1
|
Nagano T, Takada K, Hashinokuchi A, Matsudo K, Kinoshita F, Akamine T, Kohno M, Shimokawa M, Takenaka T, Oda Y, Yoshizumi T. Clinical significance of CD155 expression in surgically resected lung squamous cell carcinoma. Int J Clin Oncol 2025; 30:62-71. [PMID: 39441454 DOI: 10.1007/s10147-024-02640-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Cluster of differentiation 155 (CD155) is expressed in many tumor types. CD155 is involved in the immune avoidance of tumor cells and contributes to tumor development and progression. Therefore, CD155 is a novel target for cancer immunotherapy. The clinical significance of CD155 expression in lung squamous cell carcinoma (LUSC) has not been fully elucidated. MATERIALS AND METHODS We performed a retrospective analysis of 264 patients with surgically resected LUSC. Immunohistochemistry was used to evaluate CD155 expression. The association of CD155 expression with clinicopathological features and clinical outcomes was assessed. We also analyzed the relationship between CD155 expression and programmed cell death-ligand 1 (PD-L1) expression and tumor-infiltrating lymphocytes. RESULTS Among the 264 patients, 137 patients (51.9%) were classified in the high CD155 expression group. High CD155 expression was significantly associated with pleural invasion, vascular invasion, PD-L1 positivity, and high CD3, CD4, and CD8 expressions. In multivariate analysis, the presence of pleural invasion and PD-L1 positivity were independent predictors of high CD155 expression. Kaplan-Meier curve analysis showed that high CD155 expression was significantly associated with shorter disease-free survival and overall survival. In multivariate analysis, high CD155 expression was an independent poor prognostic factor for overall survival, but not for disease-free survival. Subgroup analyses revealed that the prognostic effect of CD155 expression was observed in the PD-L1 positive group but not the PD-L1 negative group. CONCLUSION Our analysis revealed that high CD155 expression significantly predicted poor prognosis in patients with surgically resected LUSC, especially in patients with PD-L1-positive tumors.
Collapse
Affiliation(s)
- Taichi Nagano
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuki Takada
- Department of Surgery, Saiseikai Fukuoka General Hospital, 1-3-46 Tenjin, Chuo-ku, Fukuoka, 810-0001, Japan.
| | - Asato Hashinokuchi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kyoto Matsudo
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fumihiko Kinoshita
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takaki Akamine
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mikihiro Kohno
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mototsugu Shimokawa
- Department of Biostatistics, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Tomoyoshi Takenaka
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
2
|
Lin Z, Zhao M, Zhang X, Piao J, Zheng X, Shu S, Zhao L, Zhang M, Shi GP, Lei Y, Cui R, Yue X, Cheng XW. CD8 + T-cell deficiency protects mice from abdominal aortic aneurysm formation in response to calcium chloride 2. J Hypertens 2024; 42:1966-1975. [PMID: 39146540 PMCID: PMC11451972 DOI: 10.1097/hjh.0000000000003823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/17/2024] [Accepted: 07/08/2024] [Indexed: 08/17/2024]
Abstract
OBJECTIVE Abdominal aortic aneurysm (AAA) is an aneurysm-like dilated and highly fatal cardiovascular disease. CD8 + T cells have been shown to be critical for vascular pathological processes, but the contribution of these lymphocytes to vascular diseases remains elusive. METHODS AND RESULTS Eight-week-old male wildtype (CD8 +/+ ) and Cd8a knockout (CD8 -/- ) mice were used in a calcium chloride 2 (CaCl 2 )-induced experimental AAA model. At 6 weeks after surgery, CD8 + T-cell deletion prevented the formation of AAA, accompanied by reductions of the levels of inflammatory (interferon-γ [IFN-γ], interleukin-1β, monocyte chemoattractant protein-1, intracellular adhesion molecule-1, vascular cell adhesion molecule-1, NOD-like receptor protein 3, caspase-1), oxidative stress [NADPH oxidase and gp91 phox ], and proteolysis (cathepsin S, cathepsin K, matrix metalloproteinase-2 [MMP-2] and MMP-9) proteins and/or genes in plasma and/or AAA tissues. Immunoreactivities of MMP-2 and MMP-9 were observed in macrophages. An injection of IFN-γ and adoptive transfer of CD8 + T cells of IFN-γ +/+ mice diminished CD8 -/- -mediated vasculoprotective actions in the AAA mice. In vitro, IFN-γ enhanced MMP-2 and MMP-9 gelatinolytic activities in macrophage and/or vascular smooth muscle cells. CONCLUSION The vasculoprotective effects of CD8 + T-cell deletion in a mouse CaCl 2 -induced AAA model were likely attributable to, at least in part, the attenuation of IFN-γ-dependent inflammation action, oxidative stress production, and proteolysis, suggesting a novel therapeutic target for AAA formation by regulating CD8 + T-cell-derived IFN-γ secretion.
Collapse
Affiliation(s)
- Zhuo Lin
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin
| | - Mantong Zhao
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital
| | - Xian Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui
| | - Jinshun Piao
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital
| | - Xintong Zheng
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital
| | - Shangzhi Shu
- Department of Cardiovascular Disease, the First Hospital of Jilin University, Changchun, Jilin PR, China
| | - Longguo Zhao
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital
| | - Meiping Zhang
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Yanna Lei
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital
| | - Rihua Cui
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital
| | - Xueling Yue
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, Japan
| |
Collapse
|
3
|
Bhat SA, Parveen A, Gormley E, Meade KG. Extensive differential DNA methylation between tuberculosis skin test positive and skin test negative cattle. BMC Genomics 2024; 25:762. [PMID: 39107682 PMCID: PMC11301934 DOI: 10.1186/s12864-024-10574-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/27/2024] [Indexed: 08/10/2024] Open
Abstract
Bovine tuberculosis (bTB), caused by Mycobacterium bovis (M. bovis), represents a significant problem for the agriculture industry as well as posing a risk for human health. Current diagnostic tests for bTB target the cell-mediated immune (CMI) response to infection with M. bovis, primarily through screening of animals with the tuberculin skin test. Epigenetic modifications have been shown to alter the course of the immune response and differentially methylated regions (DMRs) might also influence the outcome of the skin test in cattle. Whole Genome Bisulphite Sequencing (WGBS) was used to profile DNA methylation levels from peripheral blood of a group of cattle identified as test positive for M. bovis (positive for the single intradermal comparative tuberculin test (SICTT) and/or the interferon-γ release assay compared to a test negative control group [n = 8/group, total of 16 WGBS libraries]. Although global methylation profiles were similar for both groups across the genome, 223 DMRs and 159 Differentially Promoter Methylated Genes (DPMGs) were identified between groups with an excess of hypermethylated sites in SICTT positive cattle (threshold > 15% differential methylation). Genes located within these DMRs included the Interleukin 1 receptor (IL1R1) and MHC related genes (BOLA and BOLA-DQB). KEGG pathway analysis identified enrichment of genes involved in Calcium and MAPK signalling, as well as metabolism pathways. Analysis of DMRs in a subset of SICTT negative cattle that were IFN-γ positive showed differential methylation of genes including Interleukin 10 Receptor, alpha (IL10RA), Interleukin 17 F (IL17F) and host defence peptides (DEFB and BDEF109). This study has identified a number of immune gene loci at which differential methylation is associated with SICTT test results and the degree of methylation could influence effective host immune responses.
Collapse
Affiliation(s)
- Sajad A Bhat
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland
| | - Alia Parveen
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland
| | - Eamonn Gormley
- UCD School of Veterinary Medicine, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland
| | - Kieran G Meade
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland.
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland.
- UCD Institute of Food and Health, University College Dublin, Belfield, Dublin, C15 PW93, Ireland.
| |
Collapse
|
4
|
Qi W, Tian J, Wang G, Yan Y, Wang T, Wei Y, Wang Z, Zhang G, Zhang Y, Wang J. Advances in cellular and molecular pathways of salivary gland damage in Sjögren's syndrome. Front Immunol 2024; 15:1405126. [PMID: 39050857 PMCID: PMC11266040 DOI: 10.3389/fimmu.2024.1405126] [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: 03/22/2024] [Accepted: 05/28/2024] [Indexed: 07/27/2024] Open
Abstract
Sjögren's Syndrome (SS) is an autoimmune disorder characterized by dysfunction of exocrine glands. Primarily affected are the salivary glands, which exhibit the most frequent pathological changes. The pathogenesis involves susceptibility genes, non-genetic factors such as infections, immune cells-including T and B cells, macrophage, dendritic cells, and salivary gland epithelial cells. Inflammatory mediators such as autoantibodies, cytokines, and chemokines also play a critical role. Key signaling pathways activated include IFN, TLR, BAFF/BAFF-R, PI3K/Akt/mTOR, among others. Comprehensive understanding of these mechanisms is crucial for developing targeted therapeutic interventions. Thus, this study explores the cellular and molecular mechanisms underlying SS-related salivary gland damage, aiming to propose novel targeted therapeutic approaches.
Collapse
Affiliation(s)
- Wenxia Qi
- Gansu University of Traditional Chinese Medicine, College of Integrative Medicine, Lanzhou, China
| | - Jiexiang Tian
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Department of Rheumatology and Orthopedics, Lanzhou, China
| | - Gang Wang
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Department of Rheumatology and Orthopedics, Lanzhou, China
| | - Yanfeng Yan
- Fourth Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Department of Respiratory and Critical Care Medicine, Lanzhou, China
| | - Tao Wang
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Department of Rheumatology and Orthopedics, Lanzhou, China
| | - Yong Wei
- Gansu University of Traditional Chinese Medicine, College of Integrative Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Department of Rheumatology and Orthopedics, Lanzhou, China
| | - Zhandong Wang
- Gansu University of Traditional Chinese Medicine, College of Integrative Medicine, Lanzhou, China
| | - Guohua Zhang
- Gansu University of Traditional Chinese Medicine, College of Integrative Medicine, Lanzhou, China
| | - Yuanyuan Zhang
- Gansu University of Traditional Chinese Medicine, College of Integrative Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Department of Rheumatology and Orthopedics, Lanzhou, China
| | - Jia Wang
- Gansu University of Traditional Chinese Medicine, College of Integrative Medicine, Lanzhou, China
| |
Collapse
|
5
|
Shen T, Li Y, Liu T, Lian Y, Kong L. Association between Mycoplasma pneumoniae infection, high‑density lipoprotein metabolism and cardiovascular health (Review). Biomed Rep 2024; 20:39. [PMID: 38357242 PMCID: PMC10865299 DOI: 10.3892/br.2024.1729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/09/2024] [Indexed: 02/16/2024] Open
Abstract
The association between Mycoplasma pneumoniae (M. pneumoniae) infection, high-density lipoprotein metabolism and cardiovascular disease is an emerging research area. The present review summarizes the basic characteristics of M. pneumoniae infection and its association with high-density lipoprotein and cardiovascular health. M. pneumoniae primarily invades the respiratory tract and damages the cardiovascular system through various mechanisms including adhesion, invasion, secretion of metabolites, production of autoantibodies and stimulation of cytokine production. Additionally, the present review highlights the potential role of high-density lipoprotein for the development of prevention and intervention of M. pneumoniae infection and cardiovascular disease, and provides suggestions for future research directions and clinical practice. It is urgent to explore the specific mechanisms underlying the association between M. pneumoniae infection, high-density lipoprotein metabolism, and cardiovascular disease and analyze the roles of the immune system and inflammatory response.
Collapse
Affiliation(s)
- Tao Shen
- Department of Clinical Laboratory, Jincheng People's Hospital, Jincheng, Shanxi 048000, P.R. China
- Jincheng Hospital Affiliated to Changzhi Medical College, Jincheng, Shanxi 048000, P.R. China
| | - Yanfang Li
- Department of Clinical Laboratory, Jincheng People's Hospital, Jincheng, Shanxi 048000, P.R. China
- Jincheng Hospital Affiliated to Changzhi Medical College, Jincheng, Shanxi 048000, P.R. China
| | - Tingting Liu
- Department of Clinical Laboratory, Jincheng People's Hospital, Jincheng, Shanxi 048000, P.R. China
- Jincheng Hospital Affiliated to Changzhi Medical College, Jincheng, Shanxi 048000, P.R. China
| | - Yunzhi Lian
- Department of Clinical Laboratory, Jincheng People's Hospital, Jincheng, Shanxi 048000, P.R. China
- Jincheng Hospital Affiliated to Changzhi Medical College, Jincheng, Shanxi 048000, P.R. China
| | - Luke Kong
- Department of Clinical Laboratory, Jincheng People's Hospital, Jincheng, Shanxi 048000, P.R. China
- Jincheng Hospital Affiliated to Changzhi Medical College, Jincheng, Shanxi 048000, P.R. China
| |
Collapse
|
6
|
Schober R, Brandus B, Laeremans T, Iserentant G, Rolin C, Dessilly G, Zimmer J, Moutschen M, Aerts JL, Dervillez X, Seguin-Devaux C. Multimeric immunotherapeutic complexes activating natural killer cells towards HIV-1 cure. J Transl Med 2023; 21:791. [PMID: 37936122 PMCID: PMC10631209 DOI: 10.1186/s12967-023-04669-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/27/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Combination antiretroviral therapy (cART) has dramatically extended the life expectancy of people living with HIV-1 and improved their quality of life. There is nevertheless no cure for HIV-1 infection since HIV-1 persists in viral reservoirs of latently infected CD4+ T cells. cART does not eradicate HIV-1 reservoirs or restore cytotoxic natural killer (NK) cells which are dramatically reduced by HIV-1 infection, and express the checkpoint inhibitors NKG2A or KIR2DL upregulated after HIV-1 infection. Cytotoxic NK cells expressing the homing receptor CXCR5 were recently described as key subsets controlling viral replication. METHODS We designed and evaluated the potency of "Natural killer activating Multimeric immunotherapeutic compleXes", called as NaMiX, combining multimers of the IL-15/IL-15Rα complex with an anti-NKG2A or an anti-KIR single-chain fragment variable (scFv) to kill HIV-1 infected CD4+ T cells. The oligomerization domain of the C4 binding protein was used to associate the IL-15/IL-15Rα complex to the scFv of each checkpoint inhibitor as well as to multimerize each entity into a heptamer (α form) or a dimer (β form). Each α or β form was compared in different in vitro models using one-way ANOVA and post-hoc Tukey's tests before evaluation in humanized NSG tg-huIL-15 mice having functional NK cells. RESULTS All NaMiX significantly enhanced the cytolytic activity of NK and CD8+ T cells against Raji tumour cells and HIV-1+ ACH-2 cells by increasing degranulation, release of granzyme B, perforin and IFN-γ. Targeting NKG2A had a stronger effect than targeting KIR2DL due to higher expression of NKG2A on NK cells. In viral inhibition assays, NaMiX initially increased viral replication of CD4+ T cells which was subsequently inhibited by cytotoxic NK cells. Importantly, anti-NKG2A NaMiX enhanced activation, cytotoxicity, IFN-γ production and CXCR5 expression of NK cells from HIV-1 positive individuals. In humanized NSG tg-huIL-15 mice, we confirmed enhanced activation, degranulation, cytotoxicity of NK cells, and killing of HIV-1 infected cells from mice injected with the anti-NKG2A.α NaMiX, as compared to control mice, as well as decreased total HIV-1 DNA in the lung. CONCLUSIONS NK cell-mediated killing of HIV-1 infected cells by NaMiX represents a promising approach to support HIV-1 cure strategies.
Collapse
Affiliation(s)
- Rafaëla Schober
- Department of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg
| | - Bianca Brandus
- Department of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg
| | - Thessa Laeremans
- Neuro-Aging and Viro-Immunotherapy (NAVI) Research Group, Faculty of Pharmacy and Medicine, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Gilles Iserentant
- Department of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg
| | - Camille Rolin
- Department of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg
| | - Géraldine Dessilly
- AIDS Reference Laboratory, Catholic University of Louvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Jacques Zimmer
- Department of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg
| | - Michel Moutschen
- Department of Infectious Diseases, University of Liège, CHU de Liège, Liège, Belgium
| | - Joeri L Aerts
- Neuro-Aging and Viro-Immunotherapy (NAVI) Research Group, Faculty of Pharmacy and Medicine, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Xavier Dervillez
- Department of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg
| | - Carole Seguin-Devaux
- Department of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg.
| |
Collapse
|
7
|
Han Y, Fan X, Fan L, Wu Y, Zhou Z, Wang G, Guo L, Gao W, Chen Y, Gao Q. Liujunzi decoction exerts potent antitumor activity in oesophageal squamous cell carcinoma by inhibiting miR-34a/STAT3/IL-6R feedback loop, and modifies antitumor immunity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 111:154672. [PMID: 36701994 DOI: 10.1016/j.phymed.2023.154672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/30/2022] [Accepted: 01/12/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Liujunzi decoction (LJZD), a traditional herbal formula and one of the most commonly used adjuvant medications for the treatment of oesophageal squamous cell carcinoma (ESCC), exerts good antitumor and immunomodulatory activity. However, its specific mechanism of action remains largely unclear. PURPOSE In order to examine the potential primary and adjuvant antitumor mechanisms of LJZD, both in vitro and in vivo. METHODS IL-6 and miR-34a inhibitors were used to activate the miR-34a/STAT3/IL-6R feedback loop to observe the effects of LJZD. A humanised mouse model with a functional human immune system was constructed to evaluate the antitumor efficacy of LJZD in vivo on xenograft tumours, which was compared to that of the positive control drug anti-PD-1 monoclonal antibodies (mAb). Finally, a co-culture system of peripheral blood mononuclear and tumour cells in vitro was used to analyse the cytotoxic activity of LJZD on T cells. RESULTS LJZD significantly interfered with IL-6-induced activation of the miR-34a/STAT3/IL-6R feedback loop in ESCC by restoring the expression of the tumour suppressor miR-34a, and inhibited the proliferation of EC109 oesophageal cancer cells in a dose-dependant manner. Furthermore, LJZD effectively suppressed oesophageal tumour growth in vivo and alleviated organ injury and visceral index. Furthermore, LJZD boosted antitumor immunity by increasing IFN-γ expression and CD8+tumour-infiltrating lymphocytes (TILs) infiltration in the peripheral blood and tumour tissues, respectively, which may be related to a decrease in PD-1, but not PD-L1 expression. Finally, we confirmed that LJZD strengthens the killing ability of T cells by suppressing PD-1 expression in a co-culture system in vitro. CONCLUSION LJZD exerts excellent antitumor effect by interfering with the miR-34a/STAT3/IL-6R feedback loop and augmenting antitumor immune responses. Which provides new insights into mechanisms for LJZD and sheds light on the multifaceted role of phytomedicine in cancer.
Collapse
Affiliation(s)
- Yicun Han
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; The Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450008, China
| | - Xiuqi Fan
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; The Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450008, China
| | - Liyan Fan
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China
| | - Yaosong Wu
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China
| | - Zhexu Zhou
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China
| | - Ge Wang
- The Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450008, China
| | - Lanwei Guo
- The Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450008, China
| | - Wendong Gao
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China
| | - Yulong Chen
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China.
| | - Qilong Gao
- The Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450008, China.
| |
Collapse
|
8
|
Moetlhoa B, Tjale M, Pretorius A, Hayeshi R, Grobler A, Mokoena NB. Rift Valley Fever vaccine strategies: Enhanced stability of RVF Clone 13. Vaccine 2023; 41:1050-1058. [PMID: 36593173 DOI: 10.1016/j.vaccine.2022.12.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 01/02/2023]
Abstract
Rift Valley Fever virus (RVFV) causes the zoonotic RVF disease, which results in substantial economic losses in livestock industries. Regular vaccination of livestock against RVF is necessary to generate long-term immunity and avoid the loss of livestock. The live attenuated vaccine based on Clone 13 virus strain has been used to reduce the negative impact of RVF disease. The vaccine strain is heat labile and requires stringent conditions for storage and handling. This research evaluated lactose and sucrose-based stabilizers coupled with lyophilisation to enhance stability of the RVF Clone 13 vaccine strain. The glass transition temperature (Tg) of the sucrose-RVF vaccine was 97.0 °C with average residual moisture of below 2 %. The lactose formulation was characterised with Tg of 83.5 °C and residual moisture of above 2 %. The RVF Clone 13 sucrose-based formulation maintained higher antigen titres during lyophilisation compared to the lactose-formulated vaccine. Cellular-mediated and humoral immunity was evaluated and compared for the two newly formulated vaccines. Pheroid® technology was also investigated as a potential adjuvant and its ability to further enhance the immunogenicity conferred by the RVF Clone 13 vaccine formulations in Merino sheep. No adverse reactions were observed following injection of the vaccine formulations in mice, guinea pigs and Merino sheep. Comparable protective humoral immune responses against RVF were obtained for all animals vaccinated with the lactose and sucrose-based stabilisers with and without the Pheroid® adjuvant. No proliferation of CD8+ and CD4+ T-cells as well as expression of IFN-γ was observed for all animals group vaccinated with Pheroid® only. Specific CD8+ IFN-γ+T-cells were expressed at higher levels compared to the CD4+ IFN-γ+T-cells in the RVF Clone 13 vaccines, suggesting that cellular immunity against RVF is through the Class I antigen presentation pathway.
Collapse
Affiliation(s)
- Boitumelo Moetlhoa
- Onderstepoort Biological Products (OBP) SOC Ltd, 100 Old Soutpan Road, Onderstepoort, 0110, South Africa; DSI/NWU Preclinical Drug Development Platform (PCDDP), Faculty of Health Sciences, North-West University, Potchefstroom 2520, South Africa
| | - Mabotse Tjale
- Onderstepoort Biological Products (OBP) SOC Ltd, 100 Old Soutpan Road, Onderstepoort, 0110, South Africa; Biophotonics, National Laser Centre, Council for Scientific and Industrial Research, Meiring Naudé Road Brummeria, Pretoria, South Africa
| | - Alri Pretorius
- ARC-Onderstepoort Veterinary Research (OVR), 100 Old Soutpan Road, Onderstepoort, 0110, South Africa
| | - Rose Hayeshi
- DSI/NWU Preclinical Drug Development Platform (PCDDP), Faculty of Health Sciences, North-West University, Potchefstroom 2520, South Africa
| | - Anne Grobler
- DSI/NWU Preclinical Drug Development Platform (PCDDP), Faculty of Health Sciences, North-West University, Potchefstroom 2520, South Africa
| | - Nobalanda B Mokoena
- Onderstepoort Biological Products (OBP) SOC Ltd, 100 Old Soutpan Road, Onderstepoort, 0110, South Africa.
| |
Collapse
|
9
|
Production of Stem Cell Antigen 1 Sca-1/Ly-6A/E by Freshly Isolated NK Cells Cultured with Relevant Cytokines. Bull Exp Biol Med 2022; 174:62-65. [PMID: 36437320 DOI: 10.1007/s10517-022-05649-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Indexed: 11/29/2022]
Abstract
Activated NK cells in appropriate conditions are known to express stem cell antigen 1 (Sca-1/Ly-6A/E). To investigate its production, NK cells isolated from mouse spleens were incubated ex vivo in the presence of different combinations of cytokines (IL-12, IL-15, IL-18, and IFNγ). Expression of Sca-1 was found to be considerably higher in NK cells incubated in the presence of IL-18, IL-15, and IL-12 than in those treated with IL-15 and IL-18 only. To test the hypothesis that the effect of IL-12 was due to stimulation of IFNγ production, we replaced IL-12 with IFNγ in some samples and added specific anti-IFNγ antibody to some samples cultured with IL-15/IL-18+IL-12. In the subpopulations incubated in the presence of IL-15/IL-18 with added IFNγ instead of IL-12, the expression of Sca-1 was not increased. By contrast, in samples treated with IL-15/IL-18+IL-12 and anti-IFNγ antibody, the expression of Sca-1 was activated to a similar extent as in those stimulated by IL-15/IL-18+IL-12 combination without the antibody. The obtained data suggest that IL-12 activates the production of Sca-1 by NK cells through an IFNγ-independent mechanism.
Collapse
|
10
|
Castillo R, Albayda J. Refractory alopecia universalis associated with dermatomyositis successfully treated with tofacitinib. Mod Rheumatol Case Rep 2022; 6:199-202. [PMID: 35253877 PMCID: PMC11132691 DOI: 10.1093/mrcr/rxac012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/26/2021] [Accepted: 02/11/2022] [Indexed: 11/13/2022]
Abstract
Dermatomyositis (DM) and alopecia areata are two diseases characterised by aberrant interferon signalling. While patchy alopecia of the scalp is a known feature of DM, alopecia universalis, which involves hair loss over the entire body, has rarely been reported in conjunction with DM. Herein, we report the case of a 30-year-old female with DM who developed refractory cutaneous disease and alopecia universalis that were successfully treated with tofacitinib. This could suggest that concomitant severe alopecia and refractory cutaneous DM may reflect a strong baseline interferon gene signature that may predict responsiveness to janus kinase inhibitors.
Collapse
Affiliation(s)
- Rochelle Castillo
- Department of Medicine, Division of Rheumatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Jemima Albayda
- Department of Medicine, Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
11
|
Accelerating clinical-scale production of BCMA CAR T cells with defined maturation stages. Mol Ther Methods Clin Dev 2022; 24:181-198. [PMID: 35118163 PMCID: PMC8791860 DOI: 10.1016/j.omtm.2021.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 12/22/2021] [Indexed: 01/04/2023]
Abstract
The advent of CAR T cells targeting CD19 or BCMA on B cell neoplasm demonstrated remarkable efficacy, but rapid relapses and primary refractoriness remains challenging. A leading cause of CAR T cell failure is their lack of expansion and limited persistence. Long-lived, self-renewing multipotent T memory stem cells (TSCM) and T central memory cells (TCM) likely sustain superior tumor regression, but their low frequencies in blood from cancer patients impose a major hurdle for clinical CAR T production. We designed a clinically compliant protocol for generating BCMA CAR T cells starting with increased TSCM/TCM cell input. A CliniMACS Prodigy process was combined with flow cytometry-based enrichment of CD62L+CD95+ T cells. Although starting with only 15% of standard T cell input, the selected TSCM/TCM material was efficiently activated and transduced with a BCMA CAR-encoding retrovirus. Cultivation in the presence of IL-7/IL-15 enabled the harvest of CAR T cells containing an increased CD4+ TSCM fraction and 70% TSCM cells amongst CD8+. Strong cell proliferation yielded cell numbers sufficient for clinical application, while effector functions were maintained. Together, adaptation of a standard CliniMACS Prodigy protocol to low input numbers resulted in efficient retroviral transduction with a high CAR T cell yield.
Collapse
|
12
|
The role of CD8 + Granzyme B + T cells in the pathogenesis of Takayasu's arteritis. Clin Rheumatol 2021; 41:167-176. [PMID: 34494213 DOI: 10.1007/s10067-021-05903-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/11/2021] [Accepted: 09/01/2021] [Indexed: 01/26/2023]
Abstract
OBJECTIVE T cell-mediated immune response plays a key role in Takayasu arteritis (TAK). Although previous studies have showed the roles of CD4+T cell and its subsets in TAK, the change of CD8+ T cell subsets remains unclear. This study investigated the role of CD8+ T cell subsets in TAK. METHODS The study consisted of 56 TA patients and 51 healthy controls. The percentages of CD8+T cells, CD8+GranzymeB+ T cells, CD8+Perforin+ T cells, and CD8+IFN-γ+ T cells in blood samples were analyzed by flow cytometry. RESULTS We found that the percentages of CD8+GranzymeB+ T cells (P = 0.030), CD8+Perforin+ T cells (P = 0.000), and CD8+IFN-γ+ T cells (P = 0.002) in CD8+T cells were higher in TAK patients compared to control group. After 6 months of treatment, the proportion of CD8+T cells in lymphocytes were significantly lower in TAK patients than the baseline assessment (P = 0.033). A lower ratio of CD8+GranzymeB+ T cells/CD8+ T cells were showed in TAK patents after treatment compared with TAK patients before treatments (P = 0.011). The change of CD8+GranzymeB+ T cells/CD8+ T cells ratio was positively correlated with the change of ITAS (r = 0.721, P = 0.002) and ITAS-A (r = 0.637, P = 0.008). Finally, the immunofluorescence staining showed the infiltration of CD8+ Granzyme B + cells in the aortic tissue of TAK patients. CONCLUSION Our results disclose that the CD8+ T lymphocytes may play a role in TAK pathogenesis. Targeting CD8+GranzymeB+ T lymphocytes or Granzyme B inhibitors could be a potential therapeutic approach for the treatment of TAK. Key Points • Our study investigated role the of CD8+ T cell subsets in TAK. • We found the percentages of CD8+GranzymeB+ T cells, CD8+Perforin+ T cells, and CD8+IFN-γ+ T cells in CD3+CD8+T cells were higher in TAK patients. • The proportion of CD8+T cells in lymphocytes and the ratio of CD8+GranzymeB+ T cells/CD8+ T cells were significantly lower in TAK patients after treatment.
Collapse
|
13
|
Lacorcia M, Bhattacharjee S, Laubhahn K, Alhamdan F, Ram M, Muschaweckh A, Potaczek DP, Kosinska A, Garn H, Protzer U, Renz H, Prazeres da Costa C. Fetomaternal immune cross talk modifies T-cell priming through sustained changes to DC function. J Allergy Clin Immunol 2021; 148:843-857.e6. [PMID: 33684437 DOI: 10.1016/j.jaci.2021.02.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 02/18/2021] [Accepted: 02/26/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Prenatal exposure to infections can modify immune development. These environmental disturbances during early life potentially alter the incidence of inflammatory disorders as well as priming of immune responses. Infection with the helminth Schistosoma mansoni is widely studied for its ability to alter immune responsiveness and is associated with variations in coinfection, allergy, and vaccine efficacy in endemic populations. OBJECTIVE Exposure to maternal schistosomiasis during early life, even without transmission of infection, can result in priming effects on offspring immune responses to bystander antigenic challenges as related to allergic responsiveness and vaccination, with this article seeking to further clarify the effects and underlying immunologic imprinting. METHODS Here, we have combined a model of chronic maternal schistosomiasis infection with a thorough analysis of subsequent offspring immune responses to allergy and vaccination models, including viral challenge and steady-state changes to immune cell compartments. RESULTS We have demonstrated that maternal schistosomiasis alters CD4+ responses during allergic sensitization and challenge in a skewed IL-4/B-cell-dominant response to antigenic challenge associated with limited inflammatory response. Beyond that, we have uncovered previously unidentified alterations to CD8+ T-cell responses during immunization that are dependent on vaccine formulation and have functional impact on the efficacy of vaccination against viral infection in a murine hepatitis B virus model. CONCLUSION In addition to steady-state modifications to CD4+ T-cell polarization and B-cell priming, we have traced these modified CD8+ responses to an altered dendritic cell phenotype sustained into adulthood, providing evidence for complex priming effects imparted by infection via fetomaternal cross talk.
Collapse
Affiliation(s)
- Matthew Lacorcia
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Sonakshi Bhattacharjee
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Kristina Laubhahn
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany; Pediatric Allergology, Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University Munich, Munich, Germany; German Center for Lung Research, Ludwig Maximilian University Munich, Munich, Germany
| | - Fahd Alhamdan
- Biochemical Pharmacological Center, Translational Inflammation Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Marija Ram
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Andreas Muschaweckh
- Department of Neurology, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Daniel P Potaczek
- Biochemical Pharmacological Center, Translational Inflammation Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Anna Kosinska
- Institute for Virology Technical University of Munich, Munich, Germany
| | - Holger Garn
- Biochemical Pharmacological Center, Translational Inflammation Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Ulrike Protzer
- Institute for Virology Technical University of Munich, Munich, Germany
| | - Harald Renz
- Biochemical Pharmacological Center, Translational Inflammation Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Clarissa Prazeres da Costa
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany.
| |
Collapse
|
14
|
Melgaço JG, Azamor T, Silva AMV, Linhares JHR, dos Santos TP, Mendes YS, de Lima SMB, Fernandes CB, da Silva J, de Souza AF, Tubarão LN, Brito e Cunha D, Pereira TBS, Menezes CEL, Miranda MD, Matos AR, Caetano BC, Martins JSCC, Calvo TL, Rodrigues NF, Sacramento CQ, Siqueira MM, Moraes MO, Missailidis S, Neves PCC, Ano Bom APD. Two-Step In Vitro Model to Evaluate the Cellular Immune Response to SARS-CoV-2. Cells 2021; 10:2206. [PMID: 34571855 PMCID: PMC8465121 DOI: 10.3390/cells10092206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 01/08/2023] Open
Abstract
The cellular immune response plays an important role in COVID-19, caused by SARS-CoV-2. This feature makes use of in vitro models' useful tools to evaluate vaccines and biopharmaceutical effects. Here, we developed a two-step model to evaluate the cellular immune response after SARS-CoV-2 infection-induced or spike protein stimulation in peripheral blood mononuclear cells (PBMC) from both unexposed and COVID-19 (primo-infected) individuals (Step1). Moreover, the supernatants of these cultures were used to evaluate its effects on lung cell lines (A549) (Step2). When PBMC from the unexposed were infected by SARS-CoV-2, cytotoxic natural killer and nonclassical monocytes expressing inflammatory cytokines genes were raised. The supernatant of these cells can induce apoptosis of A549 cells (mock vs. Step2 [mean]: 6.4% × 17.7%). Meanwhile, PBMCs from primo-infected presented their memory CD4+ T cells activated with a high production of IFNG and antiviral genes. Supernatant from past COVID-19 subjects contributed to reduce apoptosis (mock vs. Step2 [ratio]: 7.2 × 1.4) and to elevate the antiviral activity (iNOS) of A549 cells (mock vs. Step2 [mean]: 31.5% × 55.7%). Our findings showed features of immune primary cells and lung cell lines response after SARS-CoV-2 or spike protein stimulation that can be used as an in vitro model to study the immunity effects after SARS-CoV-2 antigen exposure.
Collapse
Affiliation(s)
- Juliana G. Melgaço
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| | - Tamiris Azamor
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| | - Andréa M. V. Silva
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| | - José Henrique R. Linhares
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| | - Tiago P. dos Santos
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| | - Ygara S. Mendes
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| | - Sheila M. B. de Lima
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| | - Camilla Bayma Fernandes
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| | - Jane da Silva
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| | - Alessandro F. de Souza
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| | - Luciana N. Tubarão
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| | - Danielle Brito e Cunha
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| | - Tamires B. S. Pereira
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| | - Catarina E. L. Menezes
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| | - Milene D. Miranda
- Laboratório de Vírus Respiratório e do Sarampo, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (M.D.M.); (A.R.M.); (B.C.C.); (J.S.C.C.M.); (M.M.S.)
| | - Aline R. Matos
- Laboratório de Vírus Respiratório e do Sarampo, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (M.D.M.); (A.R.M.); (B.C.C.); (J.S.C.C.M.); (M.M.S.)
| | - Braulia C. Caetano
- Laboratório de Vírus Respiratório e do Sarampo, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (M.D.M.); (A.R.M.); (B.C.C.); (J.S.C.C.M.); (M.M.S.)
| | - Jéssica S. C. C. Martins
- Laboratório de Vírus Respiratório e do Sarampo, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (M.D.M.); (A.R.M.); (B.C.C.); (J.S.C.C.M.); (M.M.S.)
| | - Thyago L. Calvo
- Laboratório de Hanseníase, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.L.C.); (M.O.M.)
| | - Natalia F. Rodrigues
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (N.F.R.); (C.Q.S.)
- Centro de Desenvolvimento Tecnológico em Saúde, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil
| | - Carolina Q. Sacramento
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (N.F.R.); (C.Q.S.)
- Centro de Desenvolvimento Tecnológico em Saúde, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil
| | - Marilda M. Siqueira
- Laboratório de Vírus Respiratório e do Sarampo, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (M.D.M.); (A.R.M.); (B.C.C.); (J.S.C.C.M.); (M.M.S.)
| | - Milton O. Moraes
- Laboratório de Hanseníase, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.L.C.); (M.O.M.)
| | - Sotiris Missailidis
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| | - Patrícia C. C. Neves
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| | - Ana Paula D. Ano Bom
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil; (T.A.); (A.M.V.S.); (J.H.R.L.); (T.P.d.S.); (Y.S.M.); (S.M.B.d.L.); (C.B.F.); (J.d.S.); (A.F.d.S.); (L.N.T.); (D.B.e.C.); (T.B.S.P.); (C.E.L.M.); (S.M.); (P.C.C.N.); (A.P.D.A.B.)
| |
Collapse
|
15
|
Chen M, Brackett CM, Burdelya LG, Punnanitinont A, Patnaik SK, Matsuzaki J, Odunsi AO, Gudkov AV, Singh AK, Repasky EA, Gurova KV. Stimulation of an anti-tumor immune response with "chromatin-damaging" therapy. Cancer Immunol Immunother 2021; 70:2073-2086. [PMID: 33439292 PMCID: PMC8726059 DOI: 10.1007/s00262-020-02846-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/29/2020] [Indexed: 02/08/2023]
Abstract
Curaxins are small molecules that bind genomic DNA and interfere with DNA-histone interactions leading to the loss of histones and decondensation of chromatin. We named this phenomenon 'chromatin damage'. Curaxins demonstrated anti-cancer activity in multiple pre-clinical tumor models. Here, we present data which reveals, for the first time, a role for the immune system in the anti-cancer effects of curaxins. Using the lead curaxin, CBL0137, we observed elevated expression of several group of genes in CBL0137-treated tumor cells including interferon sensitive genes, MHC molecules, some embryo-specific antigens suggesting that CBL0137 increases tumor cell immunogenicity and improves recognition of tumor cells by the immune system. In support of this, we found that the anti-tumor activity of CBL0137 was reduced in immune deficient SCID mice when compared to immune competent mice. Anti-tumor activity of CBL0137 was abrogated in CD8+ T cell depleted mice but only partially lost when natural killer or CD4+ T cells were depleted. Further support for a key role for the immune system in the anti-tumor activity of CBL0137 is evidenced by an increased antigen-specific effector CD8+ T cell and NK cell response, and an increased ratio of effector T cells to Tregs in the tumor and spleen. CBL0137 also elevated the number of CXCR3-expressing CTLs in the tumor and the level of interferon-γ-inducible protein 10 (IP-10) in serum, suggesting IP-10/CXCR3 controls CBL0137-elicited recruitment of effector CTLs to tumors. Our collective data underscores a previously unrecognized role for both innate and adaptive immunity in the anti-tumor activity of curaxins.
Collapse
Affiliation(s)
- Minhui Chen
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Craig M Brackett
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Lyudmila G Burdelya
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Achamaporn Punnanitinont
- Cancer for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Santosh K Patnaik
- Cancer for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Junko Matsuzaki
- Cancer for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Adekunle O Odunsi
- Cancer for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Andrei V Gudkov
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Anurag K Singh
- Department of Radiation Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA.
| | - Katerina V Gurova
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Sts, Buffalo, NY, 14263, USA.
| |
Collapse
|
16
|
Demers-Mathieu V, Mathijssen GB, DaPra C, Medo E. The effects of probiotic supplementation on the gene expressions of immune cell surface markers and levels of antibodies and pro-inflammatory cytokines in human milk. J Perinatol 2021; 41:1083-1091. [PMID: 33208844 DOI: 10.1038/s41372-020-00875-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/07/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE This study investigated the impact of probiotic supplementation on the gene expressions of cluster of differentiation (CD) as cell markers and the concentrations of antibodies and cytokines in human milk. STUDY DESIGN Gene expressions of CD28, CD19, and CD38 were determined in milk from 15 women ingesting daily probiotics (from Greek yogurt) and 12 women who do not consume probiotics. Concentrations of antibodies and cytokines were measured using ELISA. RESULTS Gene expression of CD28 tended to be higher in milk from mothers ingesting daily probiotics than mothers who did not take probiotics. Interleukin-6 (IL-6) concentration in milk was higher in mothers ingesting probiotics than those who do not consume probiotics. The increase of IL-6 level in human milk was positively correlated with total IgA and IgG concentrations. CONCLUSIONS Probiotic supplementation could enhance the secretion of IL-6 in human milk. Human milk IL-6 may improve neonatal immunity due to its stimulation of total IgA and IgG.
Collapse
Affiliation(s)
- Veronique Demers-Mathieu
- Department of Neonatal Immunology and Microbiology, Medolac Laboratories A Public Benefit Corporation, Boulder City, NV, USA.
| | - Gabrielle B Mathijssen
- Department of Neonatal Immunology and Microbiology, Medolac Laboratories A Public Benefit Corporation, Boulder City, NV, USA
| | - Ciera DaPra
- Department of Neonatal Immunology and Microbiology, Medolac Laboratories A Public Benefit Corporation, Boulder City, NV, USA
| | - Elena Medo
- Department of Neonatal Immunology and Microbiology, Medolac Laboratories A Public Benefit Corporation, Boulder City, NV, USA
| |
Collapse
|
17
|
Toor SM, Sasidharan Nair V, Saleh R, Taha RZ, Murshed K, Al-Dhaheri M, Khawar M, Ahmed AA, Kurer MA, Abu Nada M, Elkord E. Transcriptome of Tumor-Infiltrating T Cells in Colorectal Cancer Patients Uncovered a Unique Gene Signature in CD4 + T Cells Associated with Poor Disease-Specific Survival. Vaccines (Basel) 2021; 9:vaccines9040334. [PMID: 33916009 PMCID: PMC8065799 DOI: 10.3390/vaccines9040334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/15/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is influenced by infiltration of immune cell populations in the tumor microenvironment. While elevated levels of cytotoxic T cells are associated with improved prognosis, limited studies have reported associations between CD4+ T cells and disease outcomes. We recently performed transcriptomic profiling and comparative analyses of sorted CD4+ and CD8+ tumor-infiltrating lymphocytes (TILs) from bulk tumors of CRC patients with varying disease stages. In this study, we compared the transcriptomes of CD4+ with CD8+ TILs. Functional annotation pathway analyses revealed the downregulation of inflammatory response-related genes, while T cell activation and angiogenesis-related genes were upregulated in CD4+ TILs. The top 200 deregulated genes in CD4+ TILs were aligned with the cancer genome atlas (TCGA) CRC dataset to identify a unique gene signature associated with poor prognosis. Moreover, 69 upregulated and 20 downregulated genes showed similar trends of up/downregulation in the TCGA dataset and were used to calculate "poor prognosis score" (ppScore), which was significantly associated with disease-specific survival. High ppScore patients showed lower expression of Treg-, Th1-, and Th17-related genes, and higher expression of Th2-related genes. Our data highlight the significance of T cells within the TME and identify a unique candidate prognostic gene signature for CD4+ TILs in CRC patients.
Collapse
Affiliation(s)
- Salman M. Toor
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), P.O. Box 34110 Doha, Qatar; (S.M.T.); (V.S.N.); (R.S.); (R.Z.T.)
| | - Varun Sasidharan Nair
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), P.O. Box 34110 Doha, Qatar; (S.M.T.); (V.S.N.); (R.S.); (R.Z.T.)
| | - Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), P.O. Box 34110 Doha, Qatar; (S.M.T.); (V.S.N.); (R.S.); (R.Z.T.)
| | - Rowaida Z. Taha
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), P.O. Box 34110 Doha, Qatar; (S.M.T.); (V.S.N.); (R.S.); (R.Z.T.)
| | - Khaled Murshed
- Department of Pathology, Hamad Medical Corporation, P.O. Box 3050 Doha, Qatar;
| | - Mahmood Al-Dhaheri
- Department of Surgery, Hamad Medical Corporation, P.O. Box 3050 Doha, Qatar; (M.A.-D.); (M.K.); (A.A.A.); (M.A.K.); (M.A.N.)
| | - Mahwish Khawar
- Department of Surgery, Hamad Medical Corporation, P.O. Box 3050 Doha, Qatar; (M.A.-D.); (M.K.); (A.A.A.); (M.A.K.); (M.A.N.)
| | - Ayman A. Ahmed
- Department of Surgery, Hamad Medical Corporation, P.O. Box 3050 Doha, Qatar; (M.A.-D.); (M.K.); (A.A.A.); (M.A.K.); (M.A.N.)
| | - Mohamed A. Kurer
- Department of Surgery, Hamad Medical Corporation, P.O. Box 3050 Doha, Qatar; (M.A.-D.); (M.K.); (A.A.A.); (M.A.K.); (M.A.N.)
| | - Mohamed Abu Nada
- Department of Surgery, Hamad Medical Corporation, P.O. Box 3050 Doha, Qatar; (M.A.-D.); (M.K.); (A.A.A.); (M.A.K.); (M.A.N.)
| | - Eyad Elkord
- Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, Manchester M5 4WT, UK
- Correspondence: ; Tel.: +44-161-295-5736
| |
Collapse
|
18
|
Huang J, Xiao Z, An Y, Han S, Wu W, Wang Y, Guo Y, Shuai X. Nanodrug with dual-sensitivity to tumor microenvironment for immuno-sonodynamic anti-cancer therapy. Biomaterials 2021; 269:120636. [PMID: 33453632 DOI: 10.1016/j.biomaterials.2020.120636] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/17/2020] [Accepted: 12/25/2020] [Indexed: 12/17/2022]
Abstract
Although a combination with photodynamic therapy (PDT) is a potential means to improve the immune checkpoint blockade (ICB)-based anticancer immunotherapy, this strategy is subjected to the extremely poor light penetration in melanoma. Herein, we develop a lipid (LP)-based micellar nanocarrier encapsulating sonosensitizer chlorin e6 (Ce6) in the core, conjugating anti-PD-L1 antibody (aPD-L1) to the interlayer through MMP-2-cleavable peptide, and bearing a PEG coating sheddable at low pH value (≈6.5) of tumor microenvironment. The unique nanocarrier design allows a tumor-targeting delivery to activate the anti-tumor immunity and meanwhile to reduce immune-related adverse effects (irAEs). Moreover, a sonodynamic therapy (SDT) is triggerable by using ultrasonic insonation to produce tumor-killing reactive oxygen species (ROS), thereby bypassing the poor light penetration which restricts PDT in melanoma. A combination of SDT with aPD-L1 immunotherapy effectively promotes tumor infiltration and activation of cytotoxic T cells, which resulted in robust anti-cancer immunity and long-term immune memory to effectively suppress melanoma growth and postoperative recurrence. This strategy for tumor-targeting codelivery of immune checkpoint inhibitors and SDT agents could be readily extended to other tumor types for better immunotherapeutic outcome and reduced irAEs.
Collapse
Affiliation(s)
- Jinsheng Huang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Zecong Xiao
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yongcheng An
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510275, China
| | - Shisong Han
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510275, China
| | - Wei Wu
- Department of Medical Oncology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Yong Wang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China.
| | - Yu Guo
- Department of General Surgery, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510275, China.
| | - Xintao Shuai
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China; PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China.
| |
Collapse
|
19
|
Gangisetty O, Palagani A, Sarkar DK. Transgenerational inheritance of fetal alcohol exposure adverse effects on immune gene interferon-ϒ. Clin Epigenetics 2020; 12:70. [PMID: 32448218 PMCID: PMC7245772 DOI: 10.1186/s13148-020-00859-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 05/12/2020] [Indexed: 11/16/2022] Open
Abstract
Background Alcohol exposures in utero have been shown to alter immune system functions in the offspring which persists into adulthood. However, it is not apparent why the in utero alcohol effect on the immune system persists into adulthood of fetal alcohol-exposed offspring. The objective of this study was to determine the long-term effects of fetal alcohol exposure on the production of interferon-ϒ (IFN-ϒ), a cytokine known to regulate both innate and adaptive immunity. Methods Isogenic Fisher 344 rats were bred to produce pregnant dams, which were fed with a liquid diet containing 6.7% alcohol between gestation days 7 and 21 and pair-fed with an isocaloric liquid diet or fed ad libitum with rat chow; their male and female offspring were used for the study. F1-F3 generation rats were used when they were 2 to 3 months old. Fetal alcohol exposure effects on the Ifn-ɣ gene was determined by measuring the gene promoter methylation and mRNA and protein expression in the spleen. Additionally, transgenerational studies were conducted to evaluate the germline-transmitted effects of fetal alcohol exposure on the Ifn-ɣ gene. Results Fetal alcohol exposure reduced the expression of Ifn-ɣ mRNA and IFN-ϒ protein while it increased the proximal promoter methylation of the Ifn-ɣ gene in both male and female offspring during the adult period. Transgenerational studies revealed that the reduced levels of Ifn-ɣ expression and increased levels of its promoter methylation persisted only in F2 and F3 generation males derived from the male germ line. Conclusion Overall, these findings provide the evidence that fetal alcohol exposures produce an epigenetic mark on the Ifn-ɣ gene that passes through multiple generations via the male germ line. These data provide the first evidence that the male germ line transmits fetal alcohol exposure's adverse effects on the immune system.
Collapse
Affiliation(s)
- Omkaram Gangisetty
- Rutgers Endocrine Research Program, Department of Animal Sciences, Rutgers University, 67 Poultry Farm Lane, New Brunswick, NJ, 08901, USA
| | - Ajay Palagani
- Rutgers Endocrine Research Program, Department of Animal Sciences, Rutgers University, 67 Poultry Farm Lane, New Brunswick, NJ, 08901, USA
| | - Dipak K Sarkar
- Rutgers Endocrine Research Program, Department of Animal Sciences, Rutgers University, 67 Poultry Farm Lane, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
20
|
de Araújo-Souza PS, Hanschke SCH, Nardy AFFR, Sécca C, Oliveira-Vieira B, Silva KL, Soares-Lima SC, Viola JPB. Differential interferon-γ production by naive and memory-like CD8 T cells. J Leukoc Biol 2020; 108:1329-1337. [PMID: 32421902 DOI: 10.1002/jlb.2ab0420-646r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/07/2020] [Accepted: 04/27/2020] [Indexed: 11/08/2022] Open
Abstract
CD8 T cells play a crucial role in immune responses to virus infections and tumors. Naïve CD8 T lymphocytes after TCR stimulation undergo differentiation into CTLs and memory cells, which are essential sources of IFN-γ. We investigated IFN-γ production by CD8 T cell subsets found in nonimmune mice. A minor fraction of in vitro TCR-stimulated CD8 T cells produce IFN-γ, and it is regulated at the transcriptional level. Antigen inexperienced C57BL/6 mice present the coexistence of 2 populations. The main population exhibits a CD44low CD122low profile, which is compatible with naïve lymphocytes. The minor expresses a phenotype of immunologic memory, CD44hi CD122hi . Both subsets are able to produce IL-2 in response to TCR activation, but only the memory-like population is responsible for IFN-γ production. Similar to memory CD8 T cells, CD44hi CD8+ T cells also present a higher level of the transcriptional factor Eomes and a lower level of T-bet (Tbx21) mRNA than CD44low CD8+ T cells. The presence of the CD44hi CD8+ T cell population in nonimmune OT-I transgenic mice reveals that the population is generated independently of antigenic stimulation. CpG methylation is an efficient epigenetic mechanism for gene silencing. DNA methylation at posttranscriptional CpG sites in the Ifng promoter is higher in CD44low CD8+ T cells than in CD44hi CD8+ T cells. Thus, memory-like CD8 T cells have a distinct epigenetic pattern in the Ifng promoter and can rapidly produce IFN-γ in response to TCR stimulation.
Collapse
Affiliation(s)
- Patrícia S de Araújo-Souza
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil.,Department of Immunobiology, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Steffi C H Hanschke
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Ana Flavia F R Nardy
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Cristiane Sécca
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Barbara Oliveira-Vieira
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Karina L Silva
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Sheila C Soares-Lima
- Program of Molecular Carcinogenesis, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - João P B Viola
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
21
|
Ghavidel AA, Shiari R, Hassan-Zadeh V, Farivar S. The expression of DNMTs is dramatically decreased in peripheral blood mononuclear cells of male patients with juvenile idiopathic arthritis. Allergol Immunopathol (Madr) 2020; 48:182-186. [PMID: 31901404 DOI: 10.1016/j.aller.2019.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 08/07/2019] [Accepted: 08/28/2019] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Juvenile idiopathic arthritis (JIA) is an autoimmune rheumatic disease, which affects primarily the joints in children under 16 years old. The etiology of JIA is yet unknown but research has shown that JIA is a multifactorial disease implicating several genes and environmental factors. Environmental factors affect immune cells via epigenetic mechanisms. One of the most important epigenetic mechanisms is DNA methylation catalyzed by DNA methyltransferases (DNMTs) and usually associated with gene silencing. In this study, we analyzed the expression of three DNA methyltransferases namely DNMT1, DNMT3a and DNMT3b in peripheral blood mononuclear cells (PBMCs) of patients with JIA and compared it with the expression of these genes in healthy young individuals. MATERIALS AND METHODS Peripheral blood mononuclear cells of 28 JIA patients and 28 healthy controls were isolated. Total RNA was extracted, cDNA was synthesized and the transcript levels of DNMTs were analyzed by quantitative PCR. RESULTS Analysis of DNMT1, DNMT3a and DNMT3b relative gene expression in PBMCs of JIA patients and control individuals shows that the expression of DNMT1 and DNMT3a is reduced significantly by 7 folds and 5.5 folds, respectively, in JIA patients compared to healthy controls. Furthermore, the expression of all three DNMTs were significantly and drastically reduced in young affected males compared to healthy males. CONCLUSION This study shows that the expression of DNMTs is reduced in JIA patients and this reduction is severe in male JIA patients.
Collapse
Affiliation(s)
- Afshin Abdi Ghavidel
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University G.C., Tehran, Iran; Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Shiari
- Department of Pediatrics, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Mofid Children's Hospital, Pediatrics Infectious Research Center (PIRC), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahideh Hassan-Zadeh
- Department of Cell and Molecular Biology, School of Biology, Faculty of Science, University of Tehran, Tehran, Iran
| | - Shirin Farivar
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University G.C., Tehran, Iran.
| |
Collapse
|
22
|
Barili V, Fisicaro P, Montanini B, Acerbi G, Filippi A, Forleo G, Romualdi C, Ferracin M, Guerrieri F, Pedrazzi G, Boni C, Rossi M, Vecchi A, Penna A, Zecca A, Mori C, Orlandini A, Negri E, Pesci M, Massari M, Missale G, Levrero M, Ottonello S, Ferrari C. Targeting p53 and histone methyltransferases restores exhausted CD8+ T cells in HCV infection. Nat Commun 2020; 11:604. [PMID: 32001678 PMCID: PMC6992697 DOI: 10.1038/s41467-019-14137-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 12/11/2019] [Indexed: 12/22/2022] Open
Abstract
Hepatitis C virus infection (HCV) represents a unique model to characterize, from early to late stages of infection, the T cell differentiation process leading to exhaustion of human CD8+ T cells. Here we show that in early HCV infection, exhaustion-committed virus-specific CD8+ T cells display a marked upregulation of transcription associated with impaired glycolytic and mitochondrial functions, that are linked to enhanced ataxia-telangiectasia mutated (ATM) and p53 signaling. After evolution to chronic infection, exhaustion of HCV-specific T cell responses is instead characterized by a broad gene downregulation associated with a wide metabolic and anti-viral function impairment, which can be rescued by histone methyltransferase inhibitors. These results have implications not only for treatment of HCV-positive patients not responding to last-generation antivirals, but also for other chronic pathologies associated with T cell dysfunction, including cancer.
Collapse
Affiliation(s)
- Valeria Barili
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Unit of Infectious Diseases and Hepatology, Laboratory of Viral Immunopathology, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Paola Fisicaro
- Unit of Infectious Diseases and Hepatology, Laboratory of Viral Immunopathology, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Barbara Montanini
- Biomolecular, Genomic and Biocomputational Sciences Unit, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy.,Biopharmanet-Tec, University of Parma, Parma, Italy
| | - Greta Acerbi
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Unit of Infectious Diseases and Hepatology, Laboratory of Viral Immunopathology, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Anita Filippi
- Unit of Infectious Diseases and Hepatology, Laboratory of Viral Immunopathology, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Giovanna Forleo
- Unit of Infectious Diseases and Hepatology, Laboratory of Viral Immunopathology, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | | | - Manuela Ferracin
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Bologna, Italy
| | | | - Giuseppe Pedrazzi
- Unit of Neuroscience, Department of Medicine and Surgery, Robust Statistics Academy (Ro.S.A.), University of Parma, Parma, Italy
| | - Carolina Boni
- Unit of Infectious Diseases and Hepatology, Laboratory of Viral Immunopathology, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Marzia Rossi
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Unit of Infectious Diseases and Hepatology, Laboratory of Viral Immunopathology, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Andrea Vecchi
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Unit of Infectious Diseases and Hepatology, Laboratory of Viral Immunopathology, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Amalia Penna
- Unit of Infectious Diseases and Hepatology, Laboratory of Viral Immunopathology, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Alessandra Zecca
- Unit of Infectious Diseases and Hepatology, Laboratory of Viral Immunopathology, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Cristina Mori
- Unit of Infectious Diseases and Hepatology, Laboratory of Viral Immunopathology, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Alessandra Orlandini
- Unit of Infectious Diseases and Hepatology, Laboratory of Viral Immunopathology, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Elisa Negri
- Unit of Infectious Diseases and Hepatology, Laboratory of Viral Immunopathology, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Marco Pesci
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Unit of Infectious Diseases and Hepatology, Laboratory of Viral Immunopathology, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Marco Massari
- Unit of Infectious Diseases, IRCCS-Azienda Ospedaliera S. Maria Nuova, Reggio Emilia, Italy
| | - Gabriele Missale
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Unit of Infectious Diseases and Hepatology, Laboratory of Viral Immunopathology, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Massimo Levrero
- Cancer Research Center of Lyon (CRCL)-INSERM U1052, Lyon, France.,Université Claude Bernard Lyon 1, Service d'Hepatologie et Gastroenterologie Hopital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France.,Center for Life Nano Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Simone Ottonello
- Biomolecular, Genomic and Biocomputational Sciences Unit, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy.,Biopharmanet-Tec, University of Parma, Parma, Italy
| | - Carlo Ferrari
- Department of Medicine and Surgery, University of Parma, Parma, Italy. .,Unit of Infectious Diseases and Hepatology, Laboratory of Viral Immunopathology, Azienda Ospedaliero-Universitaria of Parma, Parma, Italy.
| |
Collapse
|
23
|
Transcriptome Analysis and Emerging Driver Identification of CD8+ T Cells in Patients with Vitiligo. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2503924. [PMID: 31885781 PMCID: PMC6899274 DOI: 10.1155/2019/2503924] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/09/2019] [Indexed: 12/12/2022]
Abstract
Activated CD8+ T cells play important roles in the pathogenesis of vitiligo. However, driving factors about the activation and migration of CD8+ T cells remain obscure. In this study, we aim to identify differentially expressed genes (DEGs) and uncover potential factors that drive the disease in melanocyte-specific CD8+ T cells in vitiligo. A total of 1147 DEGs were found through transcriptome sequencing in CD8+ T cells from lesional skin of vitiligo patients and normal controls. Based on KEGG pathway enrichment analysis and PPI, 16 upregulated and 23 downregulated genes were identified. Ultimately, 3 genes were figured out after RT-qPCR verification. The mRNA and protein expression levels of PIK3CB, HIF-1α, and F2RL1 were all elevated in CD8+ T cells from peripheral blood in vitiligo. HIF-1α and PIK3CB were significantly increased in lesional skin of vitiligo. Two CpG sites of the HIF-1α promoter were hypomethylated in vitiligo CD8+ T cells. In conclusion, HIF-1α, F2RL1, and PIK3CB may act as novel drivers for vitiligo, which are all closely associated with reactive oxygen species and possibly contribute to the activation and/or migration of melanocyte-specific CD8+ T cells in vitiligo. In addition, we uncovered a potential role for DNA hypomethylation of HIF-1α in CD8+ T cells of vitiligo.
Collapse
|
24
|
Deng Z, Zhou W, Sun J, Li C, Zhong B, Lai K. IFN-γ Enhances the Cough Reflex Sensitivity via Calcium Influx in Vagal Sensory Neurons. Am J Respir Crit Care Med 2019; 198:868-879. [PMID: 29672123 DOI: 10.1164/rccm.201709-1813oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Cough hypersensitivity syndrome is often triggered by a viral infection. The viral infection might trigger cough hypersensitivity via increasing the release of IFN-γ from T lymphocytes in the lung. OBJECTIVES To investigate effects of IFN-γ on the vagal sensory neurons and the cough reflex. METHODS Effects of IFN-γ on the cough reflex were investigated in guinea pigs. Cellular immunofluorescence imaging, calcium imaging, and patch clamp techniques were used to study effects of IFN-γ in primary cultured rat vagal sensory neurons. MEASUREMENTS AND MAIN RESULTS Intratracheal instillation of IFN-γ enhanced the cough response to citric acid in vivo. IFN-γ significantly increased levels of phosphorylated signal transducer and activator of transcription-1 but not phosphorylated transient receptor potential vanilloid 1 in vitro. Not only did IFN-γ enhance the response of neurons to capsaicin and electric stimulation, but also it directly induced Ca2+ influx, membrane depolarization, and action potentials in neurons via the Janus kinase, protein kinase A, and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid pathways. However, IFN-γ did not elicit Ca2+ release from the endoplasmic reticulum via the phospholipase C pathway. Although IFN-γ-induced action potentials were suppressed by Ca2+ influx inhibitors, IFN-γ-induced Ca2+ influx was not altered by an inhibitor of rapid sodium channels. CONCLUSIONS The membrane potential in vagal sensory neurons may be depolarized by IFN-γ-induced Ca2+ influx. The depolarization of membrane potentials may enhance the cough reflex sensitivity and cause action potentials. IFN-γ may be a new target for treating cough hypersensitivity syndrome and postviral cough.
Collapse
Affiliation(s)
- Zheng Deng
- 1 State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,2 School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China; and
| | - Wenliang Zhou
- 3 School of Life Science, Sun Yat-sen University, Guangzhou, China
| | - Jiayang Sun
- 1 State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chenhui Li
- 1 State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bonian Zhong
- 1 State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kefang Lai
- 1 State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
25
|
Vigano S, Alatzoglou D, Irving M, Ménétrier-Caux C, Caux C, Romero P, Coukos G. Targeting Adenosine in Cancer Immunotherapy to Enhance T-Cell Function. Front Immunol 2019; 10:925. [PMID: 31244820 PMCID: PMC6562565 DOI: 10.3389/fimmu.2019.00925] [Citation(s) in RCA: 296] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 04/10/2019] [Indexed: 12/13/2022] Open
Abstract
T cells play a critical role in cancer control, but a range of potent immunosuppressive mechanisms can be upregulated in the tumor microenvironment (TME) to abrogate their activity. While various immunotherapies (IMTs) aiming at re-invigorating the T-cell-mediated anti-tumor response, such as immune checkpoint blockade (ICB), and the adoptive cell transfer (ACT) of natural or gene-engineered ex vivo expanded tumor-specific T cells, have led to unprecedented clinical responses, only a small proportion of cancer patients benefit from these treatments. Important research efforts are thus underway to identify biomarkers of response, as well as to develop personalized combinatorial approaches that can target other inhibitory mechanisms at play in the TME. In recent years, adenosinergic signaling has emerged as a powerful immuno-metabolic checkpoint in tumors. Like several other barriers in the TME, such as the PD-1/PDL-1 axis, CTLA-4, and indoleamine 2,3-dioxygenase (IDO-1), adenosine plays important physiologic roles, but has been co-opted by tumors to promote their growth and impair immunity. Several agents counteracting the adenosine axis have been developed, and pre-clinical studies have demonstrated important anti-tumor activity, alone and in combination with other IMTs including ICB and ACT. Here we review the regulation of adenosine levels and mechanisms by which it promotes tumor growth and broadly suppresses protective immunity, with extra focus on the attenuation of T cell function. Finally, we present an overview of promising pre-clinical and clinical approaches being explored for blocking the adenosine axis for enhanced control of solid tumors.
Collapse
Affiliation(s)
- Selena Vigano
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Dimitrios Alatzoglou
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Melita Irving
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Christine Ménétrier-Caux
- Department of Immunology Virology and Inflammation, INSERM 1052, CNRS 5286, Léon Bérard Cancer Center, Cancer Research Center of Lyon, University of Lyon, University Claude Bernard Lyon 1, Lyon, France
| | - Christophe Caux
- Department of Immunology Virology and Inflammation, INSERM 1052, CNRS 5286, Léon Bérard Cancer Center, Cancer Research Center of Lyon, University of Lyon, University Claude Bernard Lyon 1, Lyon, France
| | - Pedro Romero
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
26
|
Yun S, Park Y, Moon S, Ahn S, Lee K, Park HJ, Lee HS, Choe G, Lee KS. Clinicopathological and prognostic significance of programmed death ligand 1 expression in Korean melanoma patients. J Cancer 2019; 10:3070-3078. [PMID: 31281485 PMCID: PMC6590033 DOI: 10.7150/jca.30573] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 05/03/2019] [Indexed: 12/22/2022] Open
Abstract
Programmed death ligand 1 (PD-L1) expression provides significant value to predict prognosis and response following immunotherapy in several types of cancers. However, its clinicopathological and prognostic significance in melanoma remains unclear. PD-L1 and the number of tumor infiltrating lymphocytes (TILs) were investigated in 63 Korean patients with melanoma based on the melanoma scoring system. We also compared the results using the PD-L1 antibodies—22C3 and E1L3N clones. In addition, BRAF gene mutation was detected using anti-BRAF antibody and real-time polymerase chain reaction. Overall, 29 (46.0%), 16 (25.4%), and 18 (28.6%) patients exhibited the acral lentiginous type, nodular type, and other histological subtypes of melanoma, respectively. PD-L1 expression was detected in 37 (58.7%) cases and was closely associated with a CD8+TILhigh phenotype (P < 0.001). Combined survival analysis depending on PD-L1 and CD8+TILs status showed that the PD-L1-/CD8+TILhigh group demonstrated the best survival outcome, whereas patients with PD-L1+/CD8+ TILlow showed the worst prognosis (P = 0.039). However, PD-L1+/CD8+ TILlow was not an independent prognostic factor. The 22C3 and E1L3N clones showed a high concordance rate (kappa value, 0.799). BRAF mutation status was not correlated with PD-L1 expression. We suggest that evaluation of the combined status of PD-L1 and TIL might be useful to predict the survival of patients with melanoma.
Collapse
Affiliation(s)
- Sumi Yun
- Department of Diagnostic Pathology, Samkwang Medical Laboratories, Seoul, Republic of Korea
| | - Yujun Park
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Seyoung Moon
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Soomin Ahn
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kyoungyul Lee
- Department of Pathology, Kangwon National University Hospital, Chuncheon-Si, Kangwon-Do, Republic of Korea
| | - Hyo Jin Park
- Department of Pathology, Sheikh Khalifa Specialty Hospital, Ras al Khaimah, United Arab Emirates
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Gheeyoung Choe
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kyu Sang Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
27
|
Deng Q, Luo Y, Chang C, Wu H, Ding Y, Xiao R. The Emerging Epigenetic Role of CD8+T Cells in Autoimmune Diseases: A Systematic Review. Front Immunol 2019; 10:856. [PMID: 31057561 PMCID: PMC6482221 DOI: 10.3389/fimmu.2019.00856] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 04/02/2019] [Indexed: 12/19/2022] Open
Abstract
Autoimmune diseases are usually complex and multifactorial, characterized by aberrant production of autoreactive immune cells and/or autoantibodies against healthy cells and tissues. However, the pathogenesis of autoimmune diseases has not been clearly elucidated. The activation, differentiation, and development of CD8+ T cells can be affected by numerous inflammatory cytokines, transcription factors, and chemokines. In recent years, epigenetic modifications have been shown to play an important role in the fate of CD8+ T cells. The discovery of these modifications that contribute to the activation or suppression of CD8+ cells has been concurrent with the increasing evidence that CD8+ T cells play a role in autoimmunity. These relationships have been studied in various autoimmune diseases, including multiple sclerosis (MS), systemic sclerosis (SSc), type 1 diabetes (T1D), Grave's disease (GD), systemic lupus erythematosus (SLE), aplastic anemia (AA), and vitiligo. In each of these diseases, genes that play a role in the proliferation or activation of CD8+ T cells have been found to be affected by epigenetic modifications. Various cytokines, transcription factors, and other regulatory molecules have been found to be differentially methylated in CD8+ T cells in autoimmune diseases. These genes are involved in T cell regulation, including interferons, interleukin (IL),tumor necrosis factor (TNF), as well as linker for activation of T cells (LAT), cytotoxic T-lymphocyte–associated antigen 4 (CTLA4), and adapter proteins. MiRNAs also play a role in the pathogenesis of these diseases and several known miRNAs that are involved in these diseases have also been shown to play a role in CD8+ regulation.
Collapse
Affiliation(s)
- Qiancheng Deng
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yangyang Luo
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Dermatology, Hunan Children's Hospital, Changsha, China
| | - Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Haijing Wu
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yan Ding
- Department of Dermatology, Hainan Provincial Dermatology Disease Hospital, Haikou, China
| | - Rong Xiao
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
28
|
Kusumoto Y, Okuyama H, Shibata T, Konno K, Takemoto Y, Maekawa D, Kononaga T, Ishii T, Akashi-Takamura S, Saitoh SI, Ikebuchi R, Moriya T, Ueda M, Miyake K, Ono S, Tomura M. Epithelial membrane protein 3 (Emp3) downregulates induction and function of cytotoxic T lymphocytes by macrophages via TNF-α production. Cell Immunol 2019; 324:33-41. [PMID: 29269102 DOI: 10.1016/j.cellimm.2017.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/22/2017] [Accepted: 12/01/2017] [Indexed: 02/07/2023]
Abstract
Tetraspanin membrane protein, epithelial membrane protein 3 (Emp3), is expressed in lymphoid tissues. Herein, we have examined the Emp3 in antigen presenting cell (APC) function in the CD8+ cytotoxic T lymphocytes (CTLs) induction. Emp3-overexpressing RAW264.7 macrophage cell line derived from BALB/c mice reduced anti-C57BL/6 alloreactive CTL induction, while Emp3-knockdown RAW264.7 enhanced it compared with parent RAW267.4. Emp3-overexpressing RAW264.7 inhibited, but Emp3-knockdown RAW264.7 augmented, CD8+ T cell proliferation, interferon-γ secretion, IL-2 consumption, and IL-2Rα expression on CD8+ T cells. The supernatant from co-culture with Emp3-overexpressing RAW264.7 contained higher amount of TNF-α, and TNF- α neutralization significantly restored all these inhibitions and the alloreactive CTL induction. These results suggest that Emp3 in allogeneic APCs possesses the inhibitory function of alloreactive CTL induction by downregulation of IL-2Rα expression CD8+ T cells via an increase in TNF-α production. This demonstrates a novel mechanism for regulating CTL induction by Emp3 in APCs through TNF-α production.
Collapse
Affiliation(s)
- Yutaka Kusumoto
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Japan.
| | - Hiromi Okuyama
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Japan
| | - Takuma Shibata
- Division of Infectious Genetics, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Kazunori Konno
- Division of Infectious Genetics, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yusuke Takemoto
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Japan
| | - Daisuke Maekawa
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Japan
| | - Tomoyuki Kononaga
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Japan
| | - Takashi Ishii
- Division of Infectious Genetics, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Sachiko Akashi-Takamura
- Division of Infectious Genetics, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Shin-Ichiroh Saitoh
- Division of Infectious Genetics, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Ryoyo Ikebuchi
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Japan; Research Fellow of Japan Society for the Promotion of Science, Japan
| | - Taiki Moriya
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Japan
| | - Mizuki Ueda
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Japan
| | - Kensuke Miyake
- Division of Infectious Genetics, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Shiro Ono
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Japan
| | - Michio Tomura
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Japan.
| |
Collapse
|
29
|
Simonović N, Witalisz-Siepracka A, Meissl K, Lassnig C, Reichart U, Kolbe T, Farlik M, Bock C, Sexl V, Müller M, Strobl B. NK Cells Require Cell-Extrinsic and -Intrinsic TYK2 for Full Functionality in Tumor Surveillance and Antibacterial Immunity. THE JOURNAL OF IMMUNOLOGY 2019; 202:1724-1734. [PMID: 30718299 DOI: 10.4049/jimmunol.1701649] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/14/2019] [Indexed: 12/17/2022]
Abstract
Tyrosine kinase 2 (TYK2) is a widely expressed receptor-associated kinase that is involved in signaling by a variety of cytokines with important immune regulatory activities. Absence of TYK2 in mice results in impaired NK cell maturation and antitumor activity, although underlying mechanisms are largely unknown. Using conditional ablation of TYK2 in NK cells we show that TYK2 is required for IFN-γ production by NK cells in response to IL-12 and for an efficient immune defense against Listeria monocytogenes Deletion of TYK2 in NK cells did not impact NK cell maturation and IFN-γ production upon NK cell activating receptor (actR) stimulation. Similarly, NK cell-mediated tumor surveillance was unimpaired upon deletion of TYK2 in NK cells only. In line with the previously reported maturation-associated Ifng promoter demethylation, the less mature phenotype of Tyk2-/- NK cells correlated with an increased CpG methylation at the Ifng locus. Treatment with the DNA hypomethylating agent 5-aza-2-deoxycytidine restored the ability of Tyk2-/- NK cells to produce IFN-γ upon actR but not upon IL-12 stimulation. NK cell maturation was dependent on the presence of TYK2 in dendritic cells and could be rescued in Tyk2-deficient mice by treatment with exogenous IL-15/IL-15Rα complexes. IL-15 treatment also rescued the in vitro cytotoxicity defect and the impaired actR-induced IFN-γ production of Tyk2-/- NK cells. Collectively, our findings provide the first evidence, to our knowledge, for a key role of TYK2 in the host environment in promoting NK cell maturation and antitumor activity.
Collapse
Affiliation(s)
- Natalija Simonović
- Department of Biomedical Science, Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Agnieszka Witalisz-Siepracka
- Department of Biomedical Science, Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria.,Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Katrin Meissl
- Department of Biomedical Science, Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Caroline Lassnig
- Department of Biomedical Science, Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria.,Biomodels Austria, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Ursula Reichart
- Department of Biomedical Science, Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria.,Biomodels Austria, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Thomas Kolbe
- Biomodels Austria, University of Veterinary Medicine Vienna, 1210 Vienna, Austria.,Department of Agrobiotechnology IFA Tulln, University of Natural Resources and Life Sciences, 1180 Vienna, Austria; and
| | - Matthias Farlik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Mathias Müller
- Department of Biomedical Science, Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria.,Biomodels Austria, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Birgit Strobl
- Department of Biomedical Science, Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, 1210 Vienna, Austria;
| |
Collapse
|
30
|
Parsonidis P, Ntanovasilis DA, Papasotiriou I. MUC1 Antigen-Specific CD8 T Lymphocytes Targeting MCF7 and MDA-MB-231 Human Breast Adenocarcinoma Cell Lines. ACTA ACUST UNITED AC 2019. [DOI: 10.4236/jct.2019.107041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
31
|
Lopes A, Vanvarenberg K, Kos Š, Lucas S, Colau D, Van den Eynde B, Préat V, Vandermeulen G. Combination of immune checkpoint blockade with DNA cancer vaccine induces potent antitumor immunity against P815 mastocytoma. Sci Rep 2018; 8:15732. [PMID: 30356111 PMCID: PMC6200811 DOI: 10.1038/s41598-018-33933-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/24/2018] [Indexed: 02/07/2023] Open
Abstract
DNA vaccination against cancer has become a promising strategy for inducing a specific and long-lasting antitumor immunity. However, DNA vaccines fail to generate potent immune responses when used as a single therapy. To enhance their activity into the tumor, a DNA vaccine against murine P815 mastocytoma was combined with antibodies directed against the immune checkpoints CTLA4 and PD1. The combination of these two strategies delayed tumor growth and enhanced specific antitumor immune cell infiltration in comparison to the corresponding single therapies. The combination also promoted IFNg, IL12 and granzyme B production in the tumor microenvironment and decreased the formation of liver metastasis in a very early phase of tumor development, enabling 90% survival. These results underline the complementarity of DNA vaccination and immune checkpoint blockers in inducing a potent immune response, by exploiting the generation of antigen-specific T cells by the vaccine and the ability of immune checkpoint blockers to enhance T cell activity and infiltration in the tumor. These findings suggest how and why a rational combination therapy can overcome the limits of DNA vaccination but could also allow responses to immune checkpoint blockers in a larger proportion of subjects.
Collapse
Affiliation(s)
- Alessandra Lopes
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, B-1200, Belgium
| | - Kevin Vanvarenberg
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, B-1200, Belgium
| | - Špela Kos
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Zaloska 2, SI-1000, Ljubljana, Slovenia
| | - Sophie Lucas
- de Duve Institute, Université Catholique de Louvain, Brussels, B-1200, Belgium
| | - Didier Colau
- de Duve Institute, Université Catholique de Louvain, Brussels, B-1200, Belgium.,Ludwig Institute for Cancer Research, Brussels, B-1200, Belgium
| | - Benoît Van den Eynde
- de Duve Institute, Université Catholique de Louvain, Brussels, B-1200, Belgium.,Ludwig Institute for Cancer Research, Brussels, B-1200, Belgium
| | - Véronique Préat
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, B-1200, Belgium.
| | - Gaëlle Vandermeulen
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, B-1200, Belgium
| |
Collapse
|
32
|
Cole E, Brown TA, Pinkerton KE, Postma B, Malany K, Yang M, Kim YJ, Hamilton RF, Holian A, Cho YH. Perinatal exposure to environmental tobacco smoke is associated with changes in DNA methylation that precede the adult onset of lung disease in a mouse model. Inhal Toxicol 2018; 29:435-442. [PMID: 29124997 DOI: 10.1080/08958378.2017.1392655] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Prenatal and early-life environmental tobacco smoke (ETS) exposure can induce epigenetic alterations associated with inflammation and respiratory disease. The objective of this study was to address the long-term epigenetic consequences of perinatal ETS exposure on latent respiratory disease risk, which are still largely unknown. C57BL/6 mice were exposed to prenatal and early-life ETS; offspring lung pathology, global DNA, and gene-specific methylation were measured at two adult ages. Significant alterations in global DNA methylation and promoter methylation of IFN-γ and Thy-1 were found in ETS-exposed offspring at 10-12 and 20 weeks of age. These sustained epigenetic alterations preceded the onset of significant pulmonary pathologies observed at 20 weeks of age. This study suggests that perinatal ETS exposure induces persistent epigenetic alterations in global DNA, as well as IFN-γ and Thy-1 promoter methylation that precede the adult onset of fibrotic lung pathology. These epigenetic findings could represent potential biomarkers of latent respiratory disease risk.
Collapse
Affiliation(s)
- Elizabeth Cole
- a Department of Biomedical and Pharmaceutical Sciences , University of Montana , Missoula , MT , USA
| | - Traci A Brown
- a Department of Biomedical and Pharmaceutical Sciences , University of Montana , Missoula , MT , USA
| | - Kent E Pinkerton
- b Center for Health and the Environment, University of California , Davis , CA , USA
| | - Britten Postma
- a Department of Biomedical and Pharmaceutical Sciences , University of Montana , Missoula , MT , USA
| | - Keegan Malany
- a Department of Biomedical and Pharmaceutical Sciences , University of Montana , Missoula , MT , USA
| | - Mihi Yang
- c Department of Toxicology , Research Center for Cell Fate Control, Sookmyung Women's University , Seoul , Korea
| | - Yang Jee Kim
- d Da Vinci College of General Education , Chung-Ang University , Seoul , Korea
| | - Raymond F Hamilton
- a Department of Biomedical and Pharmaceutical Sciences , University of Montana , Missoula , MT , USA
| | - Andrij Holian
- a Department of Biomedical and Pharmaceutical Sciences , University of Montana , Missoula , MT , USA
| | - Yoon Hee Cho
- a Department of Biomedical and Pharmaceutical Sciences , University of Montana , Missoula , MT , USA
| |
Collapse
|
33
|
Wang M, Yu F, Wu W, Wang Y, Ding H, Qian L. Epstein-Barr virus-encoded microRNAs as regulators in host immune responses. Int J Biol Sci 2018; 14:565-576. [PMID: 29805308 PMCID: PMC5968849 DOI: 10.7150/ijbs.24562] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 03/06/2018] [Indexed: 12/12/2022] Open
Abstract
Epstein-Barr virus (EBV) is an oncogenic virus that infects over 90% of the world's adult population. EBV can establish life-long latent infection in host due to the balance between EBV and host immune system. EBV latency is associated with various malignancies such as nasopharyngeal carcinoma, gastric carcinoma and Burkitt's lymphoma. EBV is the first human virus that has the capability to encode microRNAs (miRNAs). Remarkably, EBV-encoded miRNAs are abundantly expressed in latently-infected cells and serve important function in viral infection and pathogenesis. Increasing evidence indicates that EBV miRNAs target the host mRNAs involved in cell proliferation, apoptosis and transformation. EBV miRNAs also inhibit the expression of viral antigens, thereby enabling infected cells to escape immune recognition. Intriguingly, EBV miRNAs directly suppress host antiviral immunity by interfering with antigen presentation and immune cell activation. This review will update the current knowledge about EBV miRNAs implicated in host immune responses. An in-depth understanding of the functions of EBV miRNAs in host antiviral immunity will shed light on the EBV-host interactions and provide potential therapeutic targets for the treatment of EBV-associated malignancies.
Collapse
Affiliation(s)
- Man Wang
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China
| | - Fei Yu
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China
| | - Wei Wu
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China
| | - Yu Wang
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China
| | - Han Ding
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China
| | - Lili Qian
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China
| |
Collapse
|
34
|
Suárez-Fueyo A, Bradley SJ, Katsuyama T, Solomon S, Katsuyama E, Kyttaris VC, Moulton VR, Tsokos GC. Downregulation of CD3ζ in NK Cells from Systemic Lupus Erythematosus Patients Confers a Proinflammatory Phenotype. THE JOURNAL OF IMMUNOLOGY 2018; 200:3077-3086. [PMID: 29602774 DOI: 10.4049/jimmunol.1700588] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 03/06/2018] [Indexed: 01/14/2023]
Abstract
Cytotoxic function and cytokine profile of NK cells are compromised in patients with systemic lupus erythematosus (SLE). CD3ζ, an important molecule for NK cell activation, is downregulated in SLE T cells and contributes to their altered function. However, little is known about the role of CD3ζ in SLE NK cells. We studied CD3ζ levels and its contribution to cytotoxic, degranulation, and cytokine production capacity of NK cells from patients with SLE. Furthermore, we studied the human NK cell line, NKL, in which manipulation of CD3ζ levels was achieved using small interfering RNA and NK cells from Rag2 mice deficient in CD3ζ. We found reduced CD3ζ expression in NK cells from SLE patients independent of disease activity. Downregulation of CD3ζ expression in NK cells is mediated, at least in part, by Caspase 3, the activity of which is higher in NK cells from patients with SLE compared with NK cells from healthy donors. CD3ζ levels correlated inversely with natural cytotoxicity and the percentage of cells capable of producing the proinflammatory cytokines IFN-γ and TNF. In contrast, CD3ζ levels showed a direct correlation with levels of Ab-dependent cellular cytotoxicity. Experiments performed in CD3ζ-silenced NKL and CD3ζ-deficient NK cells from Rag2 mice confirmed the dependence of NK cell function on CD3ζ levels. Our results demonstrate a differential role for CD3ζ in natural cytotoxicity and Ab-dependent cellular cytotoxicity. We conclude that downregulated CD3ζ confers a proinflammatory phenotype to SLE NK cells and contributes to their altered function in patients with SLE.
Collapse
Affiliation(s)
- Abel Suárez-Fueyo
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Sean J Bradley
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Takayuki Katsuyama
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Sarah Solomon
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Eri Katsuyama
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Vasileios C Kyttaris
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Vaishali R Moulton
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - George C Tsokos
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
35
|
Tsukahara T, Nakamura SI, Romero-Pèrez GA, Ohwaki M, Yanagisawa T, Kan T. Stimulation of murine cell-mediated immunity by dietary administration of a cell preparation of Enterococcus faecalis strain KH-2 and its possible activity against tumour development in mice. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2018; 37:49-57. [PMID: 30094120 PMCID: PMC6081610 DOI: 10.12938/bmfh.17-021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/09/2018] [Indexed: 01/09/2023]
Abstract
It is well known that dietary lactic acid bacteria (LAB) stimulate cell-mediated immunity such as natural killer (NK) activity in mice. Here, we aimed to assay the immunomodulatory effects of a cell preparation of Enterococcus faecalis strain KH-2 (CPEF). We further evaluated the possibility of antitumour activity caused by CPEF administration, because NK cells actively participate in the prevention of tumour formation. NK cell activity and gene expression of IFN-γ and Perforin 1, which were induced most likely by a synergetic action of their cytotoxic activity, were higher in splenocytes of CPEF-administered mice than they were in control mice. Moreover, unlike those of control mice, the splenocytes of CPEF-administered mice had significantly higher CD28+CD69+/CD4+ and CD28+CD69+/CD8+ ratios that resulted in a survival rate with a tendency toward improvement after 47 days of CPEF administration (p=0.1) in Meth-A fibrosarcoma-bearing mice. In conclusion, we showed that CPEF might be effective in treating Meth-A fibrosarcoma in mice, as it helped increase their survival rate via stimulation of an immune response in splenocytes, which involved systemic cellular immunity processes such as cytotoxic activity, and active T cells.
Collapse
Affiliation(s)
- Takamitsu Tsukahara
- Kyoto Institute of Nutrition and Pathology, 7-2 Furuikedani, Ujitawara-cho, Tsuzuki-gun, Kyoto 610-0231, Japan
| | - Shin-Ichi Nakamura
- Kyoto Institute of Nutrition and Pathology, 7-2 Furuikedani, Ujitawara-cho, Tsuzuki-gun, Kyoto 610-0231, Japan
| | - Gustavo A Romero-Pèrez
- Kyoto Institute of Nutrition and Pathology, 7-2 Furuikedani, Ujitawara-cho, Tsuzuki-gun, Kyoto 610-0231, Japan
| | - Makoto Ohwaki
- Non-Profit Organisation, The Japanese Association of Clinical Research on Supplements, 1-9-24 Shihogi, Hidaka-shi, Saitama 350-1248, Japan
| | - Takaharu Yanagisawa
- Broma Laboratory Ltd., 1-26 Kandasuda-cho, Chiyoda-ku, Tokyo 101-0041, Japan
| | - Tatsuhiko Kan
- Bio-Lab Co., Ltd., 2-1-3 Komagawa, Hidaka-shi, Saitama 350-1249, Japan
| |
Collapse
|
36
|
Kulling PM, Olson KC, Hamele CE, Toro MF, Tan SF, Feith DJ, Loughran TP. Dysregulation of the IFN-γ-STAT1 signaling pathway in a cell line model of large granular lymphocyte leukemia. PLoS One 2018; 13:e0193429. [PMID: 29474442 PMCID: PMC5825082 DOI: 10.1371/journal.pone.0193429] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 02/09/2018] [Indexed: 02/04/2023] Open
Abstract
T cell large granular lymphocyte leukemia (T-LGLL) is a rare incurable disease that is characterized by defective apoptosis of cytotoxic CD8+ T cells. Chronic activation of the Janus Kinase-Signal Transducer and Activator of Transcription (JAK-STAT) pathway is a hallmark of T-LGLL. One manifestation is the constitutive phosphorylation of tyrosine 701 of STAT1 (p-STAT1). T-LGLL patients also exhibit elevated serum levels of the STAT1 activator, interferon-γ (IFN-γ), thus contributing to an inflammatory environment. In normal cells, IFN-γ production is tightly controlled through induction of IFN-γ negative regulators. However, in T-LGLL, IFN-γ signaling lacks this negative feedback mechanism as evidenced by excessive IFN-γ production and decreased levels of suppressors of cytokine signaling 1 (SOCS1), a negative regulator of IFN-γ. Here we characterize the IFN-γ-STAT1 pathway in TL-1 cells, a cell line model of T-LGLL. TL-1 cells exhibited lower IFN-γ receptor protein and mRNA expression compared to an IFN-γ responsive cell line. Furthermore, IFN-γ treatment did not induce JAK2 or STAT1 activation or transcription of IFN-γ-inducible gene targets. However, IFN-β induced p-STAT1 and subsequent STAT1 gene transcription, demonstrating a specific IFN-γ signaling defect in TL-1 cells. We utilized siRNA targeting of STAT1, STAT3, and STAT5b to probe their role in IL-2-mediated IFN-γ regulation. These studies identified STAT5b as a positive regulator of IFN-γ production. We also characterized the relationship between STAT1, STAT3, and STAT5b proteins. Surprisingly, p-STAT1 was positively correlated with STAT3 levels while STAT5b suppressed the activation of both STAT1 and STAT3. Taken together, these results suggest that the dysregulation of the IFN-γ-STAT1 signaling pathway in TL-1 cells likely results from low levels of the IFN-γ receptor. The resulting inability to induce negative feedback regulators explains the observed elevated IL-2 driven IFN-γ production. Future work will elucidate the best way to target this pathway, with the ultimate goal to find a better therapeutic for T-LGLL.
Collapse
Affiliation(s)
- Paige M. Kulling
- University of Virginia Cancer Center, University of Virginia; Charlottesville, VA United States of America
- Department of Medicine, Division of Hematology/Oncology, University of Virginia; Charlottesville, VA United States of America
- Department of Pathology, University of Virginia; Charlottesville, VA United States of America
| | - Kristine C. Olson
- University of Virginia Cancer Center, University of Virginia; Charlottesville, VA United States of America
- Department of Medicine, Division of Hematology/Oncology, University of Virginia; Charlottesville, VA United States of America
| | - Cait E. Hamele
- University of Virginia Cancer Center, University of Virginia; Charlottesville, VA United States of America
- Department of Medicine, Division of Hematology/Oncology, University of Virginia; Charlottesville, VA United States of America
| | - Mariella F. Toro
- University of Virginia Cancer Center, University of Virginia; Charlottesville, VA United States of America
- Department of Medicine, Division of Hematology/Oncology, University of Virginia; Charlottesville, VA United States of America
| | - Su-Fern Tan
- University of Virginia Cancer Center, University of Virginia; Charlottesville, VA United States of America
- Department of Medicine, Division of Hematology/Oncology, University of Virginia; Charlottesville, VA United States of America
| | - David J. Feith
- University of Virginia Cancer Center, University of Virginia; Charlottesville, VA United States of America
- Department of Medicine, Division of Hematology/Oncology, University of Virginia; Charlottesville, VA United States of America
| | - Thomas P. Loughran
- University of Virginia Cancer Center, University of Virginia; Charlottesville, VA United States of America
- Department of Medicine, Division of Hematology/Oncology, University of Virginia; Charlottesville, VA United States of America
- * E-mail:
| |
Collapse
|
37
|
Chu F, Hu Y, Zhou Y, Guo M, Lu J, Zheng W, Xu H, Zhao J, Xu L. MicroRNA-126 deficiency enhanced the activation and function of CD4 + T cells by elevating IRS-1 pathway. Clin Exp Immunol 2018; 191:166-179. [PMID: 28987000 PMCID: PMC5758368 DOI: 10.1111/cei.13067] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2017] [Indexed: 01/01/2023] Open
Abstract
Recent evidence has shown that microRNA-126 (miR-126) has been involved in the development and function of immune cells, which contributed to the pathogenesis of related clinical diseases. However, the potential role of miR-126 in the development and function of CD4+ T cells remains largely unknown. Here we first found that the activation and proliferation, as well as the expression of interferon (IFN)-γ, of CD4+ T cells from miR-126 knock-down (KD) mice using the miRNA-sponge technique were enhanced significantly in vitro, compared with those in CD4+ T cells from wild-type (WT) mice. To monitor further the possible effect of miR-126 deficiency on the function of CD4+ T cells in vivo, we used dextran sulphate sodium (DSS)-induced murine model of acute autoimmune colitis and found that miR-126 deficiency could elevate the pathology of colitis. Importantly, the proportion of CD4+ T cells in splenocytes increased significantly in miR-126KD mice. Moreover, the expression levels of CD69 and CD44 on CD4+ T cells increased significantly and the expression level of CD62L decreased significantly. Of note, adoptive cell transfer assay showed that the pathology of colitis was more serious in carboxyfluorescein succinimidyl ester (CFSE)-labelled miR-126KD CD4+ T cell-transferred group, compared with that in the CFSE-labelled WT CD4+ T cells transferred group. Consistently, the expression levels of CD69 and CD44 on CFSE+ cells increased significantly. Furthermore, both the proliferation and IFN-γ secretion of CFSE+ cells also increased significantly in the CFSE-labelled miR-126KD CD4+ T cell-transferred group. Mechanistic evidence showed that the expression of insulin receptor substrate 1 (IRS-1), as a functional target of miR-126, was elevated in CD4+ T cells from miR-126KD mice, accompanied by altered transduction of the extracellular regulated kinase, protein B (AKT) and nuclear factor kappa B (NF-κB) pathway. Our data revealed a novel role in which miR-126 was an intrinsic regulator in the function of CD4+ T cells, which provided preliminary basis for exploring further the role of miR-126 in the development, function of CD4+ T cells and related clinical diseases.
Collapse
Affiliation(s)
- F. Chu
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Provincial Education DepartmentGuizhouChina
- Department of ImmunologyZunyi Medical CollegeGuizhouChina
| | - Y. Hu
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Provincial Education DepartmentGuizhouChina
- Department of ImmunologyZunyi Medical CollegeGuizhouChina
| | - Y. Zhou
- Department of Medical PhysicsZunyi Medical CollegeGuizhouChina
| | - M. Guo
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Provincial Education DepartmentGuizhouChina
- Department of ImmunologyZunyi Medical CollegeGuizhouChina
| | - J. Lu
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Provincial Education DepartmentGuizhouChina
- Department of ImmunologyZunyi Medical CollegeGuizhouChina
| | - W. Zheng
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Provincial Education DepartmentGuizhouChina
- Department of ImmunologyZunyi Medical CollegeGuizhouChina
| | - H. Xu
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Provincial Education DepartmentGuizhouChina
- Department of ImmunologyZunyi Medical CollegeGuizhouChina
| | - J. Zhao
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Provincial Education DepartmentGuizhouChina
- Department of ImmunologyZunyi Medical CollegeGuizhouChina
| | - L. Xu
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Provincial Education DepartmentGuizhouChina
- Department of ImmunologyZunyi Medical CollegeGuizhouChina
| |
Collapse
|
38
|
Ge A, Fu F, Wang X. The enhancing effect of Klebsiella pneumoniae lysate on cellular immunity. EUR J INFLAMM 2018. [DOI: 10.1177/2058739218776476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cellular immunity plays important roles in clearing intracellular pathogens or tumor cells. It is of significance to develop an adjuvant able to efficaciously induce cellular immunity, but such an approved adjuvant is not currently available. Klebsiella pneumoniae ( K. pneumoniae) possesses lipopolysaccharide and capsular polysaccharide, both of which have been demonstrated to be able to induce humoral immunity. In this study, we investigated the effect of K. pneumoniae on epidermal cellular immunity, in vitro. The effects of K. pneumoniae on the maturation of Langerhans cells (LCs), the capacity to induce T-cell proliferation, and the secretion of interferon gamma (IFN-γ) by T cells were examined. The results showed that K. pneumoniae induced significant upregulation of CD86 and human leukocyte antigen-deterodimer (HLA-DR) levels on LCs, but not CD40 and CD80. K. pneumoniae-loaded LCs induced significant CD4+ and CD8+ T-cell proliferation. Significant increases in extracellular IFN-γ secretion by CD4+ and CD8+ T cells stimulated with K. pneumoniae-pulsed LCs using enzyme-linked immunospot assay (ELISPOT) were demonstrated, while only a part of the subjects showed increases in intracellular secretion of IFN-γ using intracellular staining assay. In sum, K. pneumoniae has the potential to enhance epidermal cellular immunity and may act as a potential adjuvant in intradermal vaccines designed to enhance cellular immunity.
Collapse
Affiliation(s)
- Anxing Ge
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, China
- Research Center of Biomedical Technology, Jiangsu Vocational College of Medicine, Yancheng, China
| | - Fangjie Fu
- Third Department of Clinical Medicine, China Medical University, Shenyang, China
| | - Xuelian Wang
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| |
Collapse
|
39
|
Mastria EM, Cai LY, Kan MJ, Li X, Schaal JL, Fiering S, Gunn MD, Dewhirst MW, Nair SK, Chilkoti A. Nanoparticle formulation improves doxorubicin efficacy by enhancing host antitumor immunity. J Control Release 2017; 269:364-373. [PMID: 29146246 DOI: 10.1016/j.jconrel.2017.11.021] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 10/15/2017] [Accepted: 11/11/2017] [Indexed: 12/14/2022]
Abstract
Strategies that enhance the host antitumor immune response promise to revolutionize cancer therapy. Optimally mobilizing the immune system will likely require a multi-pronged approach to overcome the resistance developed by tumors to therapy. Recently, it has become recognized that doxorubicin can contribute to re-establishing host antitumor immunity through the generation of immunogenic cell death. However, the potential for delivery strategies to further enhance the immunological effects of doxorubicin has not been adequately examined. We report herein that Chimeric Polypeptide Doxorubicin (CP-Dox), a nanoparticle formulation of doxorubicin, enhances antitumor immunity. Compared to free doxorubicin, a single intravenous (IV) administration of CP-Dox at the maximum tolerated dose increases the infiltration of leukocytes into the tumor, slowing tumor growth and preventing metastasis in poorly immunogenic 4T1 mammary carcinoma. We demonstrate that the full efficacy of CP-Dox is dependent on CD8+ T cells and IFN-γ. CP-dox treatment also repolarized intratumoral myeloid cells towards an antitumor phenotype. These findings demonstrate that a nanoparticle drug is distinct from the free drug in its ability to productively stimulate antitumor immunity. Our study strongly argues for the use of antitumor immunotherapies combined with nanoparticle-packaged chemotherapy.
Collapse
Affiliation(s)
- Eric M Mastria
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Leon Y Cai
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Matthew J Kan
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - Xinghai Li
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Jeffrey L Schaal
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Steven Fiering
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Norris Cotton Cancer Center, Lebanon, NH, USA
| | - Michael D Gunn
- Department of Immunology, Duke University Medical Center, Durham, NC, USA; Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Mark W Dewhirst
- Department of Biomedical Engineering, Duke University, Durham, NC, USA; Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Smita K Nair
- Department of Surgery, Duke University Medical Center, Durham, NC, USA; Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, NC, USA; Center for Biologically Inspired Materials and Materials Systems, Duke University, Durham, NC, USA.
| |
Collapse
|
40
|
Behnam Sani K, Sawitzki B. Immune monitoring as prerequisite for transplantation tolerance trials. Clin Exp Immunol 2017; 189:158-170. [PMID: 28518214 DOI: 10.1111/cei.12988] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2017] [Indexed: 02/06/2023] Open
Abstract
Ever since its first application in clinical medicine, scientists have been urged to induce tolerance towards foreign allogeneic transplants and thus avoid rejection by the recipient's immune system. This would circumvent chronic use of immunosuppressive drugs (IS) and thus avoid development of IS-induced side effects, which are contributing to the still unsatisfactory long-term graft and patient survival after solid organ transplantation. Although manifold strategies of tolerance induction have been described in preclinical models, only three therapeutic approaches have been utilized successfully in a still small number of patients. These approaches are based on (i) IS withdrawal in spontaneous operational tolerant (SOT) patients, (ii) induction of a mixed chimerism and (iii) adoptive transfer of regulatory cells. Results of clinical trials utilizing these approaches show that tolerance induction does not work in all patients. Thus, there is a need for reliable biomarkers, which can be used for patient selection and post-therapeutic immune monitoring of safety, success and failure. In this review, we summarize recent achievements in the identification and validation of such immunological assays and biomarkers, focusing mainly on kidney and liver transplantation. From the published findings so far, it has become clear that indicative biomarkers may vary between different therapeutic approaches applied and organs transplanted. Also, patient numbers studied so far are very small. This is the main reason why nearly all described parameters lack validation and reproducibility testing in large clinical trials, and are therefore not yet suitable for clinical practice.
Collapse
Affiliation(s)
- K Behnam Sani
- Institute of Medical Immunology, Charité Universitaetsmedizin Berlin, Berlin, Germany
| | - B Sawitzki
- Institute of Medical Immunology, Charité Universitaetsmedizin Berlin, Berlin, Germany
| |
Collapse
|
41
|
Abstract
IFN-γ is a critical inflammatory cytokine that is closely regulated to cellular metabolic status and requires high rates of glycolysis. It has now been shown that a key mechanism to link these pathways is through maintenance of acetyl-CoA and epigenetic regulation of the IFNG locus.
Collapse
Affiliation(s)
- Peter J Siska
- Department of Pathology, Microbiology and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232-2363, USA
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232-2363, USA.
| |
Collapse
|
42
|
Yu HR, Tain YL, Sheen JM, Tiao MM, Chen CC, Kuo HC, Hung PL, Hsieh KS, Huang LT. Prenatal Dexamethasone and Postnatal High-Fat Diet Decrease Interferon Gamma Production through an Age-Dependent Histone Modification in Male Sprague-Dawley Rats. Int J Mol Sci 2016; 17:ijms17101610. [PMID: 27669212 PMCID: PMC5085643 DOI: 10.3390/ijms17101610] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/11/2016] [Accepted: 09/14/2016] [Indexed: 02/07/2023] Open
Abstract
Overexposure to prenatal glucocorticoid (GC) disturbs hypothalamic-pituitary-adrenocortical axis-associated neuroendocrine metabolism and susceptibility to metabolic syndrome. A high-fat (HF) diet is a major environmental factor that can cause metabolic syndrome. We aimed to investigate whether prenatal GC plus a postnatal HF diet could alter immune programming in rat offspring. Pregnant Sprague-Dawley rats were given intraperitoneal injections of dexamethasone or saline at 14-21 days of gestation. Male offspring were then divided into four groups: vehicle, prenatal dexamethasone exposure, postnatal HF diet (VHF), and prenatal dexamethasone exposure plus a postnatal HF diet (DHF). The rats were sacrificed and adaptive immune function was evaluated. Compared to the vehicle, the DHF group had lower interferon gamma (IFN-γ) production by splenocytes at postnatal day 120. Decreases in H3K9 acetylation and H3K36me3 levels at the IFN-γ promoter correlated with decreased IFN-γ production. The impaired IFN-γ production and aberrant site-specific histone modification at the IFN-γ promoter by prenatal dexamethasone treatment plus a postnatal HF diet resulted in resilience at postnatal day 180. Prenatal dexamethasone and a postnatal HF diet decreased IFN-γ production through a site-specific and an age-dependent histone modification. These findings suggest a mechanism by which prenatal exposure to GC and a postnatal environment exert effects on fetal immunity programming.
Collapse
Affiliation(s)
- Hong-Ren Yu
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - You-Lin Tain
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Jiunn-Ming Sheen
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Mao-Meng Tiao
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Chih-Cheng Chen
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Ho-Chang Kuo
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Pi-Lien Hung
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Kai-Sheng Hsieh
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Li-Tung Huang
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| |
Collapse
|
43
|
High-Resolution Expression Profiling of Peripheral Blood CD8 + Cells in Patients with Multiple Sclerosis Displays Fingolimod-Induced Immune Cell Redistribution. Mol Neurobiol 2016; 54:5511-5525. [PMID: 27631876 DOI: 10.1007/s12035-016-0075-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 08/23/2016] [Indexed: 10/21/2022]
Abstract
Fingolimod, a sphingosine-1-phosphate (S1P) receptor modulator, is an oral drug approved for the treatment of active relapsing-remitting multiple sclerosis (RRMS). It selectively inhibits the egress of lymphocytes from lymph nodes. We studied the changes in the transcriptome of peripheral blood CD8+ cells to unravel the effects at the molecular level during fingolimod therapy. We separated CD8+ cells from the blood of RRMS patients before the first dose of fingolimod as well as 24 h and 3 months after the start of therapy. Changes in the expression of coding and non-coding genes were measured with high-density Affymetrix Human Transcriptome Array (HTA) 2.0 microarrays. Differentially expressed genes in response to therapy were identified by t test and fold change and analyzed for their functions and molecular interactions. No gene was expressed at significantly higher or lower levels 24 h after the first administration of fingolimod compared to baseline. However, after 3 months of therapy, 861 transcripts were found to be differentially expressed, including interleukin and chemokine receptors. Some of the genes are associated to the S1P pathway, such as the receptor S1P5 and the kinase MAPK1, which were significantly increased in expression. The fingolimod-induced transcriptome changes reflect a shift in the proportions of CD8+ T cell subsets, with CCR7- effector memory T cells being relatively increased in frequency in the blood of fingolimod-treated patients. In consequence, CCR7 mRNA levels were reduced by >80 % and genes involved in T cell activation and lymphocyte cytotoxicity were increased in expression. Gene regulatory programs caused by downstream S1P signaling had only minor effects.
Collapse
|
44
|
Cao-Lei L, Veru F, Elgbeili G, Szyf M, Laplante DP, King S. DNA methylation mediates the effect of exposure to prenatal maternal stress on cytokine production in children at age 13½ years: Project Ice Storm. Clin Epigenetics 2016; 8:54. [PMID: 27182285 PMCID: PMC4866030 DOI: 10.1186/s13148-016-0219-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 05/04/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Prenatal maternal stress (PNMS) is an important programming factor of postnatal immunity. We tested here the hypothesis that DNA methylation of genes in the NF-κB signaling pathway in T cells mediates the effect of objective PNMS on Th1 and Th2 cytokine production in blood from 13½ year olds who were exposed in utero to the 1998 Quebec ice storm. RESULTS Bootstrapping analyses were performed with 47 CpGs across a selection of 20 genes for Th1-type cytokines (IFN-γ and IL-2) and Th2-type cytokines (IL-4 and IL-13). Six CpGs in six different NF-κB signaling genes (PIK3CD, PIK3R2, NFKBIA, TRAF5, TNFRSF1B, and LTBR) remained as significant negative mediators of objective PNMS on IFN-γ secretion after correcting for multiple comparisons. However, no mediation effects on IL-2, IL-4 and IL-13 survived Bonferroni correction. CONCLUSIONS The present study provides preliminary evidence supporting the mediating role of DNA methylation in the association between objective aspects of PNMS and child immune states, favoring a Th2 shift.
Collapse
Affiliation(s)
- Lei Cao-Lei
- />Department of Psychiatry, McGill University and Douglas Hospital Research Centre, 6875 LaSalle Blvd, Montreal, Quebec H4H 1R3 Canada
| | - Franz Veru
- />Douglas Hospital Research Centre, Montreal, Quebec Canada
| | | | - Moshe Szyf
- />Department of Pharmacology and Therapeutics and Sackler Program for Epigenetics and Developmental Psychobiology, McGill University, Montreal, Quebec Canada
| | | | - Suzanne King
- />Department of Psychiatry, McGill University and Douglas Hospital Research Centre, 6875 LaSalle Blvd, Montreal, Quebec H4H 1R3 Canada
| |
Collapse
|
45
|
The induction of antigen-specific CTL by in situ Ad-REIC gene therapy. Gene Ther 2016; 23:408-14. [PMID: 26836118 DOI: 10.1038/gt.2016.7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 11/17/2015] [Accepted: 01/20/2016] [Indexed: 01/03/2023]
Abstract
An adenovirus vector carrying the human Reduced Expression in Immortalized Cell (REIC)/Dkk-3 gene (Ad-REIC) mediates simultaneous induction of cancer-selective apoptosis and augmentation of anticancer immunity. In our preclinical and clinical studies, in situ Ad-REIC gene therapy showed remarkable direct and indirect antitumor effects to realize therapeutic cancer vaccines. We herein aimed to confirm the induction of tumor-associated antigen-specific cytotoxic T lymphocytes (CTLs) by Ad-REIC. Using an ovalbumin (OVA), a tumor-associated antigen, expressing E.G7 tumor-bearing mouse model, we investigated the induction and expansion of OVA-specific CTLs responsible for indirect, systemic effects of Ad-REIC. The intratumoral administration of Ad-REIC mediated clear antitumor effects with the accumulation of OVA-specific CTLs in the tumor tissues and spleen. The CD86-positive dendritic cells (DCs) were upregulated in the tumor draining lymph nodes of Ad-REIC-treated mice. In a dual tumor-bearing mouse model in the left and right back, Ad-REIC injection in one side significantly suppressed the tumor growth on both sides and significant infiltration of OVA-specific CTLs into non-injected tumor was also detected. Consequently, in situ Ad-REIC gene therapy is expected to realize a new-generation cancer vaccine via anticancer immune activation with DC and tumor antigen-specific CTL expansion.
Collapse
|
46
|
Gao Y, Wu J, Zhang M, Hou M, Ji M. Mice lack of LRG-47 display the attenuated outcome of infection with Schistosoma japonicum. Parasitol Res 2015; 115:1185-93. [PMID: 26660918 DOI: 10.1007/s00436-015-4853-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/27/2015] [Indexed: 10/22/2022]
Abstract
Interferon-inducible GTPase LRG-47 (also named immune-related GTPase M, Irgm1) is a member of the p47 GTPase family that has been shown to regulate host resistance to intracellular pathogens. Little knowledge has been known about the role of LRG-47 in host's responses to extracellular pathogens. To investigate possible roles of LRG-47 in the course of Schistosoma japonicum infection, LRG-47-deficient (LRG-47(-/-)) and wild-type (WT) mice were challenged with cercariae of S. japonicum, and the cellular and humoral responses in mice were analyzed. At the acute stage of S. japonicum infection, in contrast to WT mice, LRG-47(-/-) mice showed the significantly decreased egg burden, low schistosome-specific antibody response, and the decreased Th1 and increased Tc1 responses. Additionally, Schistosoma japonicum-specific egg antigen immunization also produced the similar humoral and cellular immune responses as S. japonicum infection. Taken together, these data suggested that the deficiency of LRG-47 might affect host's CD4(+) T cell immune response via the weakening of IFN-γ downstream signaling; however, the specific function of LRG-47 on dealing with extracellular worm needs to be further studied.
Collapse
Affiliation(s)
- Yanan Gao
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.,College of Basic Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jingjiao Wu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.,Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Meijuan Zhang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Min Hou
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Minjun Ji
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China. .,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China.
| |
Collapse
|