1
|
Wang X. Clinical and molecular prognostic nomograms for patients with papillary renal cell carcinoma. Discov Oncol 2024; 15:780. [PMID: 39692801 DOI: 10.1007/s12672-024-01669-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024] Open
Abstract
OBJECTIVE To summarize the clinicopathological characteristics and prognostic factors of papillary renal cell carcinoma (pRCC) and to construct clinical and molecular prognostic nomograms using existing databases. METHODS Clinical prognostic models were developed using the Surveillance, Epidemiology, and End Results (SEER) database, while molecular prognostic models were constructed using The Cancer Genome Atlas (TCGA) database. Cox regression and LASSO regression were employed to identify clinicopathological features and molecular markers related to prognosis. The accuracy of the prognostic models was assessed using ROC curves, C-index, decision curve analysis (DCA) curves, and calibration plots. RESULTS In the 2004-2015 SEER cohort, Cox regression analysis revealed that age, grade, AJCC stage, N stage, M stage, and surgery were independent predictors of overall survival (OS) and cancer-specific survival (CSS) in pRCC patients. ROC curves, C-index, and DCA curves indicated that the prognostic nomogram based on clinical independent predictors had better predictive ability than TNM staging and SEER staging. Additionally, in the TCGA cohort, M stage, clinical stage, and the molecular markers IDO1 and PLK1 were identified as independent risk factors. The prognostic nomogram based on molecular independent risk factors effectively predicted the 3-year and 5-year OS and CSS for pRCC patients. CONCLUSIONS The clinical and molecular nomograms constructed in this study provide robust predictive tools for individualized prognosis in pRCC patients, offering better accuracy than traditional staging systems.
Collapse
Affiliation(s)
- Xuhui Wang
- Department of Urology, The Affiliated People's Hospital of Ningbo University, No.251, Baizhang East Road, Yinzhou District, Ningbo, 315040, China.
| |
Collapse
|
2
|
Xu R, He X, Xu J, Yu G, Wu Y. Immunometabolism: signaling pathways, homeostasis, and therapeutic targets. MedComm (Beijing) 2024; 5:e789. [PMID: 39492834 PMCID: PMC11531657 DOI: 10.1002/mco2.789] [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: 10/19/2023] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 11/05/2024] Open
Abstract
Immunometabolism plays a central role in sustaining immune system functionality and preserving physiological homeostasis within the organism. During the differentiation and activation, immune cells undergo metabolic reprogramming mediated by complex signaling pathways. Immune cells maintain homeostasis and are influenced by metabolic microenvironmental cues. A series of immunometabolic enzymes modulate immune cell function by metabolizing nutrients and accumulating metabolic products. These enzymes reverse immune cells' differentiation, disrupt intracellular signaling pathways, and regulate immune responses, thereby influencing disease progression. The huge population of immune metabolic enzymes, the ubiquity, and the complexity of metabolic regulation have kept the immune metabolic mechanisms related to many diseases from being discovered, and what has been revealed so far is only the tip of the iceberg. This review comprehensively summarized the immune metabolic enzymes' role in multiple immune cells such as T cells, macrophages, natural killer cells, and dendritic cells. By classifying and dissecting the immunometabolism mechanisms and the implications in diseases, summarizing and analyzing advancements in research and clinical applications of the inhibitors targeting these enzymes, this review is intended to provide a new perspective concerning immune metabolic enzymes for understanding the immune system, and offer novel insight into future therapeutic interventions.
Collapse
Affiliation(s)
- Rongrong Xu
- National Key Laboratory of Immunity and Inflammation & Institute of ImmunologyCollege of Basic Medical SciencesNaval Medical UniversityShanghaiChina
- School of Life SciencesFudan UniversityShanghaiChina
| | - Xiaobo He
- National Key Laboratory of Immunity and Inflammation & Institute of ImmunologyCollege of Basic Medical SciencesNaval Medical UniversityShanghaiChina
| | - Jia Xu
- National Key Laboratory of Immunity and Inflammation & Institute of ImmunologyCollege of Basic Medical SciencesNaval Medical UniversityShanghaiChina
| | - Ganjun Yu
- National Key Laboratory of Immunity and Inflammation & Institute of ImmunologyCollege of Basic Medical SciencesNaval Medical UniversityShanghaiChina
| | - Yanfeng Wu
- National Key Laboratory of Immunity and Inflammation & Institute of ImmunologyCollege of Basic Medical SciencesNaval Medical UniversityShanghaiChina
| |
Collapse
|
3
|
Pan L, Zhou G, Wei G, Zhao Q, Wang Y, Chen Q, Xiao Q, Song Y, Liang X, Zou Z, Li X, Xiong X. Associations between Sjogren syndrome and psychiatric disorders in European populations: a 2-sample bidirectional Mendelian randomization study. Front Psychiatry 2024; 15:1465381. [PMID: 39479595 PMCID: PMC11521899 DOI: 10.3389/fpsyt.2024.1465381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
Background Psychiatric disorders, such as major depressive disorder (MDD), anxiety disorder (AD), bipolar disorder (BD), and schizophrenia (SCZ), are disturbances in brain activity that lead to disorders of cognition, behavior, and emotion regulation. Among Sjogren syndrome (SS) patients, psychiatric disorders are more prevalent than in the general population. Identifying associated risk factors can provide new evidence for clinical diagnosis and treatment. Methods We selected genetic instruments based on published genome-wide association studies (GWASs) to determine predisposition. Then, we conducted a 2-sample bidirectional Mendelian randomization (MR) analysis to explore the potential causal associations between SS and four major psychiatric disorders. The primary analysis was performed using MR with the inverse-variance weighted method. Confirmation was achieved through Steiger filtering and testing to determine the causal direction. Sensitivity analyses were conducted using MR-Egger, MR-PRESSO, and "leave-one-out" method methods. Results Our study showed that SS was linked to BD and SCZ, indicating that individuals with SS may have a reduced risk of developing BD (IVW: OR = 0.940, P=0.014) and SCZ (IVW: OR = 0.854, P=1.47*10-4), while there was no causal relationship between SS and MDD or AD. MR-Egger regression shows no evidence of pleiotropy (BD: intercept = 0.007, p = 0.774; SCZ: intercept = 0.051, p = 0.209). The same as the MR-PRESSO analysis (BD: global test p = 1.000; SCZ: global test p = 0.160). However, the results from the leave-one-out analysis demonstrated instability. Specifically, after excluding SNP rs3117581, the effects on BD and SCZ were found to be non-significant, suggesting the potential influence of unrecognized confounding factors. The results of the reverse MR show that four major psychiatric disorders had no causal effects on SS. Conclusions Our research findings demonstrate a causal relationship between SS and SCZ, as well as between SS and BD. There are no causal effects between the four major psychiatric disorders and SS. These findings suggest that SS may have the potential to reduce the risk of both psychiatric disorders. This study provides new insight for their prevention and treatment.
Collapse
Affiliation(s)
- Lingai Pan
- Department of Intensive Care Unit, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Guangpeng Zhou
- Endocrine Department, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Guocui Wei
- Department of Pharmacy, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qian Zhao
- Department of Pharmacy, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yanping Wang
- Department of Pharmacy, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qianlan Chen
- Department of Pharmacy, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qing Xiao
- Department of Pharmacy, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yujie Song
- Department of Pharmacy, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiangui Liang
- Department of Pharmacy, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhili Zou
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiuxia Li
- Department of Pharmacy, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xuan Xiong
- Department of Pharmacy, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
4
|
Ni M, Cui J, Yang X, Ding Y, Zhao P, Hu T, Zhan Y, Kang Q, Hu X, Zhao J, Xu Y, Chen L, Liu M, Zhao M, Zhang F, Huang S, Li Y, Yang X, Zhang L, Zhang T, Deng B, Yang B, Lu D, Wang J. Dual roles of CD11b +CD33 +HLA-DR -/lowCD14 - myeloid-derived suppressor cells with a granulocytic morphology following allogeneic hematopoietic stem cell transplantation: from inflammation promoters to immune suppressors within 90 days. Front Immunol 2024; 15:1403272. [PMID: 39040102 PMCID: PMC11260618 DOI: 10.3389/fimmu.2024.1403272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
Introduction Granulocytic myeloid-derived suppressor cells (G-MDSCs) show fast recovery following allogeneic hematopoietic stem cell transplantation (allo-HSCT) constituting the major part of peripheral blood in the early phase. Although G-MDSCs mediate immune suppression through multiple mechanisms, they may also promote inflammation under specific conditions. Methods G-MDSCs were isolated from 82 patients following allo-HSCT within 90 days after allo-HSCT, and their interactions with autologous CD3+ T-cells were examined. T-cell proliferation was assessed by flow cytometry following CFSE staining, while differentiation and interferon-γ secretion were characterized using chemokine receptor profiling and ELISpot assays, respectively. NK cell cytotoxicity was evaluated through co-culture with K562 cells. An aGVHD xenogeneic model in humanized mice was employed to study the in vivo effects of human leukocytes. Furthermore, transcriptional alterations in G-MDSCs were analyzed via RNA sequencing to investigate functional transitions. Results G-MDSCs promoted inflammation in the early-stage, by facilitating cytokine secretion and proliferation of T cells, as well as their differentiation into pro-inflammatory T helper subsets. At day 28, patients with a higher number of G-MDSCs exhibited an increased risk of developing grades II-IV aGvHD. Besides, adoptive transfer of G-MDSCs from patients at day 28 into humanized mice exacerbated aGvHD. However, at day 90, G-MDSCs led to immunosuppression, characterized by upregulated expression of indoleamine 2,3-dioxygenase gene and interleukin-10 secretion, coupled with the inhibition of T cell proliferation. Furthermore, transcriptional analysis of G-MDSCs at day 28 and day 90 revealed that 1445 genes were differentially expressed. These genes were associated with various pathways, revealing the molecular signatures of early post-transplant differentiation in G-MDSCs. In addition, genes linked to the endoplasmic reticulum stress were upregulated in patients without aGvHD. The acquisition of immunosuppressive function by G-MDSCs may depend on the activation of CXCL2 and DERL1 genes. Conclusion Our findings revealed the alteration in the immune characteristics of G-MDSCs within the first 90 days post-allo-HSCT. Moreover, the quantity of G-MDSCs at day 28 may serve as a predictive indicator for the development of aGvHD.
Collapse
Affiliation(s)
- Ming Ni
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jing Cui
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xin Yang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Hematology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, China
| | - Yuntian Ding
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Peng Zhao
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Tianzhen Hu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yun Zhan
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qian Kang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xiuying Hu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jiangyuan Zhao
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yao Xu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Lu Chen
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Min Liu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Mei Zhao
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Fengqi Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shisi Huang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ya Li
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xueying Yang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Luxin Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Tianzhuo Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Bo Deng
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Bing Yang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Deqin Lu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
| | - Jishi Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
5
|
Sayour EJ, Boczkowski D, Mitchell DA, Nair SK. Cancer mRNA vaccines: clinical advances and future opportunities. Nat Rev Clin Oncol 2024; 21:489-500. [PMID: 38760500 DOI: 10.1038/s41571-024-00902-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2024] [Indexed: 05/19/2024]
Abstract
mRNA vaccines have been revolutionary in terms of their rapid development and prevention of SARS-CoV-2 infections during the COVID-19 pandemic, and this technology has considerable potential for application to the treatment of cancer. Compared with traditional cancer vaccines based on proteins or peptides, mRNA vaccines reconcile the needs for both personalization and commercialization in a manner that is unique to each patient but not beholden to their HLA haplotype. A further advantage of mRNA vaccines is the availability of engineering strategies to improve their stability while retaining immunogenicity, enabling the induction of complementary innate and adaptive immune responses. Thus far, no mRNA-based cancer vaccines have received regulatory approval, although several phase I-II trials have yielded promising results, including in historically poorly immunogenic tumours. Furthermore, many early phase trials testing a wide range of vaccine designs are currently ongoing. In this Review, we describe the advantages of cancer mRNA vaccines and advances in clinical trials using both cell-based and nanoparticle-based delivery methods, with discussions of future combinations and iterations that might optimize the activity of these agents.
Collapse
Affiliation(s)
- Elias J Sayour
- Preston A. Wells Jr. Center for Brain Tumour Therapy, University of Florida, Gainesville, FL, USA
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - David Boczkowski
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Duane A Mitchell
- Preston A. Wells Jr. Center for Brain Tumour Therapy, University of Florida, Gainesville, FL, USA
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Smita K Nair
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA.
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA.
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA.
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
6
|
Lee J, Jang J, Cha SR, Lee SB, Hong SH, Bae HS, Lee YJ, Yang SR. Recombinant Human Bone Morphogenetic Protein-2 Priming of Mesenchymal Stem Cells Ameliorate Acute Lung Injury by Inducing Regulatory T Cells. Immune Netw 2023; 23:e48. [PMID: 38188599 PMCID: PMC10767548 DOI: 10.4110/in.2023.23.e48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) possess immunoregulatory properties and their regulatory functions represent a potential therapy for acute lung injury (ALI). However, uncertainties remain with respect to defining MSCs-derived immunomodulatory pathways. Therefore, this study aimed to investigate the mechanism underlying the enhanced effect of human recombinant bone morphogenic protein-2 (rhBMP-2) primed ES-MSCs (MSCBMP2) in promoting Tregs in ALI mice. MSC were preconditioned with 100 ng/ml rhBMP-2 for 24 h, and then administrated to mice by intravenous injection after intratracheal injection of 1 mg/kg LPS. Treating MSCs with rhBMP-2 significantly increased cellular proliferation and migration, and cytokines array reveled that cytokines release by MSCBMP2 were associated with migration and growth. MSCBMP2 ameliorated LPS induced lung injury and reduced myeloperoxidase activity and permeability in mice exposed to LPS. Levels of inducible nitric oxide synthase were decreased while levels of total glutathione and superoxide dismutase activity were further increased via inhibition of phosphorylated STAT1 in ALI mice treated with MSCBMP2. MSCBMP2 treatment increased the protein level of IDO1, indicating an increase in Treg cells, and Foxp3+CD25+ Treg of CD4+ cells were further increased in ALI mice treated with MSCBMP2. In co-culture assays with MSCs and RAW264.7 cells, the protein level of IDO1 was further induced in MSCBMP2. Additionally, cytokine release of IL-10 was enhanced while both IL-6 and TNF-α were further inhibited. In conclusion, these findings suggest that MSCBMP2 has therapeutic potential to reduce massive inflammation of respiratory diseases by promoting Treg cells.
Collapse
Affiliation(s)
- Jooyeon Lee
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Jimin Jang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Sang-Ryul Cha
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Se Bi Lee
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Han-Sol Bae
- Cellular Therapeutics Team, Daewoong Pharmaceutical, Yongin 17028, Korea
| | - Young Jin Lee
- Cellular Therapeutics Team, Daewoong Pharmaceutical, Yongin 17028, Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
7
|
Aderinto N, Abdulbasit MO, Tangmi ADE, Okesanya JO, Mubarak JM. Unveiling the growing significance of metabolism in modulating immune cell function: exploring mechanisms and implications; a review. Ann Med Surg (Lond) 2023; 85:5511-5522. [PMID: 37915697 PMCID: PMC10617839 DOI: 10.1097/ms9.0000000000001308] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 09/06/2023] [Indexed: 11/03/2023] Open
Abstract
Immunometabolism has emerged as a rapidly growing field of research, holding significant promise for personalised medicine and precision immunotherapy. This review explores the intricate relationship between immune function and metabolic processes, emphasising their profound impact on various immune-related disorders. Understanding how metabolic dysregulation contributes to the pathogenesis of these disorders remains a critical research gap. Therefore, this review aims to bridge that gap by examining the key metabolic pathways involved and their specific implications in immune cell function. Key metabolic pathways, including glycolysis, mitochondrial metabolism, fatty acid metabolism, and amino acid metabolism, are discussed in the context of immune cell function. Dysregulation of these pathways can disrupt immune cell activation, differentiation, and overall function, contributing to disease pathogenesis. Understanding these metabolic alterations' molecular mechanisms is essential for developing targeted therapeutic interventions. The review also emphasises the importance of personalised medicine in immune-related disorders. The unique metabolic profiles of individuals can influence treatment outcomes, highlighting the need for tailored approaches. Integrating metabolic profiling into clinical practice can enhance treatment efficacy and improve patient outcomes. Investigating the clinical significance of immunometabolism in diverse disease contexts will facilitate the translation of research findings into clinical practice. Moreover, refining treatment strategies based on individual metabolic profiles will contribute to advancing precision immunotherapy.
Collapse
Affiliation(s)
- Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso
| | | | | | | | | |
Collapse
|
8
|
Azimnasab-Sorkhabi P, Soltani-Asl M, Yoshinaga TT, Zaidan Dagli ML, Massoco CDO, Kfoury Junior JR. Indoleamine-2,3 dioxygenase: a fate-changer of the tumor microenvironment. Mol Biol Rep 2023:10.1007/s11033-023-08469-3. [PMID: 37217614 DOI: 10.1007/s11033-023-08469-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 04/18/2023] [Indexed: 05/24/2023]
Abstract
Indoleamine-2,3 dioxygenase is a rate-limiting enzyme in the tryptophan catabolism in kynurenine pathways that has an immunosuppressive effect and supports cancer cells to evade the immune system in different cancer types. Diverse cytokines and pathways upregulate the production of indoleamine-2,3 dioxygenase enzymes in the tumor microenvironment and cause more production and activity of this enzyme. Ultimately, this situation results in anti-tumor immune suppression which is in favor of tumor growth. Several inhibitors such as 1-methyl-tryptophan have been introduced for indoleamine-2,3 dioxygenase enzyme and some of them are widely utilized in pre-clinical and clinical trials. Importantly at the molecular level, indoleamine-2,3 dioxygenase is positioned in a series of intricate signaling and molecular networks. Here, the main objective is to provide a focused view of indoleamine-2,3 dioxygenase enhancer pathways and propose further studies to cover the gap in available information on the function of indoleamine-2,3 dioxygenase enzyme in the tumor microenvironment.
Collapse
Affiliation(s)
- Parviz Azimnasab-Sorkhabi
- Department of Surgery, School of Veterinary Medicine and Animal Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| | - Maryam Soltani-Asl
- Department of Surgery, School of Veterinary Medicine and Animal Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Túlio Teruo Yoshinaga
- Department of Surgery, School of Veterinary Medicine and Animal Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Maria Lucia Zaidan Dagli
- Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Cristina de Oliveira Massoco
- Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Jose Roberto Kfoury Junior
- Department of Surgery, School of Veterinary Medicine and Animal Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
9
|
Mannarino MR, Bianconi V, Scalisi G, Franceschini L, Manni G, Cucci A, Bagaglia F, Mencarelli G, Giglioni F, Ricciuti D, Figorilli F, Pieroni B, Cosentini E, Padiglioni E, Colangelo C, Fuchs D, Puccetti P, Follenzi A, Pirro M, Gargaro M, Fallarino F. A tryptophan metabolite prevents depletion of circulating endothelial progenitor cells in systemic low-grade inflammation. Front Immunol 2023; 14:964660. [PMID: 37081894 PMCID: PMC10110845 DOI: 10.3389/fimmu.2023.964660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 03/20/2023] [Indexed: 04/07/2023] Open
Abstract
BackgroundChronic systemic inflammation reduces the bioavailability of circulating endothelial progenitor cells (EPCs). Indoleamine 2,3-dioxygenase 1 (IDO1), a key enzyme of immune tolerance catalyzing the initial step of tryptophan degradation along the so-called l-kynurenine (l-kyn) pathway, that is induced by inflammatory stimuli and exerts anti-inflammatory effects. A specific relationship between IDO1 activity and circulating EPC numbers has not yet been investigated.MethodsIn this study, circulating EPCs were examined in mice treated with low doses of lipopolysaccharide (LPS) to mimic low-grade inflammation. Moreover, the association between IDO1 activity and circulating EPCs was studied in a cohort of 277 patients with variable systemic low-grade inflammation.ResultsRepeated low doses of LPS caused a decrease in circulating EPCs and l-kyn supplementation, mimicking IDO1 activation, significantly increased EPC numbers under homeostatic conditions preventing EPC decline in low-grade endotoxemia. Accordingly, in patients with variable systemic low-grade inflammation, there was a significant interaction between IDO1 activity and high-sensitivity C-reactive protein (hs-CRP) in predicting circulating EPCs, with high hs-CRP associated with significantly lower EPCs at low IDO1 activity but not at high IDO1 activity.InterpretationOverall, these findings demonstrate that systemic low-grade inflammation reduces circulating EPCs. However, high IDO1 activity and l-kyn supplementation limit circulating EPC loss in low-grade inflammation.
Collapse
Affiliation(s)
| | - Vanessa Bianconi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- *Correspondence: Vanessa Bianconi, ; Marco Gargaro, ; Francesca Fallarino,
| | - Giulia Scalisi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Luca Franceschini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giorgia Manni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Alessia Cucci
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Novara, Italy
| | - Francesco Bagaglia
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giulia Mencarelli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Francesco Giglioni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Doriana Ricciuti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Filippo Figorilli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Benedetta Pieroni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Elena Cosentini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Cecilia Colangelo
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Paolo Puccetti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Antonia Follenzi
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Novara, Italy
| | - Matteo Pirro
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marco Gargaro
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- *Correspondence: Vanessa Bianconi, ; Marco Gargaro, ; Francesca Fallarino,
| | - Francesca Fallarino
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- *Correspondence: Vanessa Bianconi, ; Marco Gargaro, ; Francesca Fallarino,
| |
Collapse
|
10
|
Kim SK, Cho SW. The Evasion Mechanisms of Cancer Immunity and Drug Intervention in the Tumor Microenvironment. Front Pharmacol 2022; 13:868695. [PMID: 35685630 PMCID: PMC9171538 DOI: 10.3389/fphar.2022.868695] [Citation(s) in RCA: 211] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/08/2022] [Indexed: 12/17/2022] Open
Abstract
Recently, in the field of cancer treatment, the paradigm has changed to immunotherapy that activates the immune system to induce cancer attacks. Among them, immune checkpoint inhibitors (ICI) are attracting attention as excellent and continuous clinical results. However, it shows not only limitations such as efficacy only in some patients or some indications, but also side-effects and resistance occur. Therefore, it is necessary to understand the factors of the tumor microenvironment (TME) that affect the efficacy of immunotherapy, that is, the mechanism by which cancer grows while evading or suppressing attacks from the immune system within the TME. Tumors can evade attacks from the immune system through various mechanisms such as restricting antigen recognition, inhibiting the immune system, and inducing T cell exhaustion. In addition, tumors inhibit or evade the immune system by accumulating specific metabolites and signal factors within the TME or limiting the nutrients available to immune cells. In order to overcome the limitations of immunotherapy and develop effective cancer treatments and therapeutic strategies, an approach is needed to understand the functions of cancer and immune cells in an integrated manner based on the TME. In this review, we will examine the effects of the TME on cancer cells and immune cells, especially how cancer cells evade the immune system, and examine anti-cancer strategies based on TME.
Collapse
Affiliation(s)
- Seong Keun Kim
- Cellus Inc., Seoul, South Korea
- *Correspondence: Seong Keun Kim, ; Sun Wook Cho,
| | - Sun Wook Cho
- Cellus Inc., Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- *Correspondence: Seong Keun Kim, ; Sun Wook Cho,
| |
Collapse
|
11
|
Guo R, Liu Y, Xu N, Ling G, Zhang P. Multifunctional nanomedicines for synergistic photodynamic immunotherapy based on tumor immune microenvironment. Eur J Pharm Biopharm 2022; 173:103-120. [DOI: 10.1016/j.ejpb.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 01/23/2022] [Accepted: 03/07/2022] [Indexed: 12/07/2022]
|
12
|
Nanoparticle-based delivery strategies of multifaceted immunomodulatory RNA for cancer immunotherapy. J Control Release 2022; 343:564-583. [DOI: 10.1016/j.jconrel.2022.01.047] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/25/2022] [Accepted: 01/29/2022] [Indexed: 12/18/2022]
|
13
|
Eroğlu İ, Eroğlu BÇ, Güven GS. Altered tryptophan absorption and metabolism could underlie long-term symptoms in survivors of coronavirus disease 2019 (COVID-19). Nutrition 2021; 90:111308. [PMID: 34111831 PMCID: PMC8087860 DOI: 10.1016/j.nut.2021.111308] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/18/2021] [Accepted: 04/21/2021] [Indexed: 12/26/2022]
Abstract
The global pandemic of COVID-19 has been lasting for more than one year and there is little known about the long-term health effects of the disease. Long-COVID is a new term that is used to describe the enduring symptoms of COVID-19 survivors. Huang et al. reported that fatigue, muscle weakness, sleep disturbances, anxiety, and depression were the most common complaints in COVID-19 survivors after 6 months of the infection. A recent meta-analysis showed that 80% of COVID-19 survivors have developed at least one long-term symptom and the most common five were fatigue, headache, attention deficit disorder, hair loss, and dyspnea. In this paper, we discuss the hypothesis that altered tryptophan absorption and metabolism could be the main contributor to the long-term symptoms in COVID-19 survivors.
Collapse
Affiliation(s)
- İmdat Eroğlu
- Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey.
| | - Burcu Çelik Eroğlu
- Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Gülay Sain Güven
- Department of General Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
14
|
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
|
15
|
Zhao LP, Zheng RR, Huang JQ, Chen XY, Deng FA, Liu YB, Huang CY, Yu XY, Cheng H, Li SY. Self-Delivery Photo-Immune Stimulators for Photodynamic Sensitized Tumor Immunotherapy. ACS NANO 2020; 14:17100-17113. [PMID: 33236625 DOI: 10.1021/acsnano.0c06765] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Self-delivery of photosensitizer and immune modulator to tumor site is highly recommendable to improve the photodynamic immunotherapy yet remains challenging. Herein, self-delivery photoimmune stimulators (designated as iPSs) are developed for photodynamic sensitized tumor immunotherapy. Carrier-free iPSs are constructed by optimizing the noncovalent interactions between the pure drugs of chlorine e6 (Ce6) and NLG919, which avoid the excipients-raised toxicity and immunogenicity. Intravenously administrated iPSs prefer to passively accumulate on tumor tissues for a robust photodynamic therapy (PDT) with the induction of immunogenetic cell death (ICD) cascade to activate cytotoxic T lymphocytes (CTLs) and initiate antitumor immune response. Meanwhile, the concomitant delivery of NLG919 inhibits the activation of indoleamine 2,3-dioxygenase 1 (IDO-1) to reverse the immunosuppressive tumor microenvironment. Ultimately, the photodynamic sensitized immunotherapy with iPSs efficiently inhibit the primary and distant tumor growth with a low system toxicity, which would shed light on the development of self-delivery nanomedicine for clinical transformation in tumor precision therapy.
Collapse
Affiliation(s)
- Lin-Ping Zhao
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Rong-Rong Zheng
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Jia-Qi Huang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, P.R. China
| | - Xia-Yun Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Fu-An Deng
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Yi-Bin Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Chu-Yu Huang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Xi-Yong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Hong Cheng
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, P.R. China
| | - Shi-Ying Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P.R. China
| |
Collapse
|
16
|
Meireson A, Devos M, Brochez L. IDO Expression in Cancer: Different Compartment, Different Functionality? Front Immunol 2020; 11:531491. [PMID: 33072086 PMCID: PMC7541907 DOI: 10.3389/fimmu.2020.531491] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022] Open
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) is a cytosolic haem-containing enzyme involved in the degradation of tryptophan to kynurenine. Although initially thought to be solely implicated in the modulation of innate immune responses during infection, subsequent discoveries demonstrated IDO1 as a mechanism of acquired immune tolerance. In cancer, IDO1 expression/activity has been observed in tumor cells as well as in the tumor-surrounding stroma, which is composed of endothelial cells, immune cells, fibroblasts, and mesenchymal cells. IDO1 expression/activity has also been reported in the peripheral blood. This manuscript reviews available data on IDO1 expression, mechanisms of its induction, and its function in cancer for each of these compartments. In-depth study of the biological function of IDO1 according to the expressing (tumor) cell can help to understand if and when IDO1 inhibition can play a role in cancer therapy.
Collapse
Affiliation(s)
- Annabel Meireson
- Department of Dermatology, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Michael Devos
- Department of Dermatology, Ghent University Hospital, Ghent, Belgium
| | - Lieve Brochez
- Department of Dermatology, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
17
|
Alobaid M, Richards SJ, Alexander M, Gibson M, Ghaemmaghami A. Developing immune-regulatory materials using immobilized monosaccharides with immune-instructive properties. Mater Today Bio 2020; 8:100080. [PMID: 33205040 PMCID: PMC7649522 DOI: 10.1016/j.mtbio.2020.100080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 11/15/2022] Open
Abstract
New strategies for immune modulation have shown real promise in regenerative medicine as well as the fight against autoimmune diseases, allergies, and cancer. Dendritic cells (DCs) are gatekeepers of the immune system and their ability in shaping the adaptive immune responses makes DCs ideal targets for immune modulation. Carbohydrates are abundant in different biological systems and are known to modulate DC phenotype and function. However, how simple monosaccharides instruct DC function is less well understood. In this study, we used a combinatorial array of immobilized monosaccharides to investigate how they modulate DC phenotype and function and crucially the impact of such changes on downstream adaptive immune responses. Our data show that a selection of monosaccharides significantly suppress lipopolysaccharide-induced DC activation as evidenced by a reduction in CD40 expression, IL-12 production, and indoleamine 2,3-dioxygenase activity, while inducing a significant increase in IL-10 production. These changes are indicative of the induction of an anti-inflammatory or regulatory phenotype in DCs, which was further confirmed in DC-T cell co-cultures where DCs cultured on the 'regulatory' monosaccharide-coated surfaces were shown to induce naïve T cell polarization toward regulatory phenotype. Our data also highlighted a selection of monosaccharides that are able to promote mixed Treg and Th17 cell differentiation, a T cell phenotype expected to be highly immune suppressive. These data show the potential immunomodulatory effects of immobilized monosaccharides in priming DCs and skewing T cell differentiation toward an immune-regulatory phenotype. The ability to fine-tune immune responses using these simple carbohydrate combinations (e.g. as coatings for existing materials) can be utilized as novel tools for immune modulation with potential applications in regenerative medicine, implantable medical devices, and wound healing where reduction of inflammatory responses and maintaining immune homeostasis are desirable.
Collapse
Key Words
- (Gal1), 100% 1-amino-1-deoxy-β-d-galactose
- (Gal1–Gal2), 50% 1-amino-1-deoxy-β-d-galactose + 50% 2-amino-2-deoxy-β-d-galactose
- (Gal2), 100% 2-amino-2-deoxy-β-d-galactose
- (Gal2–Man1), 90% 2-amino-2-deoxy-β-d-galactose + 10% 1-amino-1-deoxy-β-d-mannose
- (Gal2–Man2), 2-amino-2-deoxy-β-d-galactose + 10% 2-amino-2-deoxy-β-d-mannose
- (Man1–Man2), 40% 1-amino-1-deoxy-β-d-mannose + 60% 2-amino-2-deoxy-β-d-mannose
- CLR, C-type lectin receptor
- Carbohydrates
- DC-SIGN, Dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin
- DCs, Dendritic cells
- Dendritic cells
- FBS, Fetal bovine serum
- Fucose
- Galactose
- IDO, Indoleamine 2,3-dioxygenase
- Immune modulation
- Immune-instructive materials
- LPS, Lipopolysaccharide
- MFI, Median fluorescence intensity
- MR, Mannose receptor
- MT, 1-methyl-DL-tryptophan
- Mannose
- PRR, Pattern recognition receptor
- Polymers
- T cells
Collapse
Affiliation(s)
- M.A. Alobaid
- Immunology & Immuno-Bioengineering, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - S.-J. Richards
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - M.R. Alexander
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - M.I. Gibson
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
- Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - A.M. Ghaemmaghami
- Immunology & Immuno-Bioengineering, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90024, USA
| |
Collapse
|
18
|
Elmansi AM, Hussein KA, Herrero SM, Periyasamy-Thandavan S, Aguilar-Pérez A, Kondrikova G, Kondrikov D, Eisa NH, Pierce JL, Kaiser H, Ding KH, Walker AL, Jiang X, Bollag WB, Elsalanty M, Zhong Q, Shi XM, Su Y, Johnson M, Hunter M, Reitman C, Volkman BF, Hamrick MW, Isales CM, Fulzele S, McGee-Lawrence ME, Hill WD. Age-related increase of kynurenine enhances miR29b-1-5p to decrease both CXCL12 signaling and the epigenetic enzyme Hdac3 in bone marrow stromal cells. Bone Rep 2020; 12:100270. [PMID: 32395570 PMCID: PMC7210406 DOI: 10.1016/j.bonr.2020.100270] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022] Open
Abstract
Mechanisms leading to age-related reductions in bone formation and subsequent osteoporosis are still incompletely understood. We recently demonstrated that kynurenine (KYN), a tryptophan metabolite, accumulates in serum of aged mice and induces bone loss. Here, we report on novel mechanisms underlying KYN's detrimental effect on bone aging. We show that KYN is increased with aging in murine bone marrow mesenchymal stem cells (BMSCs). KYN reduces bone formation via modulating levels of CXCL12 and its receptors as well as histone deacetylase 3 (Hdac3). BMSCs responded to KYN by significantly decreasing mRNA expression levels of CXCL12 and its cognate receptors, CXCR4 and ACKR3, as well as downregulating osteogenic gene RUNX2 expression, resulting in a significant inhibition in BMSCs osteogenic differentiation. KYN's effects on these targets occur by increasing regulatory miRNAs that target osteogenesis, specifically miR29b-1-5p. Thus, KYN significantly upregulated the anti-osteogenic miRNA miR29b-1-5p in BMSCs, mimicking the up-regulation of miR-29b-1-5p in human and murine BMSCs with age. Direct inhibition of miR29b-1-5p by antagomirs rescued CXCL12 protein levels downregulated by KYN, while a miR29b-1-5p mimic further decreased CXCL12 levels. KYN also significantly downregulated mRNA levels of Hdac3, a target of miR-29b-1-5p, as well as its cofactor NCoR1. KYN is a ligand for the aryl hydrocarbon receptor (AhR). We hypothesized that AhR mediates KYN's effects in BMSCs. Indeed, AhR inhibitors (CH-223191 and 3',4'-dimethoxyflavone [DMF]) partially rescued secreted CXCL12 protein levels in BMSCs treated with KYN. Importantly, we found that treatment with CXCL12, or transfection with an miR29b-1-5p antagomir, downregulated the AhR mRNA level, while transfection with miR29b-1-5p mimic significantly upregulated its level. Further, CXCL12 treatment downregulated IDO, an enzyme responsible for generating KYN. Our findings reveal novel molecular pathways involved in KYN's age-associated effects in the bone microenvironment that may be useful translational targets for treating osteoporosis.
Collapse
Affiliation(s)
- Ahmed M Elmansi
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States of America
| | - Khaled A Hussein
- Department of Oral Surgery and Medicine, National Research Centre, Cairo, Egypt
| | | | | | - Alexandra Aguilar-Pérez
- Department of Anatomy and Cell Biology, Indiana University School of Medicine in Indianapolis, IN, United States of America.,Department of Cellular and Molecular Biology, School of Medicine, Universidad Central del Caribe, Bayamon 00956, Puerto Rico.,Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Galina Kondrikova
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States of America
| | - Dmitry Kondrikov
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States of America
| | - Nada H Eisa
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States of America.,Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Jessica L Pierce
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Helen Kaiser
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Ke-Hong Ding
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Aisha L Walker
- Department of Medicine, Vascular Medicine Institute, University of Pittsburg School of Medicine, Pittsburg, PA 15261, United States of America
| | - Xue Jiang
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wendy B Bollag
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America.,Department of Orthopaedic Surgery, Medical College of Georgia, Aueusta University, Augusta, GA 30912, United States of America.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States of America.,Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, United States of America.,Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Mohammed Elsalanty
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Qing Zhong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Xing-Ming Shi
- Department of Orthopaedic Surgery, Medical College of Georgia, Aueusta University, Augusta, GA 30912, United States of America.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Yun Su
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Maribeth Johnson
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America.,Department of Population Health Science, Augusta University, Augusta, GA 30912, United States of America
| | - Monte Hunter
- Department of Orthopaedic Surgery, Medical College of Georgia, Aueusta University, Augusta, GA 30912, United States of America
| | - Charles Reitman
- Orthopaedics and Physical Medicine Department, Medical University of South Carolina, Charleston, SC 29403, United States of America
| | - Brian F Volkman
- Biochemistry Department, Medical College of Wisconsin, Milwaukee, WI 53226, United States of America
| | - Mark W Hamrick
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America.,Department of Orthopaedic Surgery, Medical College of Georgia, Aueusta University, Augusta, GA 30912, United States of America.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States of America
| | - Carlos M Isales
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America.,Department of Orthopaedic Surgery, Medical College of Georgia, Aueusta University, Augusta, GA 30912, United States of America.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States of America.,Division of Endocrinology, Diabetes and Metabolism, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Sadanand Fulzele
- Department of Orthopaedic Surgery, Medical College of Georgia, Aueusta University, Augusta, GA 30912, United States of America.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States of America
| | - Meghan E McGee-Lawrence
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America.,Department of Orthopaedic Surgery, Medical College of Georgia, Aueusta University, Augusta, GA 30912, United States of America.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States of America
| | - William D Hill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States of America.,Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America.,Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States of America.,Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, United States of America
| |
Collapse
|
19
|
Dos Santos LM, Commodaro AG, Vasquez ARR, Kohlhoff M, de Paula Guerra DA, Coimbra RS, Martins-Filho OA, Teixeira-Carvalho A, Rizzo LV, Vieira LQ, Serra HM. Intestinal microbiota regulates tryptophan metabolism following oral infection with Toxoplasma gondii. Parasite Immunol 2020; 42:e12720. [PMID: 32275066 DOI: 10.1111/pim.12720] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 02/24/2020] [Accepted: 03/30/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The intestinal microbiota plays an important role in modulating host immune responses. Oral Toxoplasma gondii infection can promote intestinal inflammation in certain mice strains. The IDO-AhR axis may control tryptophan (Trp) metabolism constituting an important immune regulatory mechanism in inflammatory settings. AIMS In the present study, we investigated the role of the intestinal microbiota on Trp metabolism during oral infection with T gondii. METHODS AND RESULTS Mice were treated with antibiotics for four weeks and then infected with T gondii by gavage. Histopathology and immune responses were evaluated 8 days after infection. We found that depletion of intestinal microbiota by antibiotics contributed to resistance against T gondii infection and led to reduced expression of AhR on dendritic and Treg cells. Mice depleted of Gram-negative bacteria presented higher levels of systemic Trp, downregulation of AhR expression and increased resistance to infection whereas depletion of Gram-positive bacteria did not affect susceptibility or expression of AhR on immune cells. CONCLUSION Our findings indicate that the intestinal microbiota can control Trp availability and provide a link between the AhR pathway and host-microbiota interaction in acute infection with T gondii.
Collapse
Affiliation(s)
- Liliane M Dos Santos
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Alessandra G Commodaro
- Departmento de Oftalmologia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Alicia R R Vasquez
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Markus Kohlhoff
- Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ, Belo Horizonte, Brazil
| | | | - Roney S Coimbra
- Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ, Belo Horizonte, Brazil
| | | | | | - Luiz V Rizzo
- Instituto Israelita de Pesquisa e Ensino, São Paulo, Brazil
| | - Leda Q Vieira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Horacio M Serra
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| |
Collapse
|
20
|
Fedele V, Melisi D. Permissive State of EMT: The Role of Immune Cell Compartment. Front Oncol 2020; 10:587. [PMID: 32391271 PMCID: PMC7189417 DOI: 10.3389/fonc.2020.00587] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
The Epithelial to Mesenchymal Transition (EMT) type 3 is a reversible dynamic process recognized as a major determinant of the metastatic event, although many questions regarding its role throughout this process remain unanswered. The ability of cancer cells to migrate and colonize distant organs is a key aspect of tumor progression and evolution, requiring constant tumor cells and tumor microenvironment (TME) changes, as well as constant changes affecting the cross-talk between the two aforementioned compartments. Alterations affecting tumor cells, such as transcription factors, trans-membrane receptors, chromatin remodeling complexes and metabolic pathways, leading to the disappearance of the epithelial phenotype and concomitant gaining of the undifferentiated mesenchymal phenotype are undoubtedly major players of the EMT process. However, several lines of evidence point out toward a more critical role of TME composition in creating an “EMT-permissive state.” The “EMT-permissive state” consists in changes affecting physical and biochemical properties (i.e., stiffness and/or hypoxia) as well as changes of the TME cellular component (i.e., immune-cell, blood vessel, lymphatic vessels, fibroblasts, and fat cells) that favor and induce the epithelial mesenchymal transition. In this mini review, we will discuss the role of the tumor microenvironment cellular component that are involved in supporting the EMT, with particular emphasis on the immune-inflammatory cells component.
Collapse
Affiliation(s)
- Vita Fedele
- Digestive Molecular Clinical Oncology Research Unit, Section of Medical Oncology, Università degli Studi di Verona, Verona, Italy
| | - Davide Melisi
- Digestive Molecular Clinical Oncology Research Unit, Section of Medical Oncology, Università degli Studi di Verona, Verona, Italy
| |
Collapse
|
21
|
Basu A, Kodumudi K. Multimodal approaches to improve immunotherapy in breast cancer. Immunotherapy 2020; 12:161-165. [PMID: 32157934 DOI: 10.2217/imt-2019-0198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Amrita Basu
- Immunology Program & Department of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Krithika Kodumudi
- Immunology Program & Department of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA.,Department of Oncological Sciences, USF Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
22
|
Volarevic V, Markovic BS, Jankovic MG, Djokovic B, Jovicic N, Harrell CR, Fellabaum C, Djonov V, Arsenijevic N, Lukic ML. Galectin 3 protects from cisplatin-induced acute kidney injury by promoting TLR-2-dependent activation of IDO1/Kynurenine pathway in renal DCs. Theranostics 2019; 9:5976-6001. [PMID: 31534532 PMCID: PMC6735380 DOI: 10.7150/thno.33959] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 06/17/2019] [Indexed: 12/21/2022] Open
Abstract
Strategies targeting cross-talk between immunosuppressive renal dendritic cells (DCs) and T regulatory cells (Tregs) may be effective in treating cisplatin (CDDP)-induced acute kidney injury (AKI). Galectin 3 (Gal-3), expressed on renal DCs, is known as a crucial regulator of immune response in the kidneys. In this study, we investigated the role of Gal-3 for DCs-mediated expansion of Tregs in the attenuation of CDDP-induced AKI. Methods: AKI was induced in CDDP-treated wild type (WT) C57BL/6 and Gal-3 deficient (Gal-3-/-) mice. Biochemical, histological analysis, enzyme-linked immunosorbent assay (ELISA), immunohistochemistry, real-time PCR, magnetic cell sorting, flow cytometry and intracellular staining of renal-infiltrated immune cells were used to determine the differences between CDDP-treated WT and Gal-3-/- mice. Newly synthesized selective inhibitor of Gal-3 (Davanat) was used for pharmacological inhibition of Gal-3. Recombinant Gal-3 was used to demonstrate the effects of exogenously administered soluble Gal-3 on AKI progression. Pam3CSK4 was used for activation of Toll-like receptor (TLR)-2 in DCs. Cyclophosphamide or anti-CD25 antibody were used for the depletion of Tregs. 1-Methyl Tryptophan (1-MT) was used for pharmacological inhibition of Indoleamine 2,3-dioxygenase-1 (IDO1) in TLR-2-primed DCs which were afterwards used in passive transfer experiments. Results: CDDP-induced nephrotoxicity was significantly more aggravated in Gal-3-/- mice. Significantly reduced number of immunosuppressive TLR-2 and IDO1-expressing renal DCs, lower serum levels of KYN, decreased presence of IL-10-producing Tregs and significantly higher number of inflammatory IFN-γ and IL-17-producing neutrophils, Th1 and Th17 cells were observed in the CDDP-injured kidneys of Gal-3-/- mice. Pharmacological inhibitor of Gal-3 aggravated CDDP-induced AKI in WT animals while recombinant Gal-3 attenuated renal injury and inflammation in CDDP-treated Gal-3-/- mice. CDDP-induced apoptosis, driven by Bax and caspase-3, was aggravated in Gal-3-/- animals and in WT mice that received Gal-3 inhibitor (CDDP+Davanat-treated mice). Recombinant Gal-3 managed to completely attenuate CDDP-induced apoptosis in CDDP-injured kidneys of Gal-3-/- mice. Genetic deletion as well as pharmacological inhibition of Gal-3 in renal DCs remarkably reduced TLR-2-dependent activation of IDO1/KYN pathway in these cells diminishing their capacity to prevent transdifferentiation of Tregs in inflammatory Th1 and Th17 cells. Additionally, Tregs generated by Gal-3 deficient DCs were not able to suppress production of IFN-γ and IL-17 in activated neutrophils. TLR-2-primed DCs significantly enhanced capacity of Tregs for attenuation of CDDP-induced AKI and inflammation and expression of Gal-3 on TLR-2-primed DCs was crucially important for their capacity to enhance nephroprotective and immunosuppressive properties of Tregs. Adoptive transfer of TLR-2-primed WTDCs significantly expanded Tregs in the kidneys of CDDP-treated WT and Gal-3-/- recipients resulting in the suppression of IFN-γ and IL-17-driven inflammation and alleviation of AKI. Importantly, this phenomenon was not observed in CDDP-treated WT and Gal-3-/- recipients of TLR-2-primed Gal-3-/-DCs. Gal-3-dependent nephroprotective and immunosuppressive effects of renal DCs was due to the IDO1-induced expansion of renal Tregs since either inhibition of IDO1 activity in TLR-2-primed DCs or depletion of Tregs completely diminished DCs-mediated attenuation of CDDP-induced AKI. Conclusions: Gal-3 protects from CDDP-induced AKI by promoting TLR-2-dependent activation of IDO1/KYN pathway in renal DCs resulting in increased expansion of immunosuppressive Tregs in injured kidneys. Activation of Gal-3:TLR-2:IDO1 pathway in renal DCs should be further explored as new therapeutic approach for DC-based immunosuppression of inflammatory renal diseases.
Collapse
|
23
|
Giunta EF, Argenziano G, Brancaccio G, Martinelli E, Ciardiello F, Troiani T. Beyond PD-1/PD-L1 Axis Blockade: New Combination Strategies in Metastatic Melanoma Treatment. CURRENT CANCER THERAPY REVIEWS 2019. [DOI: 10.2174/1573394714666180927095650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
:
Metastatic melanoma treatment has dramatically changed in the last few years, having a
breakthrough with the introduction of targeted agents and immunotherapy. PD-1/PD-L1 pathway
is one of the physiologic mechanisms of peripheral immune tolerance, but it also represents a
mechanism of tumor immune escape. PD-1/PD-L1 inhibitors represent new immune-checkpoint
drugs currently used in metastatic melanoma treatment.
:
Resistance to PD-1/PD-L1 axis blockade, which is the main cause of therapeutic failure during
therapeutic use of these drugs, could be linked to several mechanism of immune escape. In fact,
other inhibitory receptor such as CTLA-4, LAG-3, TIM-3 and TIGIT might be co-expressed on T
cells, deleting the effect of anti-PD-1/PD-L1; overexpression of the enzyme IDO could cause immunosuppression
through the depletion of tryptophan in the tumor microenvironment; defective c
ostimulation (through reduced activity of 4-1BB and OX40 receptors) could result in T-cell
energy.
:
Combination of anti-PD-1/PD-L1 with drugs targeting inhibitory or costimulatory receptors, intracellular
pathways, enzymes or neoangiogenesis could be a possible strategy to overcome resistance
to single PD-1/PD-L1 blockade. Clinical trials evaluating combination therapies have already
showed interesting results, although most of them are still on going.
Collapse
Affiliation(s)
| | - Giuseppe Argenziano
- Dermatology unit, Università degli Studi della Campania Luigi Vanvitelli, via Pansini 5, 80131 Naples, Italy
| | - Gabriella Brancaccio
- Dermatology unit, Università degli Studi della Campania Luigi Vanvitelli, via Pansini 5, 80131 Naples, Italy
| | - Erika Martinelli
- Oncologia Medica, Dipartimento di Internistica Clinica e Sperimentale , Italy
| | | | - Teresa Troiani
- Oncologia Medica, Dipartimento di Internistica Clinica e Sperimentale , Italy
| |
Collapse
|
24
|
Orlova EG, Shirshev SV, Loginova OA. Mechanisms of Leptin and Ghrelin Action on Maturation and Functions of Dendritic Cells. BIOCHEMISTRY (MOSCOW) 2019; 84:1-10. [PMID: 30927520 DOI: 10.1134/s0006297919010012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Molecular mechanisms of the immunomodulatory effects of leptin and ghrelin in concentrations typical for pregnancy on the maturation and functional activity of dendritic cells (DCs) generated from the peripheral blood monocytes of women are investigated. The presence of leptin during DC maturation did not affect the levels of CD83+CD1c+, CD86+CD1c+, and HLA-DR+CD1c+ DCs, but increased the amount and the activity of the enzyme indoleamine 2,3-dioxygenase (IDO). Cell culturing in the presence of ghrelin or combination of leptin and ghrelin reduced the percentage of CD86+CD1c+ DCs but did not affect the levels of CD83+CD1c+ and HLA-DR+CD1c+ DCs. In addition, ghrelin reduced the number of IDO molecules without affecting its activity. Simultaneous presence of leptin and ghrelin increased induced IDO activity without affecting the amount of the enzyme in DCs. The effects of leptin and ghrelin on the investigated functions of DCs in some cases correlated with high levels of cAMP. New mechanisms for leptin and ghrelin regulation of tolerogenic functions of DCs in pregnancy are proposed.
Collapse
Affiliation(s)
- E G Orlova
- Perm Federal Research Center, Institute of Ecology and Genetics of Microorganisms, Ural Branch of the Russian Academy of Sciences, Perm, 614081, Russia.
| | - S V Shirshev
- Perm Federal Research Center, Institute of Ecology and Genetics of Microorganisms, Ural Branch of the Russian Academy of Sciences, Perm, 614081, Russia
| | - O A Loginova
- Perm Federal Research Center, Institute of Ecology and Genetics of Microorganisms, Ural Branch of the Russian Academy of Sciences, Perm, 614081, Russia
| |
Collapse
|
25
|
Domblides C, Lartigue L, Faustin B. Control of the Antitumor Immune Response by Cancer Metabolism. Cells 2019; 8:cells8020104. [PMID: 30708988 PMCID: PMC6406288 DOI: 10.3390/cells8020104] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 12/12/2022] Open
Abstract
The metabolic reprogramming of tumor cells and immune escape are two major hallmarks of cancer cells. The metabolic changes that occur during tumorigenesis, enabling survival and proliferation, are described for both solid and hematological malignancies. Concurrently, tumor cells have deployed mechanisms to escape immune cell recognition and destruction. Additionally, therapeutic blocking of tumor-mediated immunosuppression has proven to have an unprecedented positive impact in clinical oncology. Increased evidence suggests that cancer metabolism not only plays a crucial role in cancer signaling for sustaining tumorigenesis and survival, but also has wider implications in the regulation of antitumor immune signaling through both the release of signaling molecules and the expression of immune membrane ligands. Here, we review these molecular events to highlight the contribution of cancer cell metabolic reprogramming on the shaping of the antitumor immune response.
Collapse
Affiliation(s)
- Charlotte Domblides
- Bordeaux University, CNRS, UMR 5164, ImmunoConcEpT, 33000 Bordeaux, France.
- Department of Medical Oncology, Hôpital Saint-André, Bordeaux University Hospital-CHU, 33000 Bordeaux, France.
| | - Lydia Lartigue
- Curematch, Inc., 6440 Lusk Bvld, San Diego, CA 92121, USA.
| | - Benjamin Faustin
- Bordeaux University, CNRS, UMR 5164, ImmunoConcEpT, 33000 Bordeaux, France.
- Cellomet, CGFB, 146 Rue léo Saignat, F-33000 Bordeaux, France.
| |
Collapse
|
26
|
Biomarqueurs prédictifs de réponse aux inhibiteurs de points de contrôle immuns. Bull Cancer 2018; 105 Suppl 1:S80-S91. [DOI: 10.1016/s0007-4551(18)30393-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 10/23/2017] [Indexed: 11/21/2022]
|
27
|
Miyazaki A, Kandasamy S, Michael H, Langel SN, Paim FC, Chepngeno J, Alhamo MA, Fischer DD, Huang HC, Srivastava V, Kathayat D, Deblais L, Rajashekara G, Saif LJ, Vlasova AN. Protein deficiency reduces efficacy of oral attenuated human rotavirus vaccine in a human infant fecal microbiota transplanted gnotobiotic pig model. Vaccine 2018; 36:6270-6281. [PMID: 30219368 PMCID: PMC6180620 DOI: 10.1016/j.vaccine.2018.09.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/31/2018] [Accepted: 09/05/2018] [Indexed: 12/11/2022]
Abstract
Protein deficiency impacted immunity and reduced human RV vaccine efficacy. Human infant fecal microbiota exacerbated the negative effects of protein deficiency. Immunological dysfunction could have been induced by altered tryptophan catabolism. Our findings provide an explanation for RV vaccine failures in malnourished children.
Background Low efficacy of rotavirus (RV) vaccines in developing African and Asian countries, where malnutrition is prevalent, remains a major concern and a challenge for global health. Methods To understand the effects of protein malnutrition on RV vaccine efficacy, we elucidated the innate, T cell and cytokine immune responses to attenuated human RV (AttHRV) vaccine and virulent human RV (VirHRV) challenge in germ-free (GF) pigs or human infant fecal microbiota (HIFM) transplanted gnotobiotic (Gn) pigs fed protein-deficient or -sufficient bovine milk diets. We also analyzed serum levels of tryptophan (TRP), a predictor of malnutrition, and kynurenine (KYN). Results Protein-deficient pigs vaccinated with oral AttHRV vaccine had lower protection rates against diarrhea post-VirHRV challenge and significantly increased fecal virus shedding titers (HIFM transplanted but not GF pigs) compared with their protein-sufficient counterparts. Reduced vaccine efficacy in protein-deficient pigs coincided with altered serum IFN-α, TNF-α, IL-12 and IFN-γ responses to oral AttHRV vaccine and the suppression of multiple innate immune parameters and HRV-specific IFN-γ producing T cells post-challenge. In protein-deficient HIFM transplanted pigs, decreased serum KYN, but not TRP levels were observed throughout the experiment, suggesting an association between the altered TRP metabolism and immune responses. Conclusion Collectively, our findings confirm the negative effects of protein deficiency, which were exacerbated in the HIFM transplanted pigs, on innate, T cell and cytokine immune responses to HRV and on vaccine efficacy, as well as on TRP-KYN metabolism.
Collapse
Affiliation(s)
- Ayako Miyazaki
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA; Division of Viral Disease and Epidemiology, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Ibaraki 305-0856, Japan
| | - Sukumar Kandasamy
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Husheem Michael
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Stephanie N Langel
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Francine C Paim
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Juliet Chepngeno
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Moyasar A Alhamo
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - David D Fischer
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA; Division of Integrated Biomedical Sciences, School of Dentistry, University of Detroit Mercy, Detroit, MI, USA(1)
| | - Huang-Chi Huang
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Vishal Srivastava
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Dipak Kathayat
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Loic Deblais
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Gireesh Rajashekara
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Linda J Saif
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA.
| | - Anastasia N Vlasova
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA.
| |
Collapse
|
28
|
Frelau A, Pracht M, Le Sourd S, Lespagnol A, Corre R, Ménard C, Tarte K, Mosser J, Edeline J. WITHDRAWN: Biomarqueurs prédictifs de réponse aux inhibiteurs de points de contrôle immuns. Bull Cancer 2018:S0007-4551(17)30384-3. [PMID: 29525053 DOI: 10.1016/j.bulcan.2017.10.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 10/19/2017] [Accepted: 10/23/2017] [Indexed: 11/19/2022]
Affiliation(s)
- Alexandra Frelau
- CLCC Eugène-Marquis UNICANCER, oncologie médicale, avenue de la Bataille-Flandres-Dunkerque, 35042 Rennes cedex, France.
| | - Marc Pracht
- CLCC Eugène-Marquis UNICANCER, oncologie médicale, avenue de la Bataille-Flandres-Dunkerque, 35042 Rennes cedex, France
| | - Samuel Le Sourd
- CLCC Eugène-Marquis UNICANCER, oncologie médicale, avenue de la Bataille-Flandres-Dunkerque, 35042 Rennes cedex, France
| | - Alexandra Lespagnol
- CHU de Rennes, laboratoire de génétique somatique des cancers, 2, rue Henri-Le-Guilloux, 35033 Rennes cedex 9, France
| | - Romain Corre
- CHU de Rennes, service de pneumologie, 2, rue Henri-Le-Guilloux, 35033 Rennes cedex 9, France
| | - Cédric Ménard
- CHU de Rennes, Inserm U197, laboratoire d'immunologie, thérapie cellulaire et hématopoïèse, 2, rue Henri-Le-Guilloux, 35033 Rennes cedex 9, France
| | - Karin Tarte
- CHU de Rennes, Inserm U197, laboratoire d'immunologie, thérapie cellulaire et hématopoïèse, 2, rue Henri-Le-Guilloux, 35033 Rennes cedex 9, France
| | - Jean Mosser
- CHU de Rennes, laboratoire de génétique somatique des cancers, 2, rue Henri-Le-Guilloux, 35033 Rennes cedex 9, France; CHU de Rennes, institut de génétique et développement, CNRS-UR1, IGRD UMR 6290, 2, rue Henri-Le-Guilloux, 35033 Rennes cedex 9, France
| | - Julien Edeline
- CLCC Eugène-Marquis UNICANCER, oncologie médicale, avenue de la Bataille-Flandres-Dunkerque, 35042 Rennes cedex, France; CHU de Rennes, UMR991, unité de recherche foie, métabolisme et cancer, 2, rue Henri-Le-Guilloux, 35033 Rennes cedex 9, France
| |
Collapse
|
29
|
Azeredo R, Serra CR, Oliva-Teles A, Costas B. Amino acids as modulators of the European seabass, Dicentrarchus labrax, innate immune response: an in vitro approach. Sci Rep 2017; 7:18009. [PMID: 29269876 PMCID: PMC5740149 DOI: 10.1038/s41598-017-18345-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 12/11/2017] [Indexed: 02/07/2023] Open
Abstract
Teleost innate immune system is a most developed and powerful system in which fish highly rely throughout their lives. Conditions in aquaculture farms are particularly prone to disease, thus, health and welfare ensuring strategies are an urgent call to which nutrition is gradually becoming a most regarded achievement tool. This study intended to evaluate different amino acids' effect on immune-related mechanisms as well as their potential as enhancers of European seabass, Dicentrarchus labrax, leucocyte functioning. To achieve these goals, primary cultures of head-kidney leucocytes were established and kept in amino acid (glutamine, arginine, tryptophan or methionine) supplemented culture media in two doses. The effects of amino acids treatments were then evaluated after stimulation with either Vibrio anguillarum or Vibrio anguillarum lipopolysaccharides by measuring nitric oxide production, extracellular respiratory burst, ATP and arginase activities, and expression of immune-related genes. Glutamine, arginine and tryptophan showed to be particularly relevant regarding cell energy dynamics; arginine and tryptophan supplementation also resulted in down-regulation of important immune-related genes. Immune responses in cells treated with methionine were generally enhanced but further studies, particularly those of enzymes activity, are essential to complement gene expression results and to better understand this nutrient's immune role in fish.
Collapse
Affiliation(s)
- Rita Azeredo
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos s/n, 4450-208, Matosinhos, Portugal.
- Departamento de Biologia, Faculdade de Ciências da Universidade do Porto (FCUP), Rua do Campo Alegre s/n, Ed. FC4, 4169-007, Porto, Portugal.
| | - Cláudia R Serra
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos s/n, 4450-208, Matosinhos, Portugal
| | - Aires Oliva-Teles
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos s/n, 4450-208, Matosinhos, Portugal
- Departamento de Biologia, Faculdade de Ciências da Universidade do Porto (FCUP), Rua do Campo Alegre s/n, Ed. FC4, 4169-007, Porto, Portugal
| | - Benjamín Costas
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos s/n, 4450-208, Matosinhos, Portugal.
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (ICBAS-UP), Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
| |
Collapse
|
30
|
Mesenchymal traits at the convergence of tumor-intrinsic and -extrinsic mechanisms of resistance to immune checkpoint blockers. Emerg Top Life Sci 2017; 1:471-486. [PMID: 33525801 PMCID: PMC7289012 DOI: 10.1042/etls20170068] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/08/2017] [Accepted: 11/09/2017] [Indexed: 01/01/2023]
Abstract
Targeting of immune checkpoint blockers (ICBs), such as cytotoxic T-lymphocyte antigen-4 and programmed-death 1/programmed-death ligand 1, has dramatically changed the landscape of cancer treatment. Seeing patients who were refractory to conventional therapy recover after immunotherapy, with high rates of objective durable responses and increased overall survival, has raised great enthusiasm in cancer care and research. However, to date, only a restricted portion of patients benefit from these therapies, due to natural and acquired resistance relying on the ever-evolving cross-talk between tumor and stromal cells. Here, we review the convergence of tumor-intrinsic and -extrinsic cues, both affecting tumor plasticity and tumor stroma leading to an immunosuppressive tumor microenvironment, which may account for the heterogeneous responses and resistance to ICB therapies. A deeper knowledge of the mechanisms and fingerprints involved in natural and acquired resistance is likely to bring clinical benefit to the majority of patients, offering important clues for overcoming drug resistance and boosting the effectiveness of treatment. We discuss the need to define tumor subtypes based on the tumor, immune and stromal gene signature and propose that the better we understand tumor mesenchymal traits, the more we will be able to identify predictive biomarkers of response to ICB treatments.
Collapse
|
31
|
Sayour EJ, Mitchell DA. Immunotherapy for Pediatric Brain Tumors. Brain Sci 2017; 7:brainsci7100137. [PMID: 29065490 PMCID: PMC5664064 DOI: 10.3390/brainsci7100137] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 09/20/2017] [Accepted: 10/18/2017] [Indexed: 12/20/2022] Open
Abstract
Malignant brain tumors are the most common cause of solid cancer death in children. New targeted therapies are vital to improve treatment outcomes, but must be developed to enable trafficking across the blood brain barrier (BBB). Since activated T cells cross the BBB, cancer immunotherapy can be harnessed to unlock the cytotoxic potential of the immune system. However, standard of care treatments (i.e., chemotherapy and radiation) applied concomitant to pediatric brain tumor immunotherapy may abrogate induction of immunotherapeutic responses. This review will discuss the development of immunotherapies within this paradigm using emerging approaches being investigated in phase I/II trials in children with refractory brain tumors, including checkpoint inhibitors, vaccine immunotherapy, and adoptive cell therapy.
Collapse
Affiliation(s)
- Elias J Sayour
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lilian S. Wells Department of Neurosurgery, 1149 South Newell Drive, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA.
| | - Duane A Mitchell
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lilian S. Wells Department of Neurosurgery, 1149 South Newell Drive, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
32
|
Sadok I, Gamian A, Staniszewska MM. Chromatographic analysis of tryptophan metabolites. J Sep Sci 2017; 40:3020-3045. [PMID: 28590049 PMCID: PMC5575536 DOI: 10.1002/jssc.201700184] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/15/2017] [Accepted: 05/24/2017] [Indexed: 12/14/2022]
Abstract
The kynurenine pathway generates multiple tryptophan metabolites called collectively kynurenines and leads to formation of the enzyme cofactor nicotinamide adenine dinucleotide. The first step in this pathway is tryptophan degradation, initiated by the rate-limiting enzymes indoleamine 2,3-dioxygenase, or tryptophan 2,3-dioxygenase, depending on the tissue. The balanced kynurenine metabolism, which has been a subject of multiple studies in last decades, plays an important role in several physiological and pathological conditions such as infections, autoimmunity, neurological disorders, cancer, cataracts, as well as pregnancy. Understanding the regulation of tryptophan depletion provide novel diagnostic and treatment opportunities, however it requires reliable methods for quantification of kynurenines in biological samples with complex composition (body fluids, tissues, or cells). Trace concentrations, interference of sample components, and instability of some tryptophan metabolites need to be addressed using analytical methods. The novel separation approaches and optimized extraction protocols help to overcome difficulties in analyzing kynurenines within the complex tissue material. Recent developments in chromatography coupled with mass spectrometry provide new opportunity for quantification of tryptophan and its degradation products in various biological samples. In this review, we present current accomplishments in the chromatographic methodologies proposed for detection of tryptophan metabolites and provide a guide for choosing the optimal approach.
Collapse
Affiliation(s)
- Ilona Sadok
- Laboratory of Separation and Spectroscopic Method Applications, Centre for Interdisciplinary ResearchThe John Paul II Catholic University of LublinLublinPoland
| | - Andrzej Gamian
- Laboratory of Medical MicrobiologyHirszfeld Institute of Immunology and Experimental TherapyPolish Academy of SciencesWroclawPoland
- Department of Medical BiochemistryWroclaw Medical UniversityWroclawPoland
| | - Magdalena Maria Staniszewska
- Laboratory of Separation and Spectroscopic Method Applications, Centre for Interdisciplinary ResearchThe John Paul II Catholic University of LublinLublinPoland
- Laboratory of Medical MicrobiologyHirszfeld Institute of Immunology and Experimental TherapyPolish Academy of SciencesWroclawPoland
| |
Collapse
|
33
|
Central Nervous System Infection with Borna Disease Virus Causes Kynurenine Pathway Dysregulation and Neurotoxic Quinolinic Acid Production. J Virol 2017; 91:JVI.00673-17. [PMID: 28446679 PMCID: PMC5487560 DOI: 10.1128/jvi.00673-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 12/27/2022] Open
Abstract
Central nervous system infection of neonatal and adult rats with Borna disease virus (BDV) results in neuronal destruction and behavioral abnormalities with differential immune-mediated involvement. Neuroactive metabolites generated from the kynurenine pathway of tryptophan degradation have been implicated in several human neurodegenerative disorders. Here, we report that brain expression of key enzymes in the kynurenine pathway are significantly, but differentially, altered in neonatal and adult rats with BDV infection. Gene expression analysis of rat brains following neonatal infection showed increased expression of kynurenine amino transferase II (KATII) and kynurenine-3-monooxygenase (KMO) enzymes. Additionally, indoleamine 2,3-dioxygenase (IDO) expression was only modestly increased in a brain region- and time-dependent manner in neonatally infected rats; however, its expression was highly increased in adult infected rats. The most dramatic impact on gene expression was seen for KMO, whose activity promotes the production of neurotoxic quinolinic acid. KMO expression was persistently elevated in brain regions of both newborn and adult BDV-infected rats, with increases reaching up to 86-fold. KMO protein levels were increased in neonatally infected rats and colocalized with neurons, the primary target cells of BDV infection. Furthermore, quinolinic acid was elevated in neonatally infected rat brains. We further demonstrate increased expression of KATII and KMO, but not IDO, in vitro in BDV-infected C6 astroglioma cells. Our results suggest that BDV directly impacts the kynurenine pathway, an effect that may be exacerbated by inflammatory responses in immunocompetent hosts. Thus, experimental models of BDV infection may provide new tools for discriminating virus-mediated from immune-mediated impacts on the kynurenine pathway and their relative contribution to neurodegeneration.IMPORTANCE BDV causes persistent, noncytopathic infection in vitro yet still elicits widespread neurodegeneration of infected neurons in both immunoincompetent and immunocompetent hosts. Here, we show that BDV infection induces expression of key enzymes of the kynurenine pathway in brains of newborn and adult infected rats and cultured astroglioma cells, shunting tryptophan degradation toward the production of neurotoxic quinolinic acid. Thus, our findings newly implicate this metabolic pathway in BDV-induced neurodegeneration. Given the importance of the kynurenine pathway in a wide range of human infections and neurodegenerative and neuropsychiatric disorders, animal models of BDV infection may serve as important tools for contrasting direct viral and indirect antiviral immune-mediated impacts on kynurenine pathway dysregulation and the ensuing neurodevelopmental and neuropathological consequences.
Collapse
|
34
|
Rodriguez Cetina Biefer H, Vasudevan A, Elkhal A. Aspects of Tryptophan and Nicotinamide Adenine Dinucleotide in Immunity: A New Twist in an Old Tale. Int J Tryptophan Res 2017; 10:1178646917713491. [PMID: 28659716 PMCID: PMC5476425 DOI: 10.1177/1178646917713491] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/07/2017] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence underscores the interesting ability of tryptophan to regulate immune responses. However, the exact mechanisms of tryptophan's immune regulation remain to be determined. Tryptophan catabolism via the kynurenine pathway is known to play an important role in tryptophan's involvement in immune responses. Interestingly, quinolinic acid, which is a neurotoxic catabolite of the kynurenine pathway, is the major pathway for the de novo synthesis of nicotinamide adenine dinucleotide (NAD+). Recent studies have shown that NAD+, a natural coenzyme found in all living cells, regulates immune responses and creates homeostasis via a novel signaling pathway. More importantly, the immunoregulatory properties of NAD+ are strongly related to the overexpression of tryptophan hydroxylase 1 (Tph1). This review provides recent knowledge of tryptophan and NAD+ and their specific and intriguing roles in the immune system. Furthermore, it focuses on the mechanisms by which tryptophan regulates NAD+ synthesis as well as innate and adaptive immune responses.
Collapse
Affiliation(s)
| | - Anju Vasudevan
- Angiogenesis and Brain Development Laboratory, Division of Basic Neuroscience, McLean Hospital and Harvard Medical School, Belmont, MA, USA
| | - Abdallah Elkhal
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
35
|
Tanhapour M, Vaisi-Raygani A, Khazaei M, Rahimi Z, Pourmotabbed T. Cytotoxic T-lymphocyte Associated Antigen-4 (CTLA-4) Polymorphism, Cancer, and Autoimmune Diseases. AIMS MEDICAL SCIENCE 2017. [DOI: 10.3934/medsci.2017.4.395] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
36
|
Novikov O, Wang Z, Stanford EA, Parks AJ, Ramirez-Cardenas A, Landesman E, Laklouk I, Sarita-Reyes C, Gusenleitner D, Li A, Monti S, Manteiga S, Lee K, Sherr DH. An Aryl Hydrocarbon Receptor-Mediated Amplification Loop That Enforces Cell Migration in ER-/PR-/Her2- Human Breast Cancer Cells. Mol Pharmacol 2016; 90:674-688. [PMID: 27573671 PMCID: PMC5074452 DOI: 10.1124/mol.116.105361] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 08/24/2016] [Indexed: 12/18/2022] Open
Abstract
The endogenous ligand-activated aryl hydrocarbon receptor (AHR) plays an important role in numerous biologic processes. As the known number of AHR-mediated processes grows, so too does the importance of determining what endogenous AHR ligands are produced, how their production is regulated, and what biologic consequences ensue. Consequently, our studies were designed primarily to determine whether ER-/PR-/Her2- breast cancer cells have the potential to produce endogenous AHR ligands and, if so, how production of these ligands is controlled. We postulated that: 1) malignant cells produce tryptophan-derived AHR ligand(s) through the kynurenine pathway; 2) these metabolites have the potential to drive AHR-dependent breast cancer migration; 3) the AHR controls expression of a rate-limiting kynurenine pathway enzyme(s) in a closed amplification loop; and 4) environmental AHR ligands mimic the effects of endogenous ligands. Data presented in this work indicate that primary human breast cancers, and their metastases, express high levels of AHR and tryptophan-2,3-dioxygenase (TDO); representative ER-/PR-/Her2- cell lines express TDO and produce sufficient intracellular kynurenine and xanthurenic acid concentrations to chronically activate the AHR. TDO overexpression, or excess kynurenine or xanthurenic acid, accelerates migration in an AHR-dependent fashion. Environmental AHR ligands 2,3,7,8-tetrachlorodibenzo[p]dioxin and benzo[a]pyrene mimic this effect. AHR knockdown or inhibition significantly reduces TDO2 expression. These studies identify, for the first time, a positive amplification loop in which AHR-dependent TDO2 expression contributes to endogenous AHR ligand production. The net biologic effect of AHR activation by endogenous ligands, which can be mimicked by environmental ligands, is an increase in tumor cell migration, a measure of tumor aggressiveness.
Collapse
MESH Headings
- Cell Line, Tumor
- Cell Movement
- Female
- Gene Amplification
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
- Humans
- Immunohistochemistry
- Kynurenine/metabolism
- Ligands
- Models, Biological
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, ErbB-2/metabolism
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/pathology
- Tryptophan/metabolism
- Tryptophan Oxygenase/genetics
- Tryptophan Oxygenase/metabolism
- Xanthurenates/metabolism
Collapse
Affiliation(s)
- Olga Novikov
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts (O.N., Z.W., E.A.S., A.J.P., A.R.-C., D.H.S.); Boston University Molecular and Translational Medicine Program, Boston, Massachusetts (O.N., E.A.S.); Department of Medicine, Division of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts (D.G., A.L., S.Mo.); Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts (E.L., I.L., C.S.-R.); and Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts (S.Ma., K.L.)
| | - Zhongyan Wang
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts (O.N., Z.W., E.A.S., A.J.P., A.R.-C., D.H.S.); Boston University Molecular and Translational Medicine Program, Boston, Massachusetts (O.N., E.A.S.); Department of Medicine, Division of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts (D.G., A.L., S.Mo.); Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts (E.L., I.L., C.S.-R.); and Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts (S.Ma., K.L.)
| | - Elizabeth A Stanford
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts (O.N., Z.W., E.A.S., A.J.P., A.R.-C., D.H.S.); Boston University Molecular and Translational Medicine Program, Boston, Massachusetts (O.N., E.A.S.); Department of Medicine, Division of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts (D.G., A.L., S.Mo.); Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts (E.L., I.L., C.S.-R.); and Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts (S.Ma., K.L.)
| | - Ashley J Parks
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts (O.N., Z.W., E.A.S., A.J.P., A.R.-C., D.H.S.); Boston University Molecular and Translational Medicine Program, Boston, Massachusetts (O.N., E.A.S.); Department of Medicine, Division of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts (D.G., A.L., S.Mo.); Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts (E.L., I.L., C.S.-R.); and Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts (S.Ma., K.L.)
| | - Alejandra Ramirez-Cardenas
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts (O.N., Z.W., E.A.S., A.J.P., A.R.-C., D.H.S.); Boston University Molecular and Translational Medicine Program, Boston, Massachusetts (O.N., E.A.S.); Department of Medicine, Division of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts (D.G., A.L., S.Mo.); Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts (E.L., I.L., C.S.-R.); and Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts (S.Ma., K.L.)
| | - Esther Landesman
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts (O.N., Z.W., E.A.S., A.J.P., A.R.-C., D.H.S.); Boston University Molecular and Translational Medicine Program, Boston, Massachusetts (O.N., E.A.S.); Department of Medicine, Division of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts (D.G., A.L., S.Mo.); Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts (E.L., I.L., C.S.-R.); and Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts (S.Ma., K.L.)
| | - Israa Laklouk
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts (O.N., Z.W., E.A.S., A.J.P., A.R.-C., D.H.S.); Boston University Molecular and Translational Medicine Program, Boston, Massachusetts (O.N., E.A.S.); Department of Medicine, Division of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts (D.G., A.L., S.Mo.); Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts (E.L., I.L., C.S.-R.); and Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts (S.Ma., K.L.)
| | - Carmen Sarita-Reyes
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts (O.N., Z.W., E.A.S., A.J.P., A.R.-C., D.H.S.); Boston University Molecular and Translational Medicine Program, Boston, Massachusetts (O.N., E.A.S.); Department of Medicine, Division of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts (D.G., A.L., S.Mo.); Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts (E.L., I.L., C.S.-R.); and Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts (S.Ma., K.L.)
| | - Daniel Gusenleitner
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts (O.N., Z.W., E.A.S., A.J.P., A.R.-C., D.H.S.); Boston University Molecular and Translational Medicine Program, Boston, Massachusetts (O.N., E.A.S.); Department of Medicine, Division of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts (D.G., A.L., S.Mo.); Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts (E.L., I.L., C.S.-R.); and Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts (S.Ma., K.L.)
| | - Amy Li
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts (O.N., Z.W., E.A.S., A.J.P., A.R.-C., D.H.S.); Boston University Molecular and Translational Medicine Program, Boston, Massachusetts (O.N., E.A.S.); Department of Medicine, Division of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts (D.G., A.L., S.Mo.); Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts (E.L., I.L., C.S.-R.); and Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts (S.Ma., K.L.)
| | - Stefano Monti
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts (O.N., Z.W., E.A.S., A.J.P., A.R.-C., D.H.S.); Boston University Molecular and Translational Medicine Program, Boston, Massachusetts (O.N., E.A.S.); Department of Medicine, Division of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts (D.G., A.L., S.Mo.); Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts (E.L., I.L., C.S.-R.); and Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts (S.Ma., K.L.)
| | - Sara Manteiga
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts (O.N., Z.W., E.A.S., A.J.P., A.R.-C., D.H.S.); Boston University Molecular and Translational Medicine Program, Boston, Massachusetts (O.N., E.A.S.); Department of Medicine, Division of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts (D.G., A.L., S.Mo.); Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts (E.L., I.L., C.S.-R.); and Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts (S.Ma., K.L.)
| | - Kyongbum Lee
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts (O.N., Z.W., E.A.S., A.J.P., A.R.-C., D.H.S.); Boston University Molecular and Translational Medicine Program, Boston, Massachusetts (O.N., E.A.S.); Department of Medicine, Division of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts (D.G., A.L., S.Mo.); Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts (E.L., I.L., C.S.-R.); and Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts (S.Ma., K.L.)
| | - David H Sherr
- Department of Environmental Health, Boston University School of Public Health, Boston, Massachusetts (O.N., Z.W., E.A.S., A.J.P., A.R.-C., D.H.S.); Boston University Molecular and Translational Medicine Program, Boston, Massachusetts (O.N., E.A.S.); Department of Medicine, Division of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts (D.G., A.L., S.Mo.); Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts (E.L., I.L., C.S.-R.); and Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts (S.Ma., K.L.)
| |
Collapse
|
37
|
Ye Y, Wang J, Hu Q, Hochu GM, Xin H, Wang C, Gu Z. Synergistic Transcutaneous Immunotherapy Enhances Antitumor Immune Responses through Delivery of Checkpoint Inhibitors. ACS NANO 2016; 10:8956-63. [PMID: 27599066 DOI: 10.1021/acsnano.6b04989] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Despite the promising efficacy of immunoregulation in cancer therapy, the clinical benefit has been restricted by inefficient infiltration of lymphocytes in the evolution of immune evasion. Also, immune-related adverse events have often occurred due to the off-target binding of therapeutics to normal tissues after systematic treatment. In light of this, we have developed a synergistic immunotherapy strategy that locally targets the immunoinhibitory receptor programmed cell death protein 1 (PD1) and immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO) for the treatment of melanoma through a microneedle-based transcutaneous delivery approach. The embedded immunotherapeutic nanocapsule loaded with anti-PD1 antibody (aPD1) is assembled from hyaluronic acid modified with 1-methyl-dl-tryptophan (1-MT), an inhibitor of IDO. This formulation method based on the combination strategy of "drug A in carriers formed by incorporation of drug B" facilitates the loading capacity of therapeutics. Moreover, the resulting delivery device elicits the sustained release and enhances retention of checkpoint inhibitors in the tumor microenvironment. Using a B16F10 mouse melanoma model, we demonstrate that this synergistic treatment has achieved potent antitumor efficacy, which is accompanied by enhanced effective T cell immunity as well as reduced immunosuppression in the local site.
Collapse
Affiliation(s)
- Yanqi Ye
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University , Raleigh, North Carolina 27695, United States
- Molecular Pharmaceutics Division and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Jinqiang Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University , Raleigh, North Carolina 27695, United States
- Molecular Pharmaceutics Division and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Quanyin Hu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University , Raleigh, North Carolina 27695, United States
- Molecular Pharmaceutics Division and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Gabrielle M Hochu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University , Raleigh, North Carolina 27695, United States
| | - Hongliang Xin
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University , Raleigh, North Carolina 27695, United States
| | - Chao Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University , Raleigh, North Carolina 27695, United States
- Molecular Pharmaceutics Division and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University , Raleigh, North Carolina 27695, United States
- Molecular Pharmaceutics Division and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina 27599, United States
- Department of Medicine, University of North Carolina School of Medicine , Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
38
|
Kim JM, Chen DS. Immune escape to PD-L1/PD-1 blockade: seven steps to success (or failure). Ann Oncol 2016; 27:1492-504. [PMID: 27207108 DOI: 10.1093/annonc/mdw217] [Citation(s) in RCA: 475] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/13/2016] [Indexed: 12/13/2022] Open
Abstract
The emergence of programmed death-ligand 1 (PD-L1)/programmed death-1 (PD-1)-targeted therapy has demonstrated the importance of the PD-L1 : PD-1 interaction in inhibiting anticancer T-cell immunity in multiple human cancers, generating durable responses and extended overall survival. However, not all patients treated with PD-L1/PD-1-targeted therapy experience tumor shrinkage, durable responses, or prolonged survival. To extend such benefits to more cancer patients, it is necessary to understand why some patients experience primary or secondary immune escape, in which the immune response is incapable of eradicating all cancer cells. Understanding immune escape from PD-L1/PD-1-targeted therapy will be important to the development of rational immune-combination therapy and predictive diagnostics and to the identification of novel immune targets. Factors that likely relate to immune escape include the lack of strong cancer antigens or epitopes recognized by T cells, minimal activation of cancer-specific T cells, poor infiltration of T cells into tumors, downregulation of the major histocompatibility complex on cancer cells, and immunosuppressive factors and cells in the tumor microenvironment. Precisely identifying and understanding these mechanisms of immune escape in individual cancer patients will allow for personalized cancer immunotherapy, in which monotherapy and combination immunotherapy are chosen based on the presence of specific immune biology. This approach may enable treatment with immunotherapy without inducing immune escape, resulting in a larger proportion of patients obtaining clinical benefit.
Collapse
Affiliation(s)
- J M Kim
- Genentech, South San Francisco
| | - D S Chen
- Genentech, South San Francisco Stanford Medical Oncology, Stanford University School of Medicine, Stanford, USA
| |
Collapse
|
39
|
Regulatory T Cells: Molecular Actions on Effector Cells in Immune Regulation. J Immunol Res 2016; 2016:1720827. [PMID: 27298831 PMCID: PMC4889823 DOI: 10.1155/2016/1720827] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 03/27/2016] [Indexed: 01/11/2023] Open
Abstract
T regulatory cells play a key role in the control of the immune response, both in health and during illness. While the mechanisms through which T regulatory cells exert their function have been extensively described, their molecular effects on effector cells have received little attention. Thus, this revision is aimed at summarizing our current knowledge on those regulation mechanisms on the target cells from a molecular perspective.
Collapse
|
40
|
Soliman HH, Minton SE, Han HS, Ismail-Khan R, Neuger A, Khambati F, Noyes D, Lush R, Chiappori AA, Roberts JD, Link C, Vahanian NN, Mautino M, Streicher H, Sullivan DM, Antonia SJ. A phase I study of indoximod in patients with advanced malignancies. Oncotarget 2016; 7:22928-38. [PMID: 27008709 PMCID: PMC5008412 DOI: 10.18632/oncotarget.8216] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 03/10/2016] [Indexed: 01/17/2023] Open
Abstract
PURPOSE Indoximod is an oral inhibitor of the indoleamine 2,3-dioxygenase pathway, which causes tumor-mediated immunosuppression. Primary endpoints were maximum tolerated dose (MTD) and toxicity for indoximod in patients with advanced solid tumors. Secondary endpoints included response rates, pharmacokinetics, and immune correlates. EXPERIMENTAL DESIGN Our 3+3 phase I trial comprised 10 dose levels (200, 300, 400, 600, and 800 mg once/day; 600, 800, 1200, 1600, and 2000 mg twice/day). Inclusion criteria were measurable metastatic solid malignancy, age ≥18 years, and adequate organ/marrow function. Exclusion criteria were chemotherapy ≤ 3 weeks prior, untreated brain metastases, autoimmune disease, or malabsorption. RESULTS In 48 patients, MTD was not reached at 2000 mg twice/day. At 200 mg once/day, 3 patients previously treated with checkpoint inhibitors developed hypophysitis. Five patients showed stable disease >6 months. Indoximod plasma AUC and Cmax plateaued above 1200mg. Cmax (~12 μM at 2000 mg twice/day) occurred at 2.9 hours, and half-life was 10.5 hours. C reactive protein (CRP) levels increased across multiple dose levels. CONCLUSIONS Indoximod was safe at doses up to 2000 mg orally twice/day. Best response was stable disease >6 months in 5 patients. Induction of hypophysitis, increased tumor antigen autoantibodies and CRP levels were observed.
Collapse
Affiliation(s)
- Hatem H. Soliman
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Susan E. Minton
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Hyo Sook Han
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Roohi Ismail-Khan
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Anthony Neuger
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Fatema Khambati
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - David Noyes
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Richard Lush
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | | | - John D. Roberts
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | | | | | | | - Howard Streicher
- Cancer Therapeutics Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Daniel M. Sullivan
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Scott J. Antonia
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| |
Collapse
|
41
|
Zong S, Li C, Luo C, Zhao X, Liu C, Wang K, Jia W, Bai M, Yin M, Bao S, Guo J, Kang J, Duan T, Zhou Q. Dysregulated expression of IDO may cause unexplained recurrent spontaneous abortion through suppression of trophoblast cell proliferation and migration. Sci Rep 2016; 6:19916. [PMID: 26814137 PMCID: PMC4728682 DOI: 10.1038/srep19916] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 12/21/2015] [Indexed: 12/25/2022] Open
Abstract
In pregnancy, trophoblast proliferation, migration and invasion are important for the establishment and maintenance of a successful pregnancy. Impaired trophoblast function has been implicated in recurrent spontaneous abortion (RSA), a major complication of pregnancy, but the underlying mechanisms remain unclear. Indoleamine 2,3-dioxygenase (IDO), an enzyme that catabolizes tryptophan along the kynurenine pathway, is highly expressed in the placenta and serum during pregnancy. Here, we identified a novel function of IDO in regulating trophoblast cell proliferation and migration. We showed that IDO expression and activity were decreased in unexplained recurrent spontaneous abortion (URSA) compared to normal pregnancy. Furthermore, blocking IDO in human trophoblast cells led to reduced proliferation and migration, along with decreased STAT3 phosphorylation and MMP9 expression. Increased STAT3 phosphorylation reversed the IDO knockdown-suppressed trophoblast cell proliferation and migration. In addition, the overexpression of IDO promoted cell proliferation and migration, which could be abolished by the STAT3 signaling inhibitor (AG490). Finally, we observed similar reductions of STAT3 phosphorylation and MMP9 expression in URSA patients. These results indicate that the level of IDO expression may be associated with pregnancy-related complications, such as URSA, by affecting trophoblast cell proliferation and migration via the STAT3 signaling pathway.
Collapse
Affiliation(s)
- Shanshan Zong
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Chunqing Li
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Chengfeng Luo
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Xin Zhao
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Chunhong Liu
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Kai Wang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Wenwen Jia
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Mingliang Bai
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Minghong Yin
- Department of Gynaecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Shihua Bao
- Department of Gynaecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Jie Guo
- Department of Obstetrics and Gynaecology, Punan hospital, Shanghai, 2000125, China
| | - jiuhong Kang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Tao Duan
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Qian Zhou
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| |
Collapse
|
42
|
Salazar F, Hall L, Negm OH, Awuah D, Tighe PJ, Shakib F, Ghaemmaghami AM. The mannose receptor negatively modulates the Toll-like receptor 4-aryl hydrocarbon receptor-indoleamine 2,3-dioxygenase axis in dendritic cells affecting T helper cell polarization. J Allergy Clin Immunol 2015; 137:1841-1851.e2. [PMID: 26703454 DOI: 10.1016/j.jaci.2015.10.033] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 10/09/2015] [Accepted: 10/27/2015] [Indexed: 01/23/2023]
Abstract
BACKGROUND Dendritic cells (DCs) are key players in the induction and re-elicitation of TH2 responses to allergens. We have previously shown that different C-type lectin receptors on DCs play a major role in allergen recognition and uptake. In particular, mannose receptor (MR), through modulation of Toll-like receptor (TLR) 4 signaling, can regulate indoleamine 2,3-dioxygenase (IDO) activity, favoring TH2 responses. Interestingly, the aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor with an emerging role in immune modulation, has been implicated in IDO activation in response to TLR stimulation. OBJECTIVE Here we investigated how allergens and lectins modulate the TLR4-AhR-IDO axis in human monocyte-derived DCs. METHODS Using a combination of genomics, proteomics, and immunologic studies, we investigated the role of MR and AhR in IDO regulation and its effect on T helper cell differentiation. RESULTS We have demonstrated that LPS induces both IDO isoforms (IDO1 and IDO2) in DCs, with partial involvement of AhR. Additionally, we found that, like mannan, different airborne allergens can effectively downregulate TLR4-induced IDO1 and IDO2 expression, most likely through binding to the MR. Mannose-based ligands were also able to downregulate IL-12p70 production by DCs, affecting T helper cell polarization. Interestingly, AhR and some components of the noncanonical nuclear factor κB pathway were shown to be downregulated after MR engagement, which could explain the regulatory effects of MR on IDO expression. CONCLUSION Our work demonstrates a key role for MR in the modulation of the TLR4-AhR-IDO axis, which has a significant effect on DC behavior and the development of immune responses against allergens.
Collapse
Affiliation(s)
- Fabián Salazar
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Laurence Hall
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Ola H Negm
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom; Medical Microbiology and Immunology Department, Mansoura University, Mansoura, Egypt
| | - Dennis Awuah
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Patrick J Tighe
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Farouk Shakib
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Amir M Ghaemmaghami
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom.
| |
Collapse
|
43
|
Miller EA, Gopal R, Valdes V, Berger JS, Bhardwaj N, O'Brien MP. Soluble CD40 ligand contributes to dendritic cell-mediated T-cell dysfunction in HIV-1 infection. AIDS 2015; 29:1287-96. [PMID: 26091297 PMCID: PMC4478195 DOI: 10.1097/qad.0000000000000698] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Plasma soluble CD40 ligand (sCD40L) is increased during HIV-1 infection, but it is unknown whether it circulates in monomeric or multimeric forms, and whether the circulating forms have differential effects on myeloid dendritic cell function and adaptive regulation. DESIGN sCD40L forms were measured in plasma samples from HIV-infected donors. The effects of sCD40L forms on dendritic cell function were measured in vitro. METHODS To delineate which forms of sCD40L are present in plasma from HIV-infected donors, immunoblots were performed following enrichment of plasma for medium and low-abundance proteins. Dendritic cells from seronegative donors were exposed to multiple forms of sCD40L prior to Toll-like receptor stimulation and dendritic cell function and adaptive regulation was assessed in vitro. RESULTS Monomeric and multimeric forms of sCD40L were identified in plasma from antiretroviral therapy-treated HIV-infected donors. Although monomeric and multimeric forms of sCD40L had differential effects on dendritic cell activation when given alone, both strongly suppressed secretion of the Th1 skewing cytokine, interleukin-12, upon subsequent Toll-like receptor stimulation. Furthermore, dendritic cells exposed to both monomeric and multimeric sCD40L induced regulatory T-cell formation and T-cell anergy. CONCLUSION Elevated sCD40L during HIV infection impairs dendritic cell function, contributing to innate and adaptive immune dysfunction. Antiretroviral adjunctive therapies that decrease sCD40L may provide immune modulatory benefits.
Collapse
Affiliation(s)
- Elizabeth A Miller
- aDivision of Infectious Diseases, Department of Medicine bDivision of Hematology and Oncology, Hess Center for Science and Medicine, Icahn School of Medicine at Mount Sinai cDivision of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York, USA
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
IDO1 (indoleamine 2,3-dioxygenase 1) is a member of a unique class of mammalian haem dioxygenases that catalyse the oxidative catabolism of the least-abundant essential amino acid, L-Trp (L-tryptophan), along the kynurenine pathway. Significant increases in knowledge have been recently gained with respect to understanding the fundamental biochemistry of IDO1 including its catalytic reaction mechanism, the scope of enzyme reactions it catalyses, the biochemical mechanisms controlling IDO1 expression and enzyme activity, and the discovery of enzyme inhibitors. Major advances in understanding the roles of IDO1 in physiology and disease have also been realised. IDO1 is recognised as a prominent immune regulatory enzyme capable of modulating immune cell activation status and phenotype via several molecular mechanisms including enzyme-dependent deprivation of L-Trp and its conversion into the aryl hydrocarbon receptor ligand kynurenine and other bioactive kynurenine pathway metabolites, or non-enzymatic cell signalling actions involving tyrosine phosphorylation of IDO1. Through these different modes of biochemical signalling, IDO1 regulates certain physiological functions (e.g. pregnancy) and modulates the pathogenesis and severity of diverse conditions including chronic inflammation, infectious disease, allergic and autoimmune disorders, transplantation, neuropathology and cancer. In the present review, we detail the current understanding of IDO1’s catalytic actions and the biochemical mechanisms regulating IDO1 expression and activity. We also discuss the biological functions of IDO1 with a focus on the enzyme's immune-modulatory function, its medical implications in diverse pathological settings and its utility as a therapeutic target.
Collapse
|
45
|
Dąbrowski R, Hagman R, Tvarijonaviciute A, Pastor J, Kocki T, Turski WA. Serum tryptophan and its metabolites in female dogs undergoing ovariohysterectomy as treatment of pyometra or as elective spay surgery. Theriogenology 2015; 83:1279-86. [DOI: 10.1016/j.theriogenology.2015.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Revised: 11/29/2014] [Accepted: 01/09/2015] [Indexed: 10/24/2022]
|
46
|
Bischoff L, Alvarez S, Dai DL, Soukhatcheva G, Orban PC, Verchere CB. Cellular mechanisms of CCL22-mediated attenuation of autoimmune diabetes. THE JOURNAL OF IMMUNOLOGY 2015; 194:3054-64. [PMID: 25740943 DOI: 10.4049/jimmunol.1400567] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Autoimmune destruction of insulin-producing β cells in type 1 diabetes and islet transplantation involves a variety of immune pathways but is primarily mediated by self-reactive T cells. Chemokines can modulate local immune responses in inflammation and tumors by recruiting immune cells. We have reported that expression of the chemokine CCL22 in pancreatic β cells in the NOD mouse prevents autoimmune attack by recruiting T regulatory cells (Tregs), protecting mice from diabetes. In this study we show that invariant NKT cells are also recruited to CCL22-expressing islet transplants and are required for CCL22-mediated protection from autoimmunity. Moreover, CCL22 induces an influx of plasmacytoid dendritic cells, which correlates with higher levels of IDO in CCL22-expressing islet grafts. In addition to its chemotactic properties, we found that CCL22 activates Tregs and promotes their ability to induce expression of IDO by dendritic cells. Islet CCL22 expression thus produces a tolerogenic milieu through the interplay of Tregs, invariant NKT cells, and plasmacytoid dendritic cells, which results in suppression of effector T cell responses and protection of β cells. The immunomodulatory properties of CCL22 could be harnessed for prevention of graft rejection and type 1 diabetes as well as other autoimmune disorders.
Collapse
Affiliation(s)
- Loraine Bischoff
- Department of Pathology and Laboratory Medicine, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada; and
| | - Sigrid Alvarez
- Department of Pathology and Laboratory Medicine, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada; and
| | - Derek L Dai
- Department of Pathology and Laboratory Medicine, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada; and
| | - Galina Soukhatcheva
- Department of Pathology and Laboratory Medicine, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada; and
| | - Paul C Orban
- Department of Pathology and Laboratory Medicine, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada; and
| | - C Bruce Verchere
- Department of Pathology and Laboratory Medicine, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada; and Department of Surgery, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| |
Collapse
|
47
|
Lysophosphatidylcholine exacerbates Leishmania major-dendritic cell infection through interleukin-10 and a burst in arginase1 and indoleamine 2,3-dioxygenase activities. Int Immunopharmacol 2015; 25:1-9. [DOI: 10.1016/j.intimp.2015.01.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 01/01/2015] [Accepted: 01/06/2015] [Indexed: 02/08/2023]
|
48
|
Shirshev SV. Molecular mechanisms of hormonal and hormonal-cytokine control of immune tolerance in pregnancy. BIOCHEMISTRY (MOSCOW) SUPPLEMENT SERIES A: MEMBRANE AND CELL BIOLOGY 2015. [DOI: 10.1134/s1990747814050079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
49
|
Zhang A, Zheng Y, Que Z, Zhang L, Lin S, Le V, Liu J, Tian J. Astragaloside IV inhibits progression of lung cancer by mediating immune function of Tregs and CTLs by interfering with IDO. J Cancer Res Clin Oncol 2014; 140:1883-90. [PMID: 24980548 DOI: 10.1007/s00432-014-1744-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 06/10/2014] [Indexed: 12/24/2022]
Abstract
PURPOSE Tumor cells have developed multiple mechanisms to escape immune recognition mediated by T cells. Indoleamine 2,3-dioxygenase (IDO), a tryptophan-catabolizing enzyme inducing immune tolerance, is involved in tumor escape from host immune systems in mice. Astragaloside IV (AS-IV), an extract from a commonly used Chinese medicinal plant Astragalus membranaceus, has been shown to be capable of restoring the impaired T-cell functions in cancer patients. The purpose of this study was to investigate the mechanisms underlying the anticancer properties of AS-IV. METHODS Here, we used IDO-overexpressed murine Lewis lung carcinoma cells to establish an orthotopic lung cancer model in C57BL/6 mice. Next, tumor growth was evaluated in several different treatment groups: control (saline), AS-IV, paclitaxel, and 1-methyl tryptophan (an inhibitor of IDO). We then analyzed the percentages of various immune cell subsets among the splenic lymphocytes of lung cancer mice by flow cytometry. The level of IDO was measured by real-time PCR and Western blot. RESULTS We showed that the growth of tumor was suppressed by AS-IV treatment in vivo. AS-IV also could down-regulate regulatory T cells (Tregs) and up-regulate cytotoxic T lymphocytes (CTLs) in vivo and in vitro. Consistent with its ability to interfere with T-cell immunity, AS-IV blocked IDO induction both in vitro and in vivo. CONCLUSIONS The results of these studies indicate that AS-IV has in vivo anticancer activity and can enhance the immune response by inhibiting the Tregs frequency and induce the activity of CTLs, which might be related to the inhibition of IDO expression.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/enzymology
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/pathology
- Cell Line, Tumor
- Coculture Techniques
- Disease Progression
- Drug Screening Assays, Antitumor
- Female
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Mice, Inbred C57BL
- Neoplasm Transplantation
- Paclitaxel/pharmacology
- Paclitaxel/therapeutic use
- Saponins/pharmacology
- Saponins/therapeutic use
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- Triterpenes/pharmacology
- Triterpenes/therapeutic use
- Tryptophan/analogs & derivatives
- Tryptophan/pharmacology
- Tryptophan/therapeutic use
- Tumor Burden/drug effects
- Tumor Escape/drug effects
Collapse
Affiliation(s)
- Anle Zhang
- Department of Molecular & Cellular Pharmacology, Biomedical Nanotechnology Center, State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Li F, Huang J, Li S, Li H, Yu J, Ren X, Liu J. The subsets of dendritic cells and memory T cells correspond to indoleamine 2,3-dioxygenase in stomach tumor microenvironment. Tumour Biol 2014; 35:8691-8. [PMID: 24870595 DOI: 10.1007/s13277-014-2126-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 05/20/2014] [Indexed: 11/27/2022] Open
Abstract
The abnormal distributions of memory T cells (Tm) and dendritic cells (DC) in stomach cancer are not well understood. Indoleamine 2,3-dioxygenase (IDO), produced by DC, may be an important enzyme affecting function and proliferation of Tm. In this study, IDO expression was examined by immunohistochemical staining. The subsets of Tm and DC were counted by flow cytometry. The percentages of CD4 + Tm and CD4 + central Tm (Tcm) were lower in tumor tissues than in normal tissues (P < 0.05), while the CD4 + effector Tm (Tem) and CD8 + Tem percentages were higher in tumor tissues (P < 0.05). The ratio of myeloid DC (DC1)/plasmacytoid DC (DC2) was significantly lower in tumor tissues (P = 0.009). The high expression of IDO was more frequently observed in tumor tissues (P = 0.001). The percentages of CD4 + Tm and CD8 + Tm were positively associated with DC1 percentage and ratio of DC1/DC2 (P < 0.05). The higher CD8 + Tcm percentage was associated with higher DC2 percentage (P = 0.025). The patients with high IDO expression had significantly lower CD4 + Tm (P = 0.012) and CD8 + Tm percentages (P = 0.033), but higher CD8 + Tem percentage (P < 0.01). Concerning on clinicopathologic features, the higher DC2 percentage was associated with larger tumor size (P = 0.019). The CD4 + Tm and CD8 + Tem percentages were significantly associated with clinical stage and lymph node metastasis; the high IDO expressions were significantly associated with deeper tumor invasion (P = 0.016) and lymph node metastasis (P = 0.038). Thus, DC subsets, Tm subsets, and IDO expression were correlated with each other. They were associated with the established clinicopathologic features, such as tumor size, depth of invasion, lymph node metastasis, and clinical stage.
Collapse
Affiliation(s)
- Fangxuan Li
- Cancer Prevention Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | | | | | | | | | | | | |
Collapse
|