, France
| | - Aurélie S Leroyer
- Aix-Marseille Univ; INSERM, Vascular Research Center of Marseille, UMR-S 1076, 13385, Marseille, France
| | - Edwige Tellier
- Aix-Marseille Univ; INSERM, Vascular Research Center of Marseille, UMR-S 1076, 13385, Marseille, France
| | - Marlène Garrido
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, UMR S 959, Immunology-Immunopathology- Immunotherapy (I3), F-75005, Paris, France
- Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Hôpital Pitié-Salpêtrière, AP-HP, F-75651, Paris, France
| | - Anna Maciejewski-Duval
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, UMR S 959, Immunology-Immunopathology- Immunotherapy (I3), F-75005, Paris, France
- Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Hôpital Pitié-Salpêtrière, AP-HP, F-75651, Paris, France
| | - Cloé Comarmond
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, UMR S 959, Immunology-Immunopathology- Immunotherapy (I3), F-75005, Paris, France
- Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Hôpital Pitié-Salpêtrière, AP-HP, F-75651, Paris, France
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, F-75013, Paris, France
- Centre national de références Maladies Autoimmunes et systémiques rares et Maladies Autoinflammatoires rares, Paris, France
| | - Stéphane Barete
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, F-75013, Paris, France
- Centre national de références Maladies Autoimmunes et systémiques rares et Maladies Autoinflammatoires rares, Paris, France
| | - Michel Arock
- Laboratoire de biotechnologies et pharmacologie génétique appliquée, CNRS UMR 8147, ENS - Ecole normale supérieure de Cachan, Cachan, France
- AP-HP, Hôpital Pitié-Salpétrière, Laboratoire d'Hématologie Biologique, Paris, France
| | - Patrick Bruneval
- Laboratoire d'anatomopathologie, Hôpital Européen Georges Pompidou, Paris, France
| | | | - Pierre Fouret
- Laboratoire d'anatomopathologie; Groupe Hospitalier Pitié-Salpétrière, Paris, France
| | - Ulrich Blank
- Inserm U1149, CNRS ERL8252, Faculté de Médecine Site X. Bichat, Paris, France
| | - Michelle Rosenzwajg
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, UMR S 959, Immunology-Immunopathology- Immunotherapy (I3), F-75005, Paris, France
- Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Hôpital Pitié-Salpêtrière, AP-HP, F-75651, Paris, France
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, F-75013, Paris, France
- Centre national de références Maladies Autoimmunes et systémiques rares et Maladies Autoinflammatoires rares, Paris, France
| | - David Klatzmann
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, UMR S 959, Immunology-Immunopathology- Immunotherapy (I3), F-75005, Paris, France
- Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Hôpital Pitié-Salpêtrière, AP-HP, F-75651, Paris, France
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, F-75013, Paris, France
- Centre national de références Maladies Autoimmunes et systémiques rares et Maladies Autoinflammatoires rares, Paris, France
| | - Mohamed Jarraya
- Banque des tissus Humains, Hôpital saint Louis, Paris, France
| | - Philippe Cluzel
- Service de radiologie vasculaire, Groupe Hospitalier Pitié-Salpétrière, Paris, France
| | - Fabien Koskas
- Service de Chirurgie vasculaire, Groupe Hospitalier Pitié-Salpétrière, Paris, France
| | - Gilles Kaplanski
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, F-75013, Paris, France
- APHM, CHU Conception, Service de Médecine Interne, Marseille, France
| | - David Saadoun
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, UMR S 959, Immunology-Immunopathology- Immunotherapy (I3), F-75005, Paris, France.
- Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Hôpital Pitié-Salpêtrière, AP-HP, F-75651, Paris, France.
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, F-75013, Paris, France.
- Centre national de références Maladies Autoimmunes et systémiques rares et Maladies Autoinflammatoires rares, Paris, France.
| |
Collapse
|
10
|
Grunewald ME, Shaban MG, Mackin SR, Fehr AR, Perlman S. Murine Coronavirus Infection Activates the Aryl Hydrocarbon Receptor in an Indoleamine 2,3-Dioxygenase-Independent Manner, Contributing to Cytokine Modulation and Proviral TCDD-Inducible-PARP Expression. J Virol 2020; 94:e01743-19. [PMID: 31694960 PMCID: PMC7000979 DOI: 10.1128/jvi.01743-19] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 11/04/2019] [Indexed: 11/20/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a cytoplasmic receptor/transcription factor that modulates several cellular and immunological processes following activation by pathogen-associated stimuli, though its role during virus infection is largely unknown. Here, we show that AhR is activated in cells infected with mouse hepatitis virus (MHV), a coronavirus (CoV), and contributes to the upregulation of downstream effector TCDD-inducible poly(ADP-ribose) polymerase (TiPARP) during infection. Knockdown of TiPARP reduced viral replication and increased interferon expression, suggesting that TiPARP functions in a proviral manner during MHV infection. We also show that MHV replication induced the expression of other genes known to be downstream of AhR in macrophages and dendritic cells and in livers of infected mice. Further, we found that chemically inhibiting or activating AhR reciprocally modulated the expression levels of cytokines induced by infection, specifically, interleukin 1β (IL-1β), IL-10, and tumor necrosis factor alpha (TNF-α), consistent with a role for AhR activation in the host response to MHV infection. Furthermore, while indoleamine 2,3-dioxygenase (IDO1) drives AhR activation in other settings, MHV infection induced equal expression of downstream genes in wild-type (WT) and IDO1-/- macrophages, suggesting an alternative pathway of AhR activation. In summary, we show that coronaviruses elicit AhR activation by an IDO1-independent pathway, contributing to upregulation of downstream effectors, including the proviral factor TiPARP, and to modulation of cytokine gene expression, and we identify a previously unappreciated role for AhR signaling in CoV pathogenesis.IMPORTANCE Coronaviruses are a family of positive-sense RNA viruses with human and agricultural significance. Characterizing the mechanisms by which coronavirus infection dictates pathogenesis or counters the host immune response would provide targets for the development of therapeutics. Here, we show that the aryl hydrocarbon receptor (AhR) is activated in cells infected with a prototypic coronavirus, mouse hepatitis virus (MHV), resulting in the expression of several effector genes. AhR is important for modulation of the host immune response to MHV and plays a role in the expression of TiPARP, which we show is required for maximal viral replication. Taken together, our findings highlight a previously unidentified role for AhR in regulating coronavirus replication and the immune response to the virus.
Collapse
Affiliation(s)
- Matthew E Grunewald
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Mohamed G Shaban
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Samantha R Mackin
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Anthony R Fehr
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
11
|
Solito S, Pinton L, De Sanctis F, Ugel S, Bronte V, Mandruzzato S, Marigo I. Methods to Measure MDSC Immune Suppressive Activity In Vitro and In Vivo. ACTA ACUST UNITED AC 2018; 124:e61. [PMID: 30303619 DOI: 10.1002/cpim.61] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This unit presents methods to assess the immunosuppressive properties of immunoregulatory cells of myeloid origin, such as myeloid-derived suppressor cells (MDSCs), both in vitro and in vivo in mice, as well as in biological samples from cancer patients. These methods could be adapted to test the impact of different suppressive populations on T cell activation, proliferation, and cytotoxic activity; moreover, they could be useful to assess the influence exerted by genetic modifications, chemical inhibitors, and drugs on immune suppressive pathways © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Samantha Solito
- Oncology and Immunology Section, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Laura Pinton
- Veneto Institute of Oncology IOV- IRCCS, Padova, Italy
| | - Francesco De Sanctis
- Immunology Section, Department of Medicine, Verona University Hospital, Verona, Italy
| | - Stefano Ugel
- Immunology Section, Department of Medicine, Verona University Hospital, Verona, Italy
| | - Vincenzo Bronte
- Immunology Section, Department of Medicine, Verona University Hospital, Verona, Italy
| | - Susanna Mandruzzato
- Oncology and Immunology Section, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Veneto Institute of Oncology IOV- IRCCS, Padova, Italy
| | - Ilaria Marigo
- Veneto Institute of Oncology IOV- IRCCS, Padova, Italy
| |
Collapse
|
12
|
Orabona C, Mondanelli G, Pallotta MT, Carvalho A, Albini E, Fallarino F, Vacca C, Volpi C, Belladonna ML, Berioli MG, Ceccarini G, Esposito SM, Scattoni R, Verrotti A, Ferretti A, De Giorgi G, Toni S, Cappa M, Matteoli MC, Bianchi R, Matino D, Iacono A, Puccetti M, Cunha C, Bicciato S, Antognelli C, Talesa VN, Chatenoud L, Fuchs D, Pilotte L, Van den Eynde B, Lemos MC, Romani L, Puccetti P, Grohmann U. Deficiency of immunoregulatory indoleamine 2,3-dioxygenase 1in juvenile diabetes. JCI Insight 2018; 3:96244. [PMID: 29563329 DOI: 10.1172/jci.insight.96244] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 02/13/2018] [Indexed: 12/23/2022] Open
Abstract
A defect in indoleamine 2,3-dioxygenase 1 (IDO1), which is responsible for immunoregulatory tryptophan catabolism, impairs development of immune tolerance to autoantigens in NOD mice, a model for human autoimmune type 1 diabetes (T1D). Whether IDO1 function is also defective in T1D is still unknown. We investigated IDO1 function in sera and peripheral blood mononuclear cells (PBMCs) from children with T1D and matched controls. These children were further included in a discovery study to identify SNPs in IDO1 that might modify the risk of T1D. T1D in children was characterized by a remarkable defect in IDO1 function. A common haplotype, associated with dysfunctional IDO1, increased the risk of developing T1D in the discovery and also confirmation studies. In T1D patients sharing such a common IDO1 haplotype, incubation of PBMCs in vitro with tocilizumab (TCZ) - an IL-6 receptor blocker - would, however, rescue IDO1 activity. In an experimental setting with diabetic NOD mice, TCZ was found to restore normoglycemia via IDO1-dependent mechanisms. Thus, functional SNPs of IDO1 are associated with defective tryptophan catabolism in human T1D, and maneuvers aimed at restoring IDO1 function would be therapeutically effective in at least a subgroup of T1D pediatric patients.
Collapse
Affiliation(s)
- Ciriana Orabona
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Giada Mondanelli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Maria T Pallotta
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Elisa Albini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Carmine Vacca
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Claudia Volpi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Maria L Belladonna
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Maria G Berioli
- Department of Surgical and Biomedical Sciences, University of Perugia, Perugia, Italy
| | - Giulia Ceccarini
- Department of Surgical and Biomedical Sciences, University of Perugia, Perugia, Italy.,Pediatric Clinic of S. Maria della Misericordia Hospital, Perugia, Italy
| | - Susanna Mr Esposito
- Department of Surgical and Biomedical Sciences, University of Perugia, Perugia, Italy.,Pediatric Clinic of S. Maria della Misericordia Hospital, Perugia, Italy
| | - Raffaella Scattoni
- Department of Surgical and Biomedical Sciences, University of Perugia, Perugia, Italy.,Pediatric Clinic of S. Maria della Misericordia Hospital, Perugia, Italy
| | - Alberto Verrotti
- Department of Surgical and Biomedical Sciences, University of Perugia, Perugia, Italy.,Pediatric Clinic of S. Maria della Misericordia Hospital, Perugia, Italy
| | | | - Giovanni De Giorgi
- Pediatric Clinic of S. Maria della Misericordia Hospital, Perugia, Italy
| | - Sonia Toni
- Juvenile Diabetes Center, Anna Meyer Children's Hospital, Florence, Italy
| | - Marco Cappa
- Unit of Endocrinology and Diabetes, 'Bambino Gesù' Children's Hospital, Rome, Italy
| | - Maria C Matteoli
- Unit of Endocrinology and Diabetes, 'Bambino Gesù' Children's Hospital, Rome, Italy
| | - Roberta Bianchi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Davide Matino
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Alberta Iacono
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Matteo Puccetti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Cinzia Antognelli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Vincenzo N Talesa
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Lucienne Chatenoud
- INSERM U1013, Hôpital Necker-Enfants Malades and Université Paris Descartes, Paris, France
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Medical University, Innsbruck, Austria
| | - Luc Pilotte
- Ludwig Institute for Cancer Research, Walloon Excellence in Life Sciences and Biotechnology and.,De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Benoît Van den Eynde
- Ludwig Institute for Cancer Research, Walloon Excellence in Life Sciences and Biotechnology and.,De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Manuel C Lemos
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Paolo Puccetti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Ursula Grohmann
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
13
|
Volpi C, Fallarino F, Mondanelli G, Macchiarulo A, Grohmann U. Opportunities and challenges in drug discovery targeting metabotropic glutamate receptor 4. Expert Opin Drug Discov 2018; 13:411-423. [DOI: 10.1080/17460441.2018.1443076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Claudia Volpi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Giada Mondanelli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Ursula Grohmann
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
14
|
Mesenchymal Stem Cells as New Therapeutic Agents for the Treatment of Primary Biliary Cholangitis. Anal Cell Pathol (Amst) 2017; 2017:7492836. [PMID: 29410945 PMCID: PMC5749170 DOI: 10.1155/2017/7492836] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/17/2017] [Accepted: 10/26/2017] [Indexed: 12/20/2022] Open
Abstract
Primary biliary cholangitis (PBC) is a chronic autoimmune cholestatic liver disease characterized by the progressive destruction of small- and medium-sized intrahepatic bile ducts with resultant cholestasis and progressive fibrosis. Ursodeoxycholic acid and obethicholic acid are the only agents approved by the US Food and Drug Administration (FDA) for the treatment of PBC. However, for patients with advanced, end-stage PBC, liver transplantation is still the most effective treatment. Accordingly, the alternative approaches, such as mesenchymal stem cell (MSC) transplantation, have been suggested as an effective alternative therapy for these patients. Due to their immunomodulatory characteristics, MSCs are considered as promising therapeutic agents for the therapy of autoimmune liver diseases, including PBC. In this review, we have summarized the therapeutic potential of MSCs for the treatment of these diseases, emphasizing molecular and cellular mechanisms responsible for MSC-based effects in an animal model of PBC and therapeutic potential observed in recently conducted clinical trials. We have also presented several outstanding problems including safety issues regarding unwanted differentiation of transplanted MSCs which limit their therapeutic use. Efficient and safe MSC-based therapy for PBC remains a challenging issue that requires continuous cooperation between clinicians, researchers, and patients.
Collapse
|
15
|
The immune regulation in cancer by the amino acid metabolizing enzymes ARG and IDO. Curr Opin Pharmacol 2017; 35:30-39. [DOI: 10.1016/j.coph.2017.05.002] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/27/2017] [Accepted: 05/08/2017] [Indexed: 01/04/2023]
|
16
|
de Vries LV, Minović I, Franssen CFM, van Faassen M, Sanders JSF, Berger SP, Navis G, Kema IP, Bakker SJL. The tryptophan/kynurenine pathway, systemic inflammation, and long-term outcome after kidney transplantation. Am J Physiol Renal Physiol 2017; 313:F475-F486. [PMID: 28490533 DOI: 10.1152/ajprenal.00690.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 04/25/2017] [Accepted: 05/02/2017] [Indexed: 12/14/2022] Open
Abstract
Tryptophan is metabolized along the kynurenine pathway, initially to kynurenine, and subsequently to cytotoxic 3-hydroxykynurenine. There is increasing interest in this pathway because of its proinflammatory nature, and drugs interfering in it have received increasing attention. We aimed to investigate whether serum and urinary parameters of the tryptophan/kynurenine pathway, and particularly cytotoxic 3-hydroxykynurenine, are associated with systemic inflammation and long-term outcome in renal transplant recipients (RTR). Data were collected in outpatient RTR with a functioning graft for >1 yr. Tryptophan, kynurenine, and 3-hydroxykynurenine in serum and urine were measured using LC-MS/MS. A total of 561 RTR (age: 51 ± 12 yr; 56% male) were included at a median of 6.0 (2.6-11.6) yr posttransplantation. Baseline median serum tryptophan was 40.0 (34.5-46.0) µmol/l, serum kynurenine was 1.8 (1.4-2.2) µmol/l, and serum 3-hydroxykynurenine was 42.2 (31.0-61.7) nmol/l. Serum kynurenine and 3-hydroxykynurenine were strongly associated with parameters of systemic inflammation. During follow-up for 7.0 (6.2-7.5) yr, 51 RTR (9%) developed graft failure and 120 RTR (21%) died. Both serum kynurenine and 3-hydroxykynurenine were independently associated with graft failure [HR 1.72 (1.23-2.41), P = 0.002; and HR 2.03 (1.42-2.90), P < 0.001]. Serum 3-hydroxykynurenine was also independently associated with mortality [HR 1.37 (1.08-1.73), P = 0.01], whereas serum kynurenine was not. Urinary tryptophan/kynurenine pathway parameters were not associated with outcome. Of tryptophan metabolites, serum 3-hydroxykynurenine is cross-sectionally most strongly and consistently associated with systemic inflammation and prospectively with adverse long-term outcome after kidney transplantation. Serum 3-hydroxykynurenine may be an interesting biomarker and target for the evaluation of drugs interfering in the tryptophan/kynurenine pathway.
Collapse
Affiliation(s)
- Laura V de Vries
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands;
| | - Isidor Minović
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and.,Top Institute Food and Nutrition, Wageningen, The Netherlands
| | - Casper F M Franssen
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martijn van Faassen
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and
| | - Jan-Stephan F Sanders
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stefan P Berger
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerjan Navis
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and
| | - Stephan J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Top Institute Food and Nutrition, Wageningen, The Netherlands
| |
Collapse
|
17
|
Mondanelli G, Bianchi R, Pallotta MT, Orabona C, Albini E, Iacono A, Belladonna ML, Vacca C, Fallarino F, Macchiarulo A, Ugel S, Bronte V, Gevi F, Zolla L, Verhaar A, Peppelenbosch M, Mazza EMC, Bicciato S, Laouar Y, Santambrogio L, Puccetti P, Volpi C, Grohmann U. A Relay Pathway between Arginine and Tryptophan Metabolism Confers Immunosuppressive Properties on Dendritic Cells. Immunity 2017; 46:233-244. [PMID: 28214225 PMCID: PMC5337620 DOI: 10.1016/j.immuni.2017.01.005] [Citation(s) in RCA: 252] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/18/2016] [Accepted: 12/21/2016] [Indexed: 02/07/2023]
Abstract
Arginase 1 (Arg1) and indoleamine 2,3-dioxygenase 1 (IDO1) are immunoregulatory enzymes catalyzing the degradation of l-arginine and l-tryptophan, respectively, resulting in local amino acid deprivation. In addition, unlike Arg1, IDO1 is also endowed with non-enzymatic signaling activity in dendritic cells (DCs). Despite considerable knowledge of their individual biology, no integrated functions of Arg1 and IDO1 have been reported yet. We found that IDO1 phosphorylation and consequent activation of IDO1 signaling in DCs was strictly dependent on prior expression of Arg1 and Arg1-dependent production of polyamines. Polyamines, either produced by DCs or released by bystander Arg1+ myeloid-derived suppressor cells, conditioned DCs toward an IDO1-dependent, immunosuppressive phenotype via activation of the Src kinase, which has IDO1-phosphorylating activity. Thus our data indicate that Arg1 and IDO1 are linked by an entwined pathway in immunometabolism and that their joint modulation could represent an important target for effective immunotherapy in several disease settings. Dendritic cells (DCs) can co-express Arg1 and IDO1 immunosuppressive enzymes Arg1 activity is required for IDO1 induction by TGF-β in DCs Spermidine, a downstream Arg1 product, but not arginine starvation, induces IDO1 in DCs Arg1+ myeloid derived suppressor cells (MDSCs) can render DCs immunosuppressive via IDO1
Collapse
Affiliation(s)
- Giada Mondanelli
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Roberta Bianchi
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | | | - Ciriana Orabona
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Elisa Albini
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Alberta Iacono
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | | | - Carmine Vacca
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Francesca Fallarino
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, 06132 Perugia, Italy
| | - Stefano Ugel
- Department of Medicine, Verona University Hospital, 37134 Verona, Italy
| | - Vincenzo Bronte
- Department of Medicine, Verona University Hospital, 37134 Verona, Italy
| | - Federica Gevi
- Department of Ecological and Biological Sciences, University of Tuscia, 01100 Viterbo, Italy
| | - Lello Zolla
- Department of Ecological and Biological Sciences, University of Tuscia, 01100 Viterbo, Italy
| | - Auke Verhaar
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre Rotterdam, 3015 CE Rotterdam, the Netherlands
| | - Maikel Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre Rotterdam, 3015 CE Rotterdam, the Netherlands
| | | | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Yasmina Laouar
- Department of Microbiology & Immunology, University of Michigan School of Medicine, Ann Arbor, MI 48109-5620, US
| | - Laura Santambrogio
- Department of Pathology, Albert Einstein College of Medicine, New York, NY 10461, US
| | - Paolo Puccetti
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Claudia Volpi
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Ursula Grohmann
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| |
Collapse
|
18
|
Alipour R, Masoumi Karimi M, Hashemi-Beni B, Adib M, Sereshki N, Sadeghi F. Indoleamine 2,3-Dioxygenase Is Dispensable for The Immunomodulatory Function of Stem Cells from Human Exfoliated Deciduous Teeth. CELL JOURNAL 2016; 18:597-608. [PMID: 28042544 PMCID: PMC5086338 DOI: 10.22074/cellj.2016.4726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 04/06/2016] [Indexed: 12/18/2022]
Abstract
Objective In this study, we sought to better understand the immunoregulatory function
of stem cells derived from human exfoliated deciduous teeth (SHED). We studied the role
of the interferon gamma (IFN-γ)-indoleamine 2,3-dioxygenase (IDO)-axis in immunoregulation of SHED compared to bone marrow derived mesenchymal stem cells (BMMSCs)
under the same conditions.
Materials and Methods In this cross-sectional study, recently isolated human T cells
were stimulated either by mitogen or inactivated allogeneic peripheral blood mononuclear cells (PBMCs). These T cells were subsequently co-cultured with, either SHED or
BMMSCs in the presence or absence of 1-methyl-tryptophan (1-MT) or neutralizing anti-
human-IFN-γ antibodies. In all co-cultures we evaluated lymphocyte activation as well as
IDO activity.
Results SHED, similar to conventional BMMSCs, had anti-proliferative effects on stimulated T cells and reduced their cytokine production. This property of SHED and BMMSCs
was changed by IFN-γ neutralization. We detected IDO in the immunosuppressive supernatant of all co-cultures. Removal of IDO decreased the immunosuppression of BMMSCs.
Conclusion SHED, like BMMSCs, produced the IDO enzyme. Although IFN-γ is one of
inducer of IDO production in SHED, these cells were not affected by IFN-γ in the same
manner as BMMSCs. Unlike BMMSCs, the IDO enzyme did not contribute to their immunosuppression and might have other cell-type specific roles.
Collapse
Affiliation(s)
- Razieh Alipour
- Department of Immunology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Batool Hashemi-Beni
- Department of Immunology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Minoo Adib
- Department of Immunology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nasrin Sereshki
- Department of Immunology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farzaneh Sadeghi
- Department of Immunology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
19
|
Oghumu S, Nori U, Bracewell A, Zhang J, Bott C, Nadasdy GM, Brodsky SV, Pelletier R, Satoskar AR, Nadasdy T, Satoskar AA. Differential gene expression pattern in biopsies with renal allograft pyelonephritis and allograft rejection. Clin Transplant 2016; 30:1115-1133. [PMID: 27352120 PMCID: PMC5256948 DOI: 10.1111/ctr.12795] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2016] [Indexed: 02/02/2023]
Abstract
Differentiating acute pyelonephritis (APN) from acute rejection (AR) in renal allograft biopsies can sometimes be difficult because of overlapping clinical and histologic features, lack of positive urine cultures,and variable response to antibiotics. We wanted to study differential gene expression between AR and APN using biopsy tissue. Thirty-three biopsies were analyzed using NanoString multiplex platform and PCR (6 transplant baseline biopsies, 8 AR, 15 APN [8 culture positive, 7 culture negative], and 4 native pyelonephritis [NP]). Additional 22 biopsies were tested by PCR to validate the results. CXCL9, CXCL10, CXCL11, and IDO1 were the top differentially expressed genes, upregulated in AR. Lactoferrin (LTF) and CXCL1 were higher in APN and NP. No statistically significant difference in transcript levels was seen between culture-positive and culture-negative APN biopsies. Comparing the overall mRNA signature using Ingenuity pathway analysis, interferon-gamma emerged as the dominant upstream regulator in AR and allograft APN, but not in NP (which clustered separately). Our study suggests that chemokine pathways in graft APN may differ from NP and in fact resemble AR, due to a component of alloreactivity, resulting in variable response to antibiotic treatment. Therefore, cautious addition of steroids might help in resistant cases of graft APN.
Collapse
Affiliation(s)
- Steve Oghumu
- Department of Pathology, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Uday Nori
- Nephrology, Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Anna Bracewell
- Nephrology, Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jianying Zhang
- Department of Biostatistics, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Cherri Bott
- Department of Pathology, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Gyongyi M Nadasdy
- Department of Pathology, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sergey V Brodsky
- Department of Pathology, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ronald Pelletier
- Department of Surgery, Comprehensive Transplant Center, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Abhay R Satoskar
- Department of Pathology, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Tibor Nadasdy
- Department of Pathology, Ohio State University Wexner Medical Center, Columbus, OH, USA.
| | - Anjali A Satoskar
- Department of Pathology, Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
20
|
Jalili RB, Zhang Y, Hosseini-Tabatabaei A, Kilani RT, Khosravi Maharlooei M, Li Y, Salimi Elizei S, Warnock GL, Ghahary A. Fibroblast Cell-Based Therapy for Experimental Autoimmune Diabetes. PLoS One 2016; 11:e0146970. [PMID: 26765526 PMCID: PMC4713151 DOI: 10.1371/journal.pone.0146970] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 12/23/2015] [Indexed: 11/22/2022] Open
Abstract
Type 1 diabetes (T1D) results from autoimmune destruction of insulin producing β cells of the pancreatic islets. Curbing autoimmunity at the initiation of T1D can result in recovery of residual β cells and consequently remission of diabetes. Here we report a cell-based therapy for autoimmune diabetes in non-obese diabetic (NOD) mice using dermal fibroblasts. This was achieved by a single injection of fibroblasts, expressing the immunoregulatory molecule indoleamine 2,3 dioxygenase (IDO), into peritoneal cavity of NOD mice shortly after the onset of overt hyperglycemia. Mice were then monitored for reversal of hyperglycemia and changes in inflammatory / regulatory T cell profiles. Blood glucose levels dropped into the normal range in 82% of NOD mice after receiving IDO-expressing fibroblasts while all control mice remained diabetic. We found significantly reduced islet inflammation, increased regulatory T cells, and decreased T helper 17 cells and β cell specific autoreactive CD8+ T cells following IDO cell therapy. We further showed that some of intraperitoneal injected fibroblasts migrated to local lymph nodes and expressed co-inhibitory molecules. These findings suggest that IDO fibroblasts therapy can reinstate self-tolerance and alleviate β cell autoreactivity in NOD mice, resulting in remission of autoimmune diabetes.
Collapse
MESH Headings
- Animals
- Autoimmunity/genetics
- Autoimmunity/immunology
- Cell Movement/genetics
- Cell Movement/immunology
- Cell- and Tissue-Based Therapy/methods
- Diabetes Mellitus, Experimental
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/therapy
- Fibroblasts/metabolism
- Gene Expression
- Hyperglycemia/genetics
- Hyperglycemia/metabolism
- Hyperglycemia/therapy
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Insulin-Secreting Cells/metabolism
- Islets of Langerhans/immunology
- Islets of Langerhans/metabolism
- Islets of Langerhans/pathology
- Lymph Nodes/immunology
- Lymph Nodes/metabolism
- Lymphocyte Count
- Mice
- Mice, Inbred NOD
- Receptors, CCR7/metabolism
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Reza B. Jalili
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Yun Zhang
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | | | - Ruhangiz T. Kilani
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | | | - Yunyuan Li
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Sanam Salimi Elizei
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Garth L. Warnock
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Aziz Ghahary
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- * E-mail:
| |
Collapse
|
21
|
Salaroglio IC, Campia I, Kopecka J, Gazzano E, Orecchia S, Ghigo D, Riganti C. Zoledronic acid overcomes chemoresistance and immunosuppression of malignant mesothelioma. Oncotarget 2015; 6:1128-42. [PMID: 25544757 PMCID: PMC4359222 DOI: 10.18632/oncotarget.2731] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 11/11/2014] [Indexed: 11/25/2022] Open
Abstract
The human malignant mesothelioma (HMM) is characterized by a chemoresistant and immunosuppressive phenotype. An effective strategy to restore chemosensitivity and immune reactivity against HMM is lacking. We investigated whether the use of zoledronic acid is an effective chemo-immunosensitizing strategy. We compared primary HMM samples with non-transformed mesothelial cells. HMM cells had higher rate of cholesterol and isoprenoid synthesis, constitutive activation of Ras/extracellular signal-regulated kinase1/2 (ERK1/2)/hypoxia inducible factor-1α (HIF-1α) pathway and up-regulation of the drug efflux transporter P-glycoprotein (Pgp). By decreasing the isoprenoid supply, zoledronic acid down-regulated the Ras/ERK1/2/HIF-1α/Pgp axis and chemosensitized the HMM cells to Pgp substrates. The HMM cells also produced higher amounts of kynurenine, decreased the proliferation of T-lymphocytes and expanded the number of T-regulatory (Treg) cells. Kynurenine synthesis was due to the transcription of the indoleamine 1,2 dioxygenase (IDO) enzyme, consequent to the activation of the signal transducer and activator of transcription-3 (STAT3). By reducing the activity of the Ras/ERK1/2/STAT3/IDO axis, zoledronic acid lowered the kyurenine synthesis and the expansion of Treg cells, and increased the proliferation of T-lymphocytes. Thanks to its ability to decrease Ras/ERK1/2 activity, which is responsible for both Pgp-mediated chemoresistance and IDO-mediated immunosuppression, zoledronic acid is an effective chemo-immunosensitizing agent in HMM cells.
Collapse
Affiliation(s)
| | - Ivana Campia
- Department of Oncology, University of Torino, Italy
| | | | | | - Sara Orecchia
- S.C. Anatomia Patologica, Azienda Ospedaliera S.S. Antonio e Biagio, Alessandria, Italy
| | - Dario Ghigo
- Department of Oncology, University of Torino, Italy
| | | |
Collapse
|
22
|
Ajamian F, Wu Y, Ebeling C, Ilarraza R, Odemuyiwa SO, Moqbel R, Adamko DJ. Respiratory syncytial virus induces indoleamine 2,3-dioxygenase activity: a potential novel role in the development of allergic disease. Clin Exp Allergy 2015; 45:644-59. [PMID: 25627660 DOI: 10.1111/cea.12498] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 12/03/2014] [Accepted: 12/21/2014] [Indexed: 01/10/2023]
Abstract
BACKGROUND Infants that develop severe bronchiolitis due to respiratory syncytial virus (RSV) are at increased risk of developing asthma later in life. We investigated a potential immunological mechanism for the association between RSV and the development of allergic inflammation. The enzyme indoleamine 2,3-dioxygenase (IDO) has been reported to induce selective apoptosis of T helper 1 (Th1) cells and contributed to Th2-biased immune responses. OBJECTIVE To determine whether RSV infection in vitro could induce IDO expression and bioactivity in human dendritic cells, leading to a Th2-biased immune response. METHODS Human peripheral blood monocytes from healthy adult donors were isolated, differentiated to dendritic cells (moDC), in vitro. We studied RSV infection and mechanisms of IDO activation in moDC with subsequent effect on T-bet expression. RESULTS We found that moDC were infected by RSV and that this induced IDO activation. RSV-induced IDO activity was inhibited by palivizumab, UV inactivation, TL4R inhibition, and ribavirin. However, blocking endosomal TLR function with chloroquine did not inhibit IDO activity. Selective inhibitors suggested that RSV-induced IDO activity was dependent on the retinoic acid-inducible gene-I (RIG-I) related pathway via NF-κB and p38 MAPK. Coculture of RSV-infected moDC with activated T cells, in a transwell system, suppressed expression of T-bet (a Th1-associated factor) but not GATA3 (a Th2 regulator). Inhibition of IDO activity with the competitive inhibitor, 1-methyl tryptophan, blocked the effect on T-bet expression. CONCLUSION AND CLINICAL RELEVANCE Our data show for the first time that RSV can induce the expression and bioactivity of IDO in human moDC, in a virus replication-dependant fashion. We suggest that RSV activation of IDO could be a potential mechanism for the development of allergic diseases.
Collapse
Affiliation(s)
- F Ajamian
- Pulmonary Research Group, University of Alberta, Edmonton, Alberta, Canada; Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | |
Collapse
|
23
|
De Sanctis F, Solito S, Ugel S, Molon B, Bronte V, Marigo I. MDSCs in cancer: Conceiving new prognostic and therapeutic targets. Biochim Biophys Acta Rev Cancer 2015; 1865:35-48. [PMID: 26255541 DOI: 10.1016/j.bbcan.2015.08.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 07/31/2015] [Accepted: 08/01/2015] [Indexed: 12/30/2022]
Abstract
The incomplete clinical efficacy of anti-tumor immunotherapy can depend on the presence of an immunosuppressive environment in the host that supports tumor progression. Tumor-derived cytokines and growth factors induce an altered hematopoiesis that modifies the myeloid cell differentiation process, promoting proliferation and expansion of cells with immunosuppressive skills, namely myeloid derived suppressor cells (MDSCs). MDSCs promote tumor growth not only by shaping immune responses towards tumor tolerance, but also by supporting several processes necessary for the neoplastic progression such as tumor angiogenesis, cancer stemness, and metastasis dissemination. Thus, MDSC targeting represents a promising tool to eliminate host immune dysfunctions and increase the efficacy of immune-based cancer therapies.
Collapse
Affiliation(s)
- Francesco De Sanctis
- Immunology Section, Department of Pathology and Diagnostics, University of Verona, 37134 Verona, Italy
| | - Samantha Solito
- Section of Oncology and Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | - Stefano Ugel
- Immunology Section, Department of Pathology and Diagnostics, University of Verona, 37134 Verona, Italy
| | - Barbara Molon
- Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Vincenzo Bronte
- Immunology Section, Department of Pathology and Diagnostics, University of Verona, 37134 Verona, Italy.
| | - Ilaria Marigo
- Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| |
Collapse
|
24
|
Aiyaz M, Bipin C, Pantulwar V, Mugasimangalam R, Shanley CA, Ordway DJ, Orme IM. Whole genome response in guinea pigs infected with the high virulence strain Mycobacterium tuberculosis TT372. Tuberculosis (Edinb) 2015; 94:606-15. [PMID: 25621360 DOI: 10.1016/j.tube.2014.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this study we conducted a microarray-based whole genomic analysis of gene expression in the lungs after exposure of guinea pigs to a low dose aerosol of the Atypical Beijing Western Cape TT372 strain of Mycobacterium tuberculosis, after harvesting lung tissues three weeks after infection at a time that effector immunity is starting to peak. The infection resulted in a very large up-regulation of multiple genes at this time, particularly in the context of a "chemokine storm" in the lungs. Overall gene expression was considerably reduced in animals that had been vaccinated with BCG two months earlier, but in both cases strong signatures featuring gamma interferon [IFNγ] and tumor necrosis factor [TNFα] were observed indicating the potent TH1 response in these animals. Even though their effects are not seen until later in the infection, even at this early time point gene expression patterns associated with the potential emergence of regulatory T cells were observed. Genes involving lung repair, response to oxidative stress, and cell trafficking were strongly expressed, but interesting these gene patterns differed substantially between the infected and vaccinated/infected groups of animals. Given the importance of this species as a relevant and cost-effective small animal model of tuberculosis, this approach has the potential to provide new information regarding the effects of vaccination on control of the disease process.
Collapse
|
25
|
An Autocrine Cytokine/JAK/STAT-Signaling Induces Kynurenine Synthesis in Multidrug Resistant Human Cancer Cells. PLoS One 2015; 10:e0126159. [PMID: 25955018 PMCID: PMC4425697 DOI: 10.1371/journal.pone.0126159] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 03/29/2015] [Indexed: 12/01/2022] Open
Abstract
Background Multidrug resistant cancer cells are hard to eradicate for the inefficacy of conventional anticancer drugs. Besides escaping the cytotoxic effects of chemotherapy, they also bypass the pro-immunogenic effects induced by anticancer drugs: indeed they are not well recognized by host dendritic cells and do not elicit a durable anti-tumor immunity. It has not yet been investigated whether multidrug resistant cells have a different ability to induce immunosuppression than chemosensitive ones. We addressed this issue in human and murine chemosensitive and multidrug resistant cancer cells. Results We found that the activity and expression of indoleamine 2,3-dioxygenase 1 (IDO1), which catalyzes the conversion of tryptophan into the immunosuppressive metabolite kynurenine, was higher in all the multidrug resistant cells analyzed and that IDO1 inhibition reduced the growth of drug-resistant tumors in immunocompetent animals. In chemoresistant cells the basal activity of JAK1/STAT1 and JAK1/STAT3 signaling was higher, the STAT3 inhibitor PIAS3 was down-regulated, and the autocrine production of STAT3-target and IDO1-inducers cytokines IL-6, IL-4, IL-1β, IL-13, TNF-α and CD40L, was increased. The disruption of the JAK/STAT signaling lowered the IDO1 activity and reversed the kynurenine-induced pro-immunosuppressive effects, as revealed by the restored proliferation of T-lymphocytes in STAT-silenced chemoresistant cells. Conclusions Our work shows that multidrug resistant cells have a stronger immunosuppressive attitude than chemosensitive cells, due to the constitutive activation of the JAK/STAT/IDO1 axis, thus resulting chemo- and immune-evasive. Disrupting this axis may significantly improve the efficacy of chemo-immunotherapy protocols against resistant tumors.
Collapse
|
26
|
Williams AC, Dunbar RIM. Big brains, meat, tuberculosis and the nicotinamide switches: co-evolutionary relationships with modern repercussions on longevity and disease? Med Hypotheses 2014; 83:79-87. [PMID: 24767939 DOI: 10.1016/j.mehy.2014.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 04/01/2014] [Indexed: 11/27/2022]
Abstract
Meat eating has been an important trigger for human evolution however the responsible component in meat has not been clearly identified. Here we propose that the limiting factors for expanding brains and increasing longevity were the micronutrient nicotinamide (vitamin B3) and the metabolically related essential amino-acid, tryptophan. Meat offers significant sourcing challenges and lack causes a deficiency of nicotinamide and tryptophan and consequently the energy carrier nicotinamide adenine dinucleotide (NAD) that gets consumed in regulatory circuits important for survival, resulting in premature ageing, poor cognition and brain atrophy. If a trophic supply of dietary nicotinamide/tryptophan is so essential for building brains, constraining their size and connectivity, we hypothesise that back-up mechanisms to ensure the supply evolved. One strategy may be increasing the reliance on gut symbionts to break down celluloses that produces NADH and only nicotinamide indirectly, and may cause diarrhoea. We suggest that a direct supplier was the chronic mycobacterial infection tuberculosis (TB) that is a surprise candidate but it co-evolved early, does not inevitably cause disease (90-95% of those infected are healthy), and secretes (and is inhibited by) nicotinamide. We hypothesise that TB evolved first as a symbiont that enabled humans to cope with short-lived shortages of meat and only later behaved as a pathogen when the supply deteriorated chronically, for those in poverty. (TB immunology and epidemiology is riddled with paradoxes for a conventional pathogen). We test this in pilot data showing that sharp declines in TB (and diarrhoea) - `environmental enteropathy' strongly correlate with increasing meat consumption and therefore nicotinamide exposure, unlike later onset cancers and Parkinson's disease that increased in incidence, perhaps - as we propose a hypothetical hypervitaminosis B3 (to include obesity and the metabolic syndrome) - as the trade-off for increased brain power and longevity, a recently evolved human characteristic.
Collapse
Affiliation(s)
- Adrian C Williams
- Institute for Cognitive and Evolutionary Anthropology, University of Oxford, 64 Banbury Road, Oxford OX2 6PN, UK.
| | - Robin I M Dunbar
- Department of Experimental Psychology, University of Oxford, South Parks Rd, Oxford OX1 3UD, UK
| |
Collapse
|
27
|
Mattox ML, D'Angelo JA, Dickinson BL. Redox control of indoleamine 2,3-dioxygenase expression and activity in human monocyte-derived dendritic cells is independent of changes in oxygen tension. Scand J Immunol 2014; 79:325-32. [PMID: 24612287 DOI: 10.1111/sji.12164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 02/26/2014] [Indexed: 11/28/2022]
Abstract
Dendritic cells (DCs) initiate adaptive immune responses to pathogens and tumours and maintain tolerance to self and innocuous antigens. These functions occur in organs and tissues exhibiting wide variations in nutrients, growth factors, redox and oxygen tension. Understanding how these microenvironmental factors influence DCs to affect immunological outcomes is of increasing relevance with the emerging success of DC-based cellular vaccines. In a previous study, we examined whether redox, an important environmental cue, could influence DC expression of the immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO). IDO-competent DCs promote long-term immune homoeostasis by limiting exaggerated inflammatory responses and directing regulatory T-cell effector function. To alter redox, we manipulated the activity of the cystine/glutamate antiporter, which functions to maintain intracellular and extracellular redox. The results of that study showed that redox perturbation strongly induced IDO expression and activity in DCs. While this study was performed using standard cell culture techniques with DCs cultured under 5% CO₂ and 20% O₂, it is clear that DCs capture and present antigens in inflamed tissues and secondary lymphoid organs which exhibit low oxygen tension (1-5% O₂). Therefore, here we investigated whether oxygen tension influences DC expression of IDO in the context of homoeostatic and altered redox.
Collapse
Affiliation(s)
- M L Mattox
- The West Virginia School of Osteopathic Medicine, Lewisburg, WV, USA
| | | | | |
Collapse
|
28
|
Rytelewski M, Meilleur CE, Atef Yekta M, Szabo PA, Garg N, Schell TD, Jevnikar AM, Sharif S, Singh B, Haeryfar SMM. Suppression of immunodominant antitumor and antiviral CD8+ T cell responses by indoleamine 2,3-dioxygenase. PLoS One 2014; 9:e90439. [PMID: 24587363 PMCID: PMC3938761 DOI: 10.1371/journal.pone.0090439] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 01/30/2014] [Indexed: 11/19/2022] Open
Abstract
Indoleamine 2,3-dioxygenase (IDO) is a tryptophan-degrading enzyme known to suppress antitumor CD8+ T cells (TCD8). The role of IDO in regulation of antiviral TCD8 responses is far less clear. In addition, whether IDO controls both immunodominant and subdominant TCD8 is not fully understood. This is an important question because the dominance status of tumor- and virus-specific TCD8 may determine their significance in protective immunity and in vaccine design. We evaluated the magnitude and breadth of cross-primed TCD8 responses to simian virus 40 (SV40) large T antigen as well as primary and recall TCD8 responses to influenza A virus (IAV) in the absence or presence of IDO. IDO−/− mice and wild-type mice treated with 1-methyl-D-tryptophan, a pharmacological inhibitor of IDO, exhibited augmented responses to immunodominant epitopes encoded by T antigen and IAV. IDO-mediated suppression of these responses was independent of CD4+CD25+FoxP3+ regulatory T cells, which remained numerically and functionally intact in IDO−/− mice. Treatment with L-kynurenine failed to inhibit TCD8 responses, indicating that tryptophan metabolites are not responsible for the suppressive effect of IDO in our models. Immunodominant T antigen-specific TCD8 from IDO−/− mice showed increased Ki-67 expression, suggesting that they may have acquired a more vigorous proliferative capacity in vivo. In conclusion, IDO suppresses immunodominant TCD8 responses to tumor and viral antigens. Our work also demonstrates that systemic primary and recall TCD8 responses to IAV are controlled by IDO. Inhibition of IDO thus represents an attractive adjuvant strategy in boosting anticancer and antiviral TCD8 targeting highly immunogenic antigens.
Collapse
MESH Headings
- Animals
- Antigens, Polyomavirus Transforming/immunology
- Antigens, Viral/immunology
- CD4 Antigens/genetics
- CD4 Antigens/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/immunology
- Gene Expression
- Immune Tolerance/genetics
- Immunity, Innate
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Indoleamine-Pyrrole 2,3,-Dioxygenase/deficiency
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology
- Influenza A virus/immunology
- Interleukin-2 Receptor alpha Subunit/genetics
- Interleukin-2 Receptor alpha Subunit/immunology
- Kynurenine/pharmacology
- Lymphocyte Activation
- Mice
- Mice, Knockout
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tryptophan/analogs & derivatives
- Tryptophan/pharmacology
Collapse
Affiliation(s)
- Mateusz Rytelewski
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Courtney E. Meilleur
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Maryam Atef Yekta
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Peter A. Szabo
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Nitan Garg
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Todd D. Schell
- Department of Microbiology and Immunology, The Pennsylvania State University, Hershey, Pennsylvania, United States of America
| | - Anthony M. Jevnikar
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Department of Medicine, Western University, London, Ontario, Canada
- Department of Pathology, Western University, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
| | - Shayan Sharif
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Bhagirath Singh
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
| | - S. M. Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
29
|
Mittal D, Kassianos AJ, Tran LS, Bergot AS, Gosmann C, Hofmann J, Blumenthal A, Leggatt GR, Frazer IH. Indoleamine 2,3-dioxygenase activity contributes to local immune suppression in the skin expressing human papillomavirus oncoprotein e7. J Invest Dermatol 2013; 133:2686-2694. [PMID: 23652797 PMCID: PMC3779505 DOI: 10.1038/jid.2013.222] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 03/25/2013] [Accepted: 04/16/2013] [Indexed: 12/20/2022]
Abstract
Chronic infection of anogenital epithelium with human papillomavirus (HPV) promotes development of cancer. Many pathogens evoke immunosuppressive mechanisms to enable persistent infection. We have previously shown that grafted skin expressing HPV16 E7 oncoprotein from a keratin-14 promoter (K14E7) is not rejected by a syngeneic, immunocompetent host. In this study we show that indoleamine 2,3-dioxygenase (IDO) 1, an IFN-γ-inducible immunoregulatory molecule, is more highly expressed by langerin(-ve) dermal dendritic cells (DCs) from K14E7 skin than nontransgenic control skin. Furthermore, inhibiting IDO activity using 1-methyl-dl-tryptophan (1-D/L-MT) promotes K14E7 skin graft rejection. Increased IDO1 expression and activity in K14E7 skin requires IFN-γ and invariant natural killer T (iNKT) cells, both of which have been shown to negatively regulate T-cell effector function and suppress K14E7 graft rejection. Furthermore, DCs from K14E7 skin express higher levels of IFN-γ receptor (IFN-γR) than DCs from control skin. K14E7 transgenic skin recruits significantly higher numbers of DCs, independent of IFN-γ and IFN-γR expression. Consistent with these observations in a murine model, we found higher expression of IDO1 and IFN-γ but not IDO2 in the cervical epithelium of patients with HPV-associated cervical intraepithelial neoplasia (CIN) 2/3. Our data support a hypothesis that induction of IDO1 in HPV-infected skin contributes to evasion of host immunity.
Collapse
Affiliation(s)
- Deepak Mittal
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Andrew J Kassianos
- Kidney Research Laboratory, Queensland Health/Queensland Institute of Medical Research, Brisbane,Queensland, Australia
| | - Lee S Tran
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Anne-Sophie Bergot
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Christine Gosmann
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Janin Hofmann
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Antje Blumenthal
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia; Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Queensland, Australia
| | - Graham R Leggatt
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Ian H Frazer
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia.
| |
Collapse
|
30
|
Williams AC, Dunbar RIM. Big brains, meat, tuberculosis, and the nicotinamide switches: co-evolutionary relationships with modern repercussions? Int J Tryptophan Res 2013; 6:73-88. [PMID: 24250227 PMCID: PMC3825668 DOI: 10.4137/ijtr.s12838] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Meat-eating was a game changer for human evolution. We suggest that the limiting factors for expanding brains earlier were scarcities of nicotinamide and tryptophan. In humans and some other omnivores, lack of meat causes these deficiencies. Nicotinamide adenine dinucleotide (NADH) is necessary to synthesize adenosine triphosphate (ATP) via either glycolysis or via the mitochondrial respiratory chain. NAD consumption is also necessary for developmental and repair circuits. Inadequate supplies result in "de-evolutionary" brain atrophy, as seen with pellagra. If trophic nicotinamide/tryptophan was a "prime mover" in building bigger brains, back-up mechanisms should have evolved. One strategy may be to recruit extra gut symbionts that produce NADH precursors or export nicotinamide (though this may cause diarrhea). We propose a novel supplier TB that co-evolved early, which did not originally and does not now inevitably cause disease. TB has highly paradoxical immunology for a pathogen, and secretes and is inhibited by nicotinamide and its analogue, isoniazid. Sharp declines in TB and diarrhea correlated with increased meat intake in the past, suggesting that dietary vitamin B3 and tryptophan deficiencies (also associated with poor cognition and decreased lifespans) are still common where meat is unaffordable.
Collapse
Affiliation(s)
- Adrian C Williams
- Institute for Cognitive and Evolutionary Anthropology, University of Oxford, 64 Banbury Road, Oxford, OX2 6PN, UK
| | | |
Collapse
|
31
|
Nittby H, Ericsson P, Förnvik K, Strömblad S, Jansson L, Xue Z, Skagerberg G, Widegren B, Sjögren HO, Salford LG. Zebularine induces long-term survival of pancreatic islet allotransplants in streptozotocin treated diabetic rats. PLoS One 2013; 8:e71981. [PMID: 23991016 PMCID: PMC3753325 DOI: 10.1371/journal.pone.0071981] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 07/05/2013] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Coping with the immune rejection of allotransplants or autologous cells in patients with an active sensitization towards their autoantigens and autoimmunity presently necessitates life-long immune suppressive therapy acting on the immune system as a whole, which makes the patients vulnerable to infections and increases their risk of developing cancer. New technologies to induce antigen selective long-lasting immunosuppression or immune tolerance are therefore much needed. METHODOLOGY/PRINCIPAL FINDINGS The DNA demethylating agent Zebularine, previously demonstrated to induce expression of the genes for the immunosuppressive enzymes indolamine-2,3-deoxygenase-1 (IDO1) and kynureninase of the kynurenine pathway, is tested for capacity to suppress rejection of allotransplants. Allogeneic pancreatic islets from Lewis rats were transplanted under the kidney capsule of Fischer rats previously made diabetic by a streptozotocin injection (40 mg/kg). One group was treated with Zebularine (225 mg/kg) daily for 14 days from day 6 or 8 after transplantation, and a control group received no further treatment. Survival of the transplants was monitored by blood sugar measurements. Rats, normoglycemic for 90 days after allografting, were subjected to transplant removal by nephrectomy to confirm whether normoglycemia was indeed due to a surviving insulin producing transplant, or alternatively was a result of recovery of pancreatic insulin production in some toxin-treated rats. Of 9 Zebularine treated rats, 4 were still normoglycemic after 90 days and became hyperglycemic after nephrectomy. The mean length of normoglycemia in the Zebularine group was 67±8 days as compared to 14±3 days in 9 controls. Seven rats (2 controls and 5 Zebularine treated) were normoglycemic at 90 days due to pancreatic recovery as demonstrated by failure of nephrectomy to induce hyperglycemia. CONCLUSIONS/SIGNIFICANCE Zebularine treatment in vivo induces a long-lasting suppression of the immune destruction of allogeneic pancreatic islets resulting in protection of allograft function for more than 10 weeks after end of treatment.
Collapse
MESH Headings
- Animals
- Blood Glucose/metabolism
- Cytidine/analogs & derivatives
- Cytidine/pharmacology
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Experimental/surgery
- Gene Expression Regulation, Enzymologic/drug effects
- Graft Survival/drug effects
- Graft Survival/immunology
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Islets of Langerhans Transplantation/immunology
- Islets of Langerhans Transplantation/methods
- Male
- Rats
- Rats, Inbred F344
- Rats, Inbred Lew
- Rats, Wistar
- Reverse Transcriptase Polymerase Chain Reaction
- Spleen/drug effects
- Spleen/metabolism
- Time Factors
- Transplantation, Homologous
- Treatment Outcome
Collapse
Affiliation(s)
- Henrietta Nittby
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
- * E-mail:
| | - Peter Ericsson
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| | - Karolina Förnvik
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| | - Susanne Strömblad
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| | - Linda Jansson
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| | - Zhongtian Xue
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| | - Gunnar Skagerberg
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| | - Bengt Widegren
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| | - Hans-Olov Sjögren
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| | - Leif G. Salford
- Institute of Clinical Sciences, Department of Neurosurgery, the Rausing Laboratory, Lund University, Lund, Sweden
| |
Collapse
|
32
|
Dextromethorphan inhibits activations and functions in dendritic cells. Clin Dev Immunol 2013; 2013:125643. [PMID: 23781253 PMCID: PMC3679715 DOI: 10.1155/2013/125643] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 03/25/2013] [Indexed: 12/22/2022]
Abstract
Dendritic cells (DCs) play an important role in connecting innate and adaptive immunity. Thus, DCs have been regarded as a major target for the development of immunomodulators. In this study, we examined the effect of dextromethorphan (DXM), a common cough suppressant with a high safety profile, on the activation and function of DCs. In the presence of DXM, the LPS-induced expression of the costimulatory molecules in murine bone marrow-derived dendritic cells (BMDCs) was significantly suppressed. In addition, DXM treatment reduced the production of reactive oxygen species (ROS), proinflammatory cytokines, and chemokines in maturing BMDCs that were activated by LPS. Therefore, DXM abrogated the ability of LPS-stimulated DCs to induce Ag-specific T-cell activation, as determined by their decreased proliferation and IFN-γ secretion in mixed leukocyte cultures. Moreover, the inhibition of LPS-induced MAPK activation and NF-κB translocation may contribute to the suppressive effect of DXM on BMDCs. Remarkably, DXM decreased the LPS-induced surface expression of CD80, CD83, and HLA-DR and the secretion of IL-6 and IL-12 in human monocyte-derived dendritic cells (MDDCs). These findings provide a new insight into the impact of DXM treatment on DCs and suggest that DXM has the potential to be used in treating DC-related acute and chronic diseases.
Collapse
|
33
|
Regulatory macrophages as therapeutic targets and therapeutic agents in solid organ transplantation. Curr Opin Organ Transplant 2013; 17:332-42. [PMID: 22790067 DOI: 10.1097/mot.0b013e328355a979] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW This review aims to provide a basic introduction to human macrophage biology and an appreciation of the diverse roles played by macrophage subsets in allograft damage and repair. Current and future strategies for therapeutically manipulating macrophage behaviour are discussed. RECENT FINDINGS Macrophages are extremely versatile effector cells that exert both immunostimulatory and immunosuppressive effects. This adaptability cannot be explained by differentiation into committed sublineages, but instead reflects the ability of macrophages to rapidly transition between states of functional polarisation. Consequently, categorisation of macrophage subpopulations is not straightforward and this, in turn, creates difficulties in studying their pathophysiology. Nevertheless, particular macrophage subpopulations have been implicated in exacerbating or attenuating ischaemia-reperfusion injury, rejection reactions and allograft fibrosis. Three general strategies for therapeutically targeting macrophages can be envisaged, namely, depletional approaches, in-situ repolarisation towards a regulatory or tissue-reparative phenotype, and ex-vivo generation of regulatory macrophages (M reg) as a cell-based therapy. SUMMARY As critical determinants of the local and systemic immune response to solid organ allografts, macrophage subpopulations represent attractive therapeutic targets. Rapid progress is being made in the implementation of novel macrophage-targeted therapies, particularly in the use of ex-vivo-generated M regs as a cell-based medicinal product.
Collapse
|
34
|
Decreased expression of indoleamine 2,3-dioxygenase 1 in dendritic cells contributes to impaired regulatory T cell development in immune thrombocytopenia. Ann Hematol 2012; 92:67-78. [DOI: 10.1007/s00277-012-1556-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 08/13/2012] [Indexed: 12/31/2022]
|
35
|
Zheng X, Koropatnick J, Chen D, Velenosi T, Ling H, Zhang X, Jiang N, Navarro B, Ichim TE, Urquhart B, Min W. Silencing IDO in dendritic cells: a novel approach to enhance cancer immunotherapy in a murine breast cancer model. Int J Cancer 2012; 132:967-77. [PMID: 22870862 DOI: 10.1002/ijc.27710] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 06/14/2012] [Indexed: 12/15/2022]
Abstract
Cancer immunotherapeutic agents (vaccines) in the form of antigen-loaded dendritic cells (DCs) reached an important milestone with the recent approval of Provenge, the first DC vaccine for treatment of prostate cancer. Although this heralds a new era of tumor immunotherapy, it also highlights the compelling need to optimize such DC-based therapies as they are increasingly tested and used to treat human patients. In this study we sought to augment and enhance the antitumor activity of a DC-based vaccine using siRNA to silence expression of immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO) in DCs. We report here that DCs loaded with tumor antigens, but with siRNA-silenced IDO expression, were introduced into 4T1 breast tumor-bearing mice, the treatment: (i) lengthened the time required for tumor onset, (ii) decreased tumor size compared to tumors grown for equal lengths of time in mice treated with antigen-loaded DCs without IDO silencing and (iii) reduced CD4(+) and CD8(+) T cell apoptosis. Furthermore, immunization with IDO-silenced DCs enhanced tumor antigen-specific T cell proliferation and CTL activity, and decreased numbers of CD4(+) CD25(+) Foxp3(+) T(reg). This study provides evidence to support silencing of immunosuppressive genes (IDO) as an effective strategy to enhance the efficacy of DC-based cancer immunotherapeutic.
Collapse
Affiliation(s)
- Xiufen Zheng
- Department of Surgery, University of Western Ontario, London, Ontario, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Orabona C, Pallotta MT, Grohmann U. Different partners, opposite outcomes: a new perspective of the immunobiology of indoleamine 2,3-dioxygenase. Mol Med 2012; 18:834-42. [PMID: 22481272 DOI: 10.2119/molmed.2012.00029] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 03/30/2012] [Indexed: 01/07/2023] Open
Abstract
Indoleamine 2,3-dioxygenase (IDO), a metabolic enzyme that catalyzes tryptophan conversion into kynurenines, is a crucial regulator of immunity. Altered IDO activity is often associated with pathology, including neoplasia and autoimmunity. IDO is highly expressed in dendritic cells (DCs) that exploit the enzyme's activity and the production of tryptophan catabolites to regulate immune responses by acting on several cell types, including T lymphocytes, of which they promote a regulatory phenotype. IDO also contains immunoreceptor tyrosine-based inhibitory motifs (ITIMs) that, once bound by distinct molecular partners, will either promote degradation or initiate signaling activity and self-maintenance of the enzyme. We here discuss how ITIM-dependent molecular events can affect the functional plasticity of IDO by modifying the protein half-life and its enzymic and nonenzymic functions.
Collapse
Affiliation(s)
- Ciriana Orabona
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
| | | | | |
Collapse
|
37
|
Wolowczuk I, Hennart B, Leloire A, Bessede A, Soichot M, Taront S, Caiazzo R, Raverdy V, Pigeyre M, Guillemin GJ, Allorge D, Pattou F, Froguel P, Poulain-Godefroy O. Tryptophan metabolism activation by indoleamine 2,3-dioxygenase in adipose tissue of obese women: an attempt to maintain immune homeostasis and vascular tone. Am J Physiol Regul Integr Comp Physiol 2012; 303:R135-43. [DOI: 10.1152/ajpregu.00373.2011] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Human obesity is characterized by chronic low-grade inflammation in white adipose tissue and is often associated with hypertension. The potential induction of indoleamine 2,3-dioxygenase-1 (IDO1), the rate-limiting enzyme in tryptophan/kynurenine degradation pathway, by proinflammatory cytokines, could be associated with these disorders but has remained unexplored in obesity. Using immunohistochemistry, we detected IDO1 expression in white adipose tissue of obese patients, and we focused on its contribution in the regulation of vascular tone and on its immunoregulatory effects. Concentrations of tryptophan and kynurenine were measured in sera of 36 obese and 15 lean women. The expression of IDO1 in corresponding omental and subcutaneous adipose tissues and liver was evaluated. Proinflammatory markers and T-cell subsets were analyzed in adipose tissue via the expression of CD14, IL-18, CD68, TNFα, CD3ε, FOXP3 [a regulatory T-cell (Treg) marker] and RORC (a Th17 marker). In obese subjects, the ratio of kynurenine to tryptophan, which reflects IDO1 activation, is higher than in lean subjects. Furthermore, IDO1 expression in both adipose tissues and liver is increased and is inversely correlated with arterial blood pressure. Inflammation is associated with a T-cell infiltration in obese adipose tissue, with predominance of Th17 in the omental compartment and of Treg in the subcutaneous depot. The Th17/Treg balance is decreased in subcutaneous fat and correlates with IDO1 activation. In contrast, in the omental compartment, despite IDO1 activation, the Th17/Treg balance control is impaired. Taken together, our results suggest that IDO1 activation represents a local compensatory mechanism to limit obesity-induced inflammation and hypertension.
Collapse
Affiliation(s)
- Isabelle Wolowczuk
- Centre National de la Recherche Scientifique, UMR 8199, Pasteur Institute, Université Lille Nord de France, Lille, France
| | - Benjamin Hennart
- EA4483, Faculty of Medicine, Université Lille Nord de France, Lille, France
| | - Audrey Leloire
- Centre National de la Recherche Scientifique, UMR 8199, Pasteur Institute, Université Lille Nord de France, Lille, France
| | - Alban Bessede
- Neuroinflammation Group, Department of Pharmacology, University of New South Wales, Sydney, New South Wales, Australia; and
| | - Marion Soichot
- EA4483, Faculty of Medicine, Université Lille Nord de France, Lille, France
| | - Solenne Taront
- Centre National de la Recherche Scientifique, UMR 8199, Pasteur Institute, Université Lille Nord de France, Lille, France
| | - Robert Caiazzo
- Institut National de la Santé et de la Recherche Médicale, UMR 859, Université Lille Nord de France, Lille, France
| | - Violeta Raverdy
- Institut National de la Santé et de la Recherche Médicale, UMR 859, Université Lille Nord de France, Lille, France
| | - Marie Pigeyre
- Institut National de la Santé et de la Recherche Médicale, UMR 859, Université Lille Nord de France, Lille, France
| | - Gilles J. Guillemin
- Neuroinflammation Group, Department of Pharmacology, University of New South Wales, Sydney, New South Wales, Australia; and
| | - Delphine Allorge
- EA4483, Faculty of Medicine, Université Lille Nord de France, Lille, France
| | - François Pattou
- Institut National de la Santé et de la Recherche Médicale, UMR 859, Université Lille Nord de France, Lille, France
| | - Philippe Froguel
- Centre National de la Recherche Scientifique, UMR 8199, Pasteur Institute, Université Lille Nord de France, Lille, France
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London, United Kingdom
| | - Odile Poulain-Godefroy
- Centre National de la Recherche Scientifique, UMR 8199, Pasteur Institute, Université Lille Nord de France, Lille, France
| | | |
Collapse
|
38
|
Nicotinamide, NAD(P)(H), and Methyl-Group Homeostasis Evolved and Became a Determinant of Ageing Diseases: Hypotheses and Lessons from Pellagra. Curr Gerontol Geriatr Res 2012; 2012:302875. [PMID: 22536229 PMCID: PMC3318212 DOI: 10.1155/2012/302875] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 12/19/2011] [Indexed: 01/22/2023] Open
Abstract
Compartmentalized redox faults are common to ageing diseases. Dietary constituents are catabolized to NAD(H) donating electrons producing proton-based bioenergy in coevolved, cross-species and cross-organ networks. Nicotinamide and NAD deficiency from poor diet or high expenditure causes pellagra, an ageing and dementing disorder with lost robustness to infection and stress. Nicotinamide and stress induce Nicotinamide-N-methyltransferase (NNMT) improving choline retention but consume methyl groups. High NNMT activity is linked to Parkinson's, cancers, and diseases of affluence. Optimising nicotinamide and choline/methyl group availability is important for brain development and increased during our evolution raising metabolic and methylome ceilings through dietary/metabolic symbiotic means but strict energy constraints remain and life-history tradeoffs are the rule. An optimal energy, NAD and methyl group supply, avoiding hypo and hyper-vitaminoses nicotinamide and choline, is important to healthy ageing and avoids utilising double-edged symbionts or uncontrolled autophagy or reversions to fermentation reactions in inflammatory and cancerous tissue that all redistribute NAD(P)(H), but incur high allostatic costs.
Collapse
|
39
|
Identification of a variable number of tandem repeats polymorphism and characterization of LEF-1 response elements in the promoter of the IDO1 gene. PLoS One 2011; 6:e25470. [PMID: 21980470 PMCID: PMC3181322 DOI: 10.1371/journal.pone.0025470] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 09/05/2011] [Indexed: 11/19/2022] Open
Abstract
Background Indoleamine 2,3-dioxygenase (IDO) catalyzes the first and rate-limiting step of the kynurenine pathway that is an important component of immunomodulatory and neuromodulatory processes. The IDO1 gene is highly inducible by IFN-γ and TNF-α through interaction with cis-acting regulatory elements of the promoter region. Accordingly, functional polymorphisms in the IDO1 promoter could partly explain the interindividual variability in IDO expression that has been previously documented. Methodology/Principal Findings A PCR-sequencing strategy, applied to DNA samples from healthy Caucasians, allowed us to identify a VNTR polymorphism in the IDO1 promoter, which correlates significantly with serum tryptophan concentration, controlled partially by IDO activity, in female subjects, but not in males. Although this VNTR does not appear to affect basal or cytokine-induced promoter activity in gene reporter assays, it contains novel cis-acting elements. Three putative LEF-1 binding sites, one being located within the VNTR repeat motif, were predicted in silico and confirmed by chromatin immunoprecipitation. Overexpression of LEF-1 in luciferase assays confirmed an interaction between LEF-1 and the predicted transcription factor binding sites, and modification of the LEF-1 core sequence within the VNTR repeat motif, by site-directed mutagenesis, resulted in an increase in promoter activity. Conclusions/Significance The identification of a VNTR in the IDO1 promoter revealed a cis-acting element interacting with the most downstream factor of the Wnt signaling pathway, suggesting novel mechanisms of regulation of IDO1 expression. These data offer new insights, and suggest further studies, into the role of IDO in various pathological conditions, particularly in cancer where IDO and the Wnt pathway are strongly dysregulated.
Collapse
|
40
|
Croitoru-Lamoury J, Lamoury FMJ, Caristo M, Suzuki K, Walker D, Takikawa O, Taylor R, Brew BJ. Interferon-γ regulates the proliferation and differentiation of mesenchymal stem cells via activation of indoleamine 2,3 dioxygenase (IDO). PLoS One 2011; 6:e14698. [PMID: 21359206 PMCID: PMC3040184 DOI: 10.1371/journal.pone.0014698] [Citation(s) in RCA: 175] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Accepted: 01/13/2011] [Indexed: 12/15/2022] Open
Abstract
The kynurenine pathway (KP) of tryptophan metabolism is linked to antimicrobial activity and modulation of immune responses but its role in stem cell biology is unknown. We show that human and mouse mesenchymal and neural stem cells (MSCs and NSCs) express the complete KP, including indoleamine 2,3 dioxygenase 1 (IDO) and IDO2, that it is highly regulated by type I (IFN-β) and II interferons (IFN-γ), and that its transcriptional modulation depends on the type of interferon, cell type and species. IFN-γ inhibited proliferation and altered human and mouse MSC neural, adipocytic and osteocytic differentiation via the activation of IDO. A functional KP present in MSCs, NSCs and perhaps other stem cell types offers novel therapeutic opportunities for optimisation of stem cell proliferation and differentiation.
Collapse
|