1
|
Miranda S, Vermeesen R, Janssen A, Rehnberg E, Etlioglu E, Baatout S, Tabury K, Baselet B. Effects of simulated space conditions on CD4+ T cells: a multi modal analysis. Front Immunol 2024; 15:1443936. [PMID: 39286254 PMCID: PMC11402665 DOI: 10.3389/fimmu.2024.1443936] [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: 06/04/2024] [Accepted: 08/08/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction The immune system is an intricate network of cellular components that safeguards against pathogens and aberrant cells, with CD4+ T cells playing a central role in this process. Human space travel presents unique health challenges, such as heavy ion ionizing radiation, microgravity, and psychological stress, which can collectively impede immune function. The aim of this research was to examine the consequences of simulated space stressors on CD4+ T cell activation, cytokine production, and gene expression. Methods CD4+ T cells were obtained from healthy individuals and subjected to Fe ion particle radiation, Photon irradiation, simulated microgravity, and hydrocortisone, either individually or in different combinations. Cytokine levels for Th1 and Th2 cells were determined using multiplex Luminex assays, and RNA sequencing was used to investigate gene expression patterns and identify essential genes and pathways impacted by these stressors. Results Simulated microgravity exposure resulted in an apparent Th1 to Th2 shift, evidenced on the level of cytokine secretion as well as altered gene expression. RNA sequencing analysis showed that several gene pathways were altered, particularly in response to Fe ions irradiation and simulated microgravity exposures. Individually, each space stressor caused differential gene expression, while the combination of stressors revealed complex interactions. Discussion The research findings underscore the substantial influence of the space exposome on immune function, particularly in the regulation of T cell responses. Future work should focus expanding the limited knowledge in this field. Comprehending these modifications will be essential for devising effective strategies to safeguard the health of astronauts during extended space missions. Conclusion The effects of simulated space stressors on CD4+ T cell function are substantial, implying that space travel poses a potential threat to immune health. Additional research is necessary to investigate the intricate relationship between space stressors and to develop effective countermeasures to mitigate these consequences.
Collapse
Affiliation(s)
- Silvana Miranda
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Randy Vermeesen
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
| | - Ann Janssen
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
| | - Emil Rehnberg
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Emre Etlioglu
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Kevin Tabury
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
- Department of Biomedical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, SC, United States
| | - Bjorn Baselet
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
| |
Collapse
|
2
|
Hai S, Li X, Xie E, Wu W, Gao Q, Yu B, Hu J, Xu F, Zheng X, Zhang BH, Wu D, Yan W, Ning Q, Wang X. Intestinal IL-33 promotes microbiota-derived trimethylamine N -oxide synthesis and drives metabolic dysfunction-associated steatotic liver disease progression by exerting dual regulation on HIF-1α. Hepatology 2024:01515467-990000000-00950. [PMID: 38985971 DOI: 10.1097/hep.0000000000000985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND AND AIMS Gut microbiota plays a prominent role in the pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD). IL-33 is highly expressed at mucosal barrier sites and regulates intestinal homeostasis. Herein, we aimed to investigate the role and mechanism of intestinal IL-33 in MASLD. APPROACH AND RESULTS In both humans and mice with MASLD, hepatic expression of IL-33 and its receptor suppression of tumorigenicity 2 (ST2) showed no significant change compared to controls, while serum soluble ST2 levels in humans, as well as intestinal IL-33 and ST2 expression in mice were significantly increased in MASLD. Deletion of global or intestinal IL-33 in mice alleviated metabolic disorders, inflammation, and fibrosis associated with MASLD by reducing intestinal barrier permeability and rectifying gut microbiota dysbiosis. Transplantation of gut microbiota from IL-33 deficiency mice prevented MASLD progression in wild-type mice. Moreover, IL-33 deficiency resulted in a decrease in the abundance of trimethylamine N -oxide-producing bacteria. Inhibition of trimethylamine N -oxide synthesis by 3,3-dimethyl-1-butanol mitigated hepatic oxidative stress in mice with MASLD. Nuclear IL-33 bound to hypoxia-inducible factor-1α and suppressed its activation, directly damaging the integrity of the intestinal barrier. Extracellular IL-33 destroyed the balance of intestinal Th1/Th17 and facilitated Th1 differentiation through the ST2- Hif1a - Tbx21 axis. Knockout of ST2 resulted in a diminished MASLD phenotype resembling that observed in IL-33 deficiency mice. CONCLUSIONS Intestinal IL-33 enhanced gut microbiota-derived trimethylamine N -oxide synthesis and aggravated MASLD progression through dual regulation on hypoxia-inducible factor-1α. Targeting IL-33 and its associated microbiota may provide a potential therapeutic strategy for managing MASLD.
Collapse
Affiliation(s)
- Suping Hai
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Xitang Li
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Erliang Xie
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Wenhui Wu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Gao
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Binghui Yu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Junjian Hu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Feiyang Xu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Xizhe Zheng
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Bin-Hao Zhang
- Department of Surgery, Hepatic Surgery Center, Institute of Hepato-Pancreato-Biliary Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Di Wu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Weiming Yan
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Ning
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojing Wang
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Nayak V, Patra S, Singh KR, Ganguly B, Kumar DN, Panda D, Maurya GK, Singh J, Majhi S, Sharma R, Pandey SS, Singh RP, Kerry RG. Advancement in precision diagnosis and therapeutic for triple-negative breast cancer: Harnessing diagnostic potential of CRISPR-cas & engineered CAR T-cells mediated therapeutics. ENVIRONMENTAL RESEARCH 2023; 235:116573. [PMID: 37437865 DOI: 10.1016/j.envres.2023.116573] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/29/2023] [Accepted: 07/06/2023] [Indexed: 07/14/2023]
Abstract
Cancer is characterized by uncontrolled cell growth, disrupted regulatory pathways, and the accumulation of genetic mutations. These mutations across different types of cancer lead to disruptions in signaling pathways and alterations in protein expression related to cellular growth and proliferation. This review highlights the AKT signaling cascade and the retinoblastoma protein (pRb) regulating cascade as promising for novel nanotheranostic interventions. Through synergizing state-of-the-art gene editing tools like the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas system with nanomaterials and targeting AKT, there is potential to enhance cancer diagnostics significantly. Furthermore, the integration of modified CAR-T cells into multifunctional nanodelivery systems offers a promising approach for targeted cancer inhibition, including the eradication of cancer stem cells (CSCs). Within the context of highly aggressive and metastatic Triple-negative Breast Cancer (TNBC), this review specifically focuses on devising innovative nanotheranostics. For both pre-clinical and post-clinical TNBC detection, the utilization of the CRISPR-Cas system, guided by RNA (gRNA) and coupled with a fluorescent reporter specifically designed to detect TNBC's mutated sequence, could be promising. Additionally, a cutting-edge approach involving the engineering of TNBC-specific iCAR and syn-Notch CAR T-cells, combined with the co-delivery of a hybrid polymeric nano-liposome encapsulating a conditionally replicative adenoviral vector (CRAdV) against CSCs, could present an intriguing intervention strategy. This review thus paves the way for exciting advancements in the field of nanotheranostics for the treatment of TNBC and beyond.
Collapse
Affiliation(s)
- Vinayak Nayak
- Indian Council of Agricultural Research- National Institute on Foot and Mouth Disease- International Center for Foot and Mouth Disease, Bhubaneswar, Odisha, India
| | - Sushmita Patra
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi-Mumbai 410210, India
| | - Kshitij Rb Singh
- Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, 2-4 Hibikino, Wakamatsu, Kitakyushu, Japan.
| | - Bristy Ganguly
- Fish Health Management Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar, Odisha, India
| | - Das Nishant Kumar
- PG Department of Biotechnology, Utkal University, Bhubaneswar, Odisha, India
| | - Deepak Panda
- PG Department of Biotechnology, Utkal University, Bhubaneswar, Odisha, India
| | - Ganesh Kumar Maurya
- Zoology Section, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Jay Singh
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Sanatan Majhi
- PG Department of Biotechnology, Utkal University, Bhubaneswar, Odisha, India
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Shyam S Pandey
- Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, 2-4 Hibikino, Wakamatsu, Kitakyushu, Japan.
| | - Ravindra Pratap Singh
- Department of Biotechnology, Indira Gandhi National Tribal University, Amarkantak, Madhya Pradesh, India.
| | - Rout George Kerry
- PG Department of Biotechnology, Utkal University, Bhubaneswar, Odisha, India.
| |
Collapse
|
4
|
Imširović V, Lenartić M, Wensveen FM, Polić B, Jelenčić V. Largely preserved functionality after the combined loss of NKG2D, NCR1 and CD16 demonstrates the remarkable plasticity of NK cell responsiveness. Front Immunol 2023; 14:1191884. [PMID: 37520575 PMCID: PMC10374020 DOI: 10.3389/fimmu.2023.1191884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Natural killer (NK) cells play an important role in the early defense against tumors and virally infected cells. Their function is thought to be controlled by the balance between activating and inhibitory receptors, which often compete for the same ligands. Several activating receptors expressed on virtually all NK cells lack an inhibitory partner, most notably CD16, NCR1 and NKG2D. We therefore hypothesized that a signal through at least one of these receptors is always required for full NK cell activation. We generated animals lacking all three receptors (TKO) and analyzed their NK cells. In vitro, TKO NK cells did not show reduced ability to kill tumor targets but displayed hyperresponsiveness to NK1.1 stimulation. In vivo, TKO animals had a minor reduction in their ability to control non-hematopoietic tumors and cytomegalovirus infection, which was the result of reduced NK cell activity. Together, our findings show that activating NK cell receptors without an inhibitory partner do not provide a 'master' signal but are integrated in the cumulative balance of activating and inhibitory signals. Their activity is controlled through regulation of the responsiveness and expression of other activating receptors. Our findings may be important for future development of NK cell-based cancer immunotherapy.
Collapse
|
5
|
Cheung J, Zahorowska B, Suranyi M, Wong JKW, Diep J, Spicer ST, Verma ND, Hodgkinson SJ, Hall BM. CD4 +CD25 + T regulatory cells in renal transplantation. Front Immunol 2022; 13:1017683. [PMID: 36426347 PMCID: PMC9681496 DOI: 10.3389/fimmu.2022.1017683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/13/2022] [Indexed: 09/14/2023] Open
Abstract
The immune response to an allograft activates lymphocytes with the capacity to cause rejection. Activation of CD4+CD25+Foxp3+T regulatory cells (Treg) can down-regulate allograft rejection and can induce immune tolerance to the allograft. Treg represent <10% of peripheral CD4+T cells and do not markedly increase in tolerant hosts. CD4+CD25+Foxp3+T cells include both resting and activated Treg that can be distinguished by several markers, many of which are also expressed by effector T cells. More detailed characterization of Treg to identify increased activated antigen-specific Treg may allow reduction of non-specific immunosuppression. Natural thymus derived resting Treg (tTreg) are CD4+CD25+Foxp3+T cells and only partially inhibit alloantigen presenting cell activation of effector cells. Cytokines produced by activated effector cells activate these tTreg to more potent alloantigen-activated Treg that may promote a state of operational tolerance. Activated Treg can be distinguished by several molecules they are induced to express, or whose expression they have suppressed. These include CD45RA/RO, cytokine receptors, chemokine receptors that alter pathways of migration and transcription factors, cytokines and suppression mediating molecules. As the total Treg population does not increase in operational tolerance, it is the activated Treg which may be the most informative to monitor. Here we review the methods used to monitor peripheral Treg, the effect of immunosuppressive regimens on Treg, and correlations with clinical outcomes such as graft survival and rejection. Experimental therapies involving ex vivo Treg expansion and administration in renal transplantation are not reviewed.
Collapse
Affiliation(s)
- Jason Cheung
- Renal Unit, Liverpool Hospital, Sydney, NSW, Australia
| | | | - Michael Suranyi
- Renal Unit, Liverpool Hospital, Sydney, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
| | | | - Jason Diep
- Renal Unit, Liverpool Hospital, Sydney, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Stephen T. Spicer
- Renal Unit, Liverpool Hospital, Sydney, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Nirupama D. Verma
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
- Immune Tolerance Laboratory, Ingham Institute for Applied Medical Research, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Suzanne J. Hodgkinson
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
- Immune Tolerance Laboratory, Ingham Institute for Applied Medical Research, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Bruce M. Hall
- Renal Unit, Liverpool Hospital, Sydney, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia
- Immune Tolerance Laboratory, Ingham Institute for Applied Medical Research, University of New South Wales (UNSW), Sydney, NSW, Australia
| |
Collapse
|
6
|
Xiang S, Zhang J, Zhang M, Qian S, Wang R, Wang Y, Xiang Y, Ding X. Imbalance of helper T cell type 1, helper T cell type 2 and associated cytokines in patients with systemic lupus erythematosus: A meta-analysis. Front Pharmacol 2022; 13:988512. [PMID: 36249802 PMCID: PMC9556996 DOI: 10.3389/fphar.2022.988512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Th1 and Th2 cells and their associated cytokines function in the pathogenesis of systemic lupus erythematosus (SLE), but their exact roles are uncertain. We performed a meta-analysis to examine the relationship of these cells and cytokines with SLE. Methods: Multiple databases were searched to identify publications that reported the percentages of Th1 and Th2 cells and their associated cytokines in SLE patients and healthy controls (HCs). Meta-analysis was performed using Stata MP version 16. Results: SLE patients had a lower percentage of Th1 cells, a higher percentage of Th2 cells, and higher levels of Th1- and Th2-associated cytokines than HCs. SLE treatments normalized some but not all of these indicators. For studies in which the proportion of females was less than 94%, the percentage of Th2 cells and the level of IL-10 were higher in patients than HCs. SLE patients who had abnormal kidney function and were younger than 30 years old had a higher proportion of Th1 cells than HCs. SLE patients more than 30 years old had a higher level of IL-6 than HCs. Conclusion: Medications appeared to restore the balance of Th1 cells and other disease indicators in patients with SLE. Gender and age affected the levels of Th1 and Th2 cells, and the abnormally elevated levels of Th2 cells appear to be more pronounced in older patients and males. Systematic Review Registration: [https://www.crd.york.ac.uk/prospero/], identifier [CRD42022296540].
Collapse
Affiliation(s)
- Shate Xiang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingjing Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengge Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Suhai Qian
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rongyun Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yao Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yingshi Xiang
- First Clinical School of Medicine, Nanjing Medical University, Nanjing, China
| | - Xinghong Ding
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Xinghong Ding,
| |
Collapse
|
7
|
Yang HX, Liu QP, Zhou YX, Chen YY, An P, Xing YZ, Zhang L, Jia M, Zhang H. Forsythiasides: A review of the pharmacological effects. Front Cardiovasc Med 2022; 9:971491. [PMID: 35958429 PMCID: PMC9357976 DOI: 10.3389/fcvm.2022.971491] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Forsythiasides are a kind of phenylethanol glycosides existing in Forsythia suspensa (Thunb.) Vahl, which possesses extensive pharmacological activities. According to the different groups connected to the nucleus, forsythiasides can be divided into A-K. In recent years, numerous investigations have been carried out on forsythiasides A, B, C, D, E, and I, which have the effects of cardiovascular protection, anti-inflammation, anti-oxidation, neuroprotection, et al. Mechanistically, forsythiasides regulate toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MyD88)/nuclear factor kappaB (NF-κB), nuclear factor-erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) and other signaling pathways, as well as the expression of related cytokines and kinases. Further exploration and development may unearth more treatment potential of forsythiasides and provide more evidence for their clinical applications. In summary, forsythiasides have high development and application value.
Collapse
Affiliation(s)
- Hong-Xuan Yang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiu-Ping Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan-Xi Zhou
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Library, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Ying Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pei An
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi-Zhuo Xing
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Zhang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Lei Zhang,
| | - Min Jia
- Department of Chinese Medicine Authentication, School of Pharmacy, Naval Medical University, Shanghai, China
- Min Jia,
| | - Hong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Hong Zhang,
| |
Collapse
|
8
|
Pan J, Zeng W, Jia J, Shi Y, Wang D, Dong J, Fang Z, He J, Yang X, Zhang R, He M, Huang M, Fu B, Zhong B, Liu H. A Novel Therapeutic Tumor Vaccine Targeting MUC1 in Combination with PD-L1 Elicits Specific Anti-Tumor Immunity in Mice. Vaccines (Basel) 2022; 10:vaccines10071092. [PMID: 35891256 PMCID: PMC9325010 DOI: 10.3390/vaccines10071092] [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: 05/19/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
Dendritic cells (DCs), as professional antigen-presenting cells (APCs), play a key role in the initiation and regulation of humoral and cellular immunity. DC vaccines loaded with different tumor-associated antigens (TAAs) have been widely used to study their therapeutic effects on cancer. A number of clinical trials have shown that DCs are safe as an antitumor vaccine and can activate certain anti-tumor immune responses; however, the overall clinical efficacy of DC vaccine is not satisfactory, so its efficacy needs to be enhanced. MUC1 is a TAA with great potential, and the immune checkpoint PD-L1 also has great potential for tumor treatment. Both of them are highly expressed on the surface of various tumors. In this study, we generated a novel therapeutic MUC1-Vax tumor vaccine based on the method of PD-L1-Vax vaccine we recently developed; this novel PD-L1-containing MUC1-Vax vaccine demonstrated an elevated persistent anti-PD-L1 antibody production and elicited a much stronger protective cytotoxic T lymphocyte (CTL) response in immunized mice. Furthermore, the MUC1-Vax vaccine exhibited a significant therapeutic anti-tumor effect, which significantly inhibited tumor growth by expressing a high MUC1+ and PD-L1+ level of LLC and Panc02 tumor cells, and prolonged the survival of cancer-bearing animals. Taken together, our study provides a new immunotherapy strategy for improving the cross-presentation ability of therapeutic vaccine, which may be applicable to pancreatic cancer, lung cancer and for targeting other types of solid tumors that highly express MUC1 and PD-L1.
Collapse
Affiliation(s)
- Jiayi Pan
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
- Clinical Laboratory, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Science, Guangzhou 510080, China
| | - Wuyi Zeng
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Jiangtao Jia
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Yi Shi
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Danni Wang
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Jun Dong
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Zixuan Fang
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Jiashan He
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Xinyu Yang
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Rong Zhang
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Menghua He
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Maoping Huang
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Bishi Fu
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou 510260, China
| | - Bei Zhong
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
- Correspondence: (B.Z.); (H.L.); Tel./Fax: +86-020-8320-5013 (H.L.)
| | - Hui Liu
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou 510260, China
- Correspondence: (B.Z.); (H.L.); Tel./Fax: +86-020-8320-5013 (H.L.)
| |
Collapse
|
9
|
Immunopathogenesis and distinct role of Th17 in Periodontitis: A review. J Oral Biosci 2022; 64:193-201. [PMID: 35489583 DOI: 10.1016/j.job.2022.04.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 01/02/2023]
Abstract
BACKGROUND Periodontitis is a multifactorial inflammatory disease mediated by the host immune response to dental plaque. Periodontitis is characterized by periodontal bone loss and loss of tooth support. Several studies have corroborated the infiltration of T lymphocytes in periodontitis and correlated the infiltration with chronic inflammation in a dysregulated T cell-mediated immune response. The complexity of the disease has prompted multiple studies aiming to understand T cell-mediated pathogenesis. HIGHLIGHT Recent findings have demonstrated the pivotal role of helper T cells in many autoimmune diseases, such as rheumatoid arthritis, which has been conventionally correlated with periodontal bone loss. In contrast, the roles of helper T subsets, Th1, Th2, and particularly Th17, have not been explored. Th17-mediated pathogenesis is a significant aspect of the progression and therapy of periodontitis. CONCLUSION In this review, we highlight the complex role of Th17 in the underlying pro-inflammatory cascades mediated by a repertoire of Th17-released molecules and their role in aggravated inflammation in periodontitis. We also summarize recent therapeutics targeting Th17 and related molecules, primarily to ameliorate inflammation and maintain periodontal care.
Collapse
|
10
|
Hall BM, Verma ND, Tran GT, Hodgkinson SJ. Transplant Tolerance, Not Only Clonal Deletion. Front Immunol 2022; 13:810798. [PMID: 35529847 PMCID: PMC9069565 DOI: 10.3389/fimmu.2022.810798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
The quest to understand how allogeneic transplanted tissue is not rejected and how tolerance is induced led to fundamental concepts in immunology. First, we review the research that led to the Clonal Deletion theory in the late 1950s that has since dominated the field of immunology and transplantation. At that time many basic mechanisms of immune response were unknown, including the role of lymphocytes and T cells in rejection. These original observations are reassessed by considering T regulatory cells that are produced by thymus of neonates to prevent autoimmunity. Second, we review "operational tolerance" induced in adult rodents and larger animals such as pigs. This can occur spontaneously especially with liver allografts, but also can develop after short courses of a variety of rejection inhibiting therapies. Over time these animals develop alloantigen specific tolerance to the graft but retain the capacity to reject third-party grafts. These animals have a "split tolerance" as peripheral lymphocytes from these animals respond to donor alloantigen in graft versus host assays and in mixed lymphocyte cultures, indicating there is no clonal deletion. Investigation of this phenomenon excludes many mechanisms, including anti-donor antibody blocking rejection as well as anti-idiotypic responses mediated by antibody or T cells. This split tolerance is transferred to a second immune-depleted host by T cells that retain the capacity to effect rejection of third-party grafts by the same host. Third, we review research on alloantigen specific inhibitory T cells that led to the first identification of the CD4+CD25+T regulatory cell. The key role of T cell derived cytokines, other than IL-2, in promoting survival and expansion of antigen specific T regulatory cells that mediate transplant tolerance is reviewed. The precise methods for inducing and diagnosing operational tolerance remain to be defined, but antigen specific T regulatory cells are key mediators.
Collapse
Affiliation(s)
- Bruce M. Hall
- Immune Tolerance Laboratory, School of Medicine, University of New South Wales (UNSW) Sydney, Ingham Institute, and Renal Service and Multiple Sclerosis Clinic, Liverpool Hospital, Liverpool, NSW, Australia
| | | | | | | |
Collapse
|
11
|
Mesenchymal Stromal Cells Mediate Clinically Unpromising but Favourable Immune Responses in Kidney Transplant Patients. Stem Cells Int 2022; 2022:2154544. [PMID: 35211176 PMCID: PMC8863486 DOI: 10.1155/2022/2154544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/11/2022] [Indexed: 01/22/2023] Open
Abstract
Background Allograft rejection postkidney transplantation (KTx) is a major clinical challenge despite increased access to a healthcare system and improvement in immunosuppressive (IS) drugs. In recent years, mesenchymal stromal cells (MSCs) have aroused considerable interest in field of transplantation due to their immunomodulatory and regenerative properties. This study was aimed at investigating safety, feasibility, and immunological effects of autologous MSCs (auto-MSCs) and allogeneic MSCs (allo-MSCs) as a complement to IS drug therapy in KTx patients. Methods 10 patients undergoing KTx with a living-related donor were analysed along with 5 patients in the control group. Patients were given auto-MSCs or allo-MSCs at two time points, i.e., one day before transplant (D-0) and 30 days after transplant (D-30) at the rate of 1.0-1.5 × 106 MSCs per kg body weight in addition to immunosuppressants. Patients were followed up for 2 years, and 29 immunologically relevant lymphocyte subsets and 8 cytokines and important biomarkers were analysed at all time points. Results Patients displayed no signs of discomfort or dose-related toxicities in response to MSC infusion. Flow cytometric analysis revealed an increase in B regulatory lymphocyte populations and nonconventional T regulatory cells and a decrease in T effector lymphocyte proportions in auto-MSC-infused patients. No such favourable immune responses were observed in all MSC-infused patients. Conclusion This study provides evidence that auto-MSCs are safe and well tolerated. This is the first ever report to compare autologous and allogeneic MSC infusion in KTx patients. Importantly, our data demonstrated that MSC-induced immune responses in patients did not completely correlate with clinical outcomes. Our findings add to the current perspective of using MSCs in KTx and explore possibilities through which donor/recipient chimerism can be achieved to induce immune tolerance in KTx patients.
Collapse
|
12
|
Hall BM, Hall RM, Tran GT, Robinson CM, Wilcox PL, Rakesh PK, Wang C, Sharland AF, Verma ND, Hodgkinson SJ. Interleukin-5 (IL-5) Therapy Prevents Allograft Rejection by Promoting CD4 +CD25 + Ts2 Regulatory Cells That Are Antigen-Specific and Express IL-5 Receptor. Front Immunol 2021; 12:714838. [PMID: 34912327 PMCID: PMC8667344 DOI: 10.3389/fimmu.2021.714838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 11/01/2021] [Indexed: 12/26/2022] Open
Abstract
CD4+CD25+Foxp3+T cell population is heterogenous and contains three major sub-groups. First, thymus derived T regulatory cells (tTreg) that are naïve/resting. Second, activated/memory Treg that are produced by activation of tTreg by antigen and cytokines. Third, effector lineage CD4+CD25+T cells generated from CD4+CD25- T cells' activation by antigen to transiently express CD25 and Foxp3. We have shown that freshly isolated CD4+CD25+T cells are activated by specific alloantigen and IL-4, not IL-2, to Ts2 cells that express the IL-5 receptor alpha. Ts2 cells are more potent than naïve/resting tTreg in suppressing specific alloimmunity. Here, we showed rIL-5 promoted further activation of Ts2 cells to Th2-like Treg, that expressed foxp3, irf4, gata3 and il5. In vivo, we studied the effects of rIL-5 treatment on Lewis heart allograft survival in F344 rats. Host CD4+CD25+T cells were assessed by FACS, in mixed lymphocyte culture and by RT-PCR to examine mRNA of Ts2 or Th2-like Treg markers. rIL-5 treatment given 7 days after transplantation reduced the severity of rejection and all grafts survived ≥60d whereas sham treated rats fully rejected by day 31 (p<0.01). Treatment with anti-CD25 or anti-IL-4 monoclonal antibody abolished the benefits of treatment with rIL-5 and accelerated rejection. After 10d treatment with rIL-5, hosts' CD4+CD25+ cells expressed more Il5ra and responded to specific donor Lewis but not self. Enriched CD4+CD25+ cells from rIL-5 treated rats with allografts surviving >60 days proliferated to specific donor only when rIL-5 was present and did not proliferate to self or third party. These cells had more mRNA for molecules expressed by Th2-like Treg including Irf4, gata3 and Il5. These findings were consistent with IL-5 treatment preventing rejection by activation of Ts2 cells and Th2-like Treg.
Collapse
Affiliation(s)
- Bruce M Hall
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Rachael M Hall
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Giang T Tran
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Catherine M Robinson
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Paul L Wilcox
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Prateek K Rakesh
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Chuanmin Wang
- Transplantation Immunobiology Group, Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Alexandra F Sharland
- Transplantation Immunobiology Group, Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Nirupama D Verma
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Suzanne J Hodgkinson
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| |
Collapse
|
13
|
Qu Y, Wang X, Bai S, Niu L, Zhao G, Yao Y, Li B, Li H. The effects of TNF-α/TNFR2 in regulatory T cells on the microenvironment and progression of gastric cancer. Int J Cancer 2021; 150:1373-1391. [PMID: 34766338 PMCID: PMC9298834 DOI: 10.1002/ijc.33873] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 10/06/2021] [Accepted: 10/27/2021] [Indexed: 12/16/2022]
Abstract
TNFR2+ regulatory T cells preferentially accumulate in the tumor microenvironment, express high levels of immunosuppressive molecules and possess strong suppressive activity. Our study aimed to explore the characteristics and role of TNFR2+ Tregs in the microenvironment and progression of gastric cancer via polychromatic immunofluorescence, single-cell RNA sequencing and flow cytometry assays. The TNFR2+ Treg infiltration level in the tumor microenvironment increased significantly as gastric cancer progressed and was demonstrated to be a prognostic marker. Single-cell RNA sequencing revealed high levels of TNFR2 in tumor-infiltrating Tregs. The TNF-α/TNFR2 signaling pathway was activated, accompanied by the upregulation of costimulatory molecules. Unlike blood Tregs, tumor-infiltrating Tregs existed in activated and effector states. In addition to expressing costimulatory molecules such as TNFR2, 4-1BB, OX40 and GITR, tumor-infiltrating Tregs were also characterized by high expression levels of immune checkpoints such as CTLA-4 and TIGIT and chemokines such as CCR6. In vitro studies showed that the TNF-α/TNFR2 pathway increased the Foxp3 expression in CD4+ CD25+ T cells and the latent TGF-β production in Tregs as well as enhanced the immunosuppressive function of Tregs. In summary, our study revealed high infiltration levels of TNFR2+ Tregs that were in activated and effector states in the tumor microenvironment. The infiltration level of TNFR2+ Tregs is a prognostic marker and an independent risk factor for gastric cancer. Activation of the TNF-α/TNFR2 pathway promotes the immunosuppressive phenotype and function of Tregs. Our study provides a new theoretical basis for TNFR2+ Tregs as a therapeutic target in gastric cancer.
Collapse
Affiliation(s)
- Yang Qu
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China
| | - Xianhao Wang
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China
| | - Shuai Bai
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China
| | - Liling Niu
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China
| | - Gang Zhao
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China
| | - Yuan Yao
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China
| | - Bin Li
- National Clinical Research Center for Cancer, Tianjin, China.,Gastric Surgery Department, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hui Li
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
14
|
Tan J, Zhang QY, Huang LP, Huang K, Xie HQ. Decellularized scaffold and its elicited immune response towards the host: the underlying mechanism and means of immunomodulatory modification. Biomater Sci 2021; 9:4803-4820. [PMID: 34018503 DOI: 10.1039/d1bm00470k] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The immune response of the host towards a decellularized scaffold is complex. Not only can a number of immune cells influence this process, but also the characteristics, preparation and modification of the decellularized scaffold can significantly impact this reaction. Such factors can, together or alone, trigger immune cells to polarize towards either a pro-healing or pro-inflammatory direction. In this article, we have comprehensively reviewed factors which may influence the immune response of the host towards a decellularized scaffold, including the source of the biomaterial, biophysical properties or modifications of the scaffolds with bioactive peptides, drugs and cytokines. Furthermore, the underlying mechanism has also been recapitulated.
Collapse
Affiliation(s)
- Jie Tan
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China.
| | - Qing-Yi Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China.
| | - Li-Ping Huang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China.
| | - Kai Huang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China.
| | - Hui-Qi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China.
| |
Collapse
|
15
|
Li H, Wang C, Li X, Kong Y, Sun W. CCL17-CCR4 axis contributes to the onset of vitiligo in mice. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:702-709. [PMID: 34077992 PMCID: PMC8342221 DOI: 10.1002/iid3.423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/18/2021] [Accepted: 03/08/2021] [Indexed: 01/09/2023]
Abstract
BACKGROUND Destruction of melanocytes mediated by autoimmunity is currently believed as the main cause of vitiligo. This article aims to identify the role of CC chemokine ligand 17 (CCL17)-CC chemokine receptor 4 (CCR4) axis in vitiligo and provide new possibilities for the clinical treatment of vitiligo. METHODS A total of 30 patients with vitiligo from Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University were recruited based on the inclusion and exclusion criteria. Trephine was used to obtain skin samples from the lesion area and its surrounding normal areas, and the expression levels of CCL17, CCR4, Tbx21, Eomes, and Blimp1 were determined by quantitative reverse transcription polymerase chain reaction. Vitiligo mouse model was established by adoptively transferring CFP-PMEL CD8+ T cells into sublethally irradiated Krt14-Kitl* mice. Recipient mice received intraperitoneal injection of 1 × 106 plaque-forming units of rVV-hPMEL on the same day of transfer. The degree of depigmentation was scored blindly by one observer 5 weeks after vitiligo induction. CFP-PMEL CD8+ T cells migration to skin, draining lymph nodes, spleen, and blood were detected by flow cytometry. CCR4 blockade was performed by intraperitoneal injection of neutralizing antibody. RESULTS The expression levels of CCL17, CCR4, Tbx21, Eomes, and Blimp1 in skin lesions were significantly increased compared with that in surrounding normal areas. CCL17-/- and CCR4-/- mice exhibited significantly lower disease scores than WT mice. The CFP-PMEL CD8+ T cells accumulation was significantly decreased in the skin of CCL17-/- and CCR4-/- mice, but was not changed in draining lymph nodes, spleen, and blood. Administration of CCR4 neutralizing antibody decreased the degree of depigmentation and the recruitment of CFP-PMEL CD8+ T cells to the skin, while keeping the number of T cells in draining lymph nodes unchanged. CONCLUSION Targeting CCL17-CCR4 axis might inhibit T cell migrating to skin and alleviate vitiligo progression.
Collapse
Affiliation(s)
- He Li
- Department of Dermatology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Congpin Wang
- Department of Pharmacy, Eye Ear Nose & Throat Hospital, Fudan University, Shanghai, China
| | - Xiaoqing Li
- Department of Dermatology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yinghui Kong
- Department of Dermatology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Weiguo Sun
- Department of Dermatology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| |
Collapse
|
16
|
Multiple sclerosis patients have reduced resting and increased activated CD4 +CD25 +FOXP3 +T regulatory cells. Sci Rep 2021; 11:10476. [PMID: 34006899 PMCID: PMC8131694 DOI: 10.1038/s41598-021-88448-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/01/2021] [Indexed: 12/26/2022] Open
Abstract
Resting and activated subpopulations of CD4+CD25+CD127loT regulatory cells (Treg) and CD4+CD25+CD127+ effector T cells in MS patients and in healthy individuals were compared. Peripheral blood mononuclear cells isolated using Ficoll Hypaque were stained with monoclonal antibodies and analysed by flow cytometer. CD45RA and Foxp3 expression within CD4+ cells and in CD4+CD25+CD127loT cells identified Population I; CD45RA+Foxp3+, Population II; CD45RA−Foxp3hi and Population III; CD45RA−Foxp3+ cells. Effector CD4+CD127+ T cells were subdivided into Population IV; memory /effector CD45RA− CD25−Foxp3− and Population V; effector naïve CD45RA+CD25−Foxp3−CCR7+ and terminally differentiated RA+ (TEMRA) effector memory cells. Chemokine receptor staining identified CXCR3+Th1-like Treg, CCR6+Th17-like Treg and CCR7+ resting Treg. Resting Treg (Population I) were reduced in MS patients, both in untreated and treated MS compared to healthy donors. Activated/memory Treg (Population II) were significantly increased in MS patients compared to healthy donors. Activated effector CD4+ (Population IV) were increased and the naïve/ TEMRA CD4+ (Population V) were decreased in MS compared to HD. Expression of CCR7 was mainly in Population I, whereas expression of CCR6 and CXCR3 was greatest in Populations II and intermediate in Population III. In MS, CCR6+Treg were lower in Population III. This study found MS is associated with significant shifts in CD4+T cells subpopulations. MS patients had lower resting CD4+CD25+CD45RA+CCR7+ Treg than healthy donors while activated CD4+CD25hiCD45RA−Foxp3hiTreg were increased in MS patients even before treatment. Some MS patients had reduced CCR6+Th17-like Treg, which may contribute to the activity of MS.
Collapse
|
17
|
Identifying the Immunological Gene Signatures of Immune Cell Subtypes. BIOMED RESEARCH INTERNATIONAL 2021. [DOI: 10.1155/2021/6639698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The immune system is a complicated defensive system that comprises multiple functional cells and molecules acting against endogenous and exogenous pathogenic factors. Identifying immune cell subtypes and recognizing their unique immunological functions are difficult because of the complicated cellular components and immunological functions of the immune system. With the development of transcriptomics and high-throughput sequencing, the gene expression profiling of immune cells can provide a new strategy to explore the immune cell subtyping. On the basis of the new profiling data of mouse immune cell gene expression from the Immunological Genome Project (ImmGen), a novel computational pipeline was applied to identify different immune cell subtypes, including αβ T cells, B cells, γδ T cells, and innate lymphocytes. First, the profiling data was analyzed by a powerful feature selection method, Monte-Carlo Feature Selection, resulting in a feature list and some informative features. For the list, the two-stage incremental feature selection method, incorporating random forest as the classification algorithm, was applied to extract essential gene signatures and build an efficient classifier. On the other hand, a rule learning scheme was applied on the informative features to construct quantitative expression rules. A group of gene signatures was found as qualitatively related to the biological processes of four immune cell subtypes. The quantitative expression rules can efficiently cluster immune cells. This work provides a novel computational tool for immune cell quantitative subtyping and biomarker recognition.
Collapse
|
18
|
Fedorka CE, El-Sheikh Ali H, Walker OF, Scoggin KE, Dini P, Loux SC, Troedsson MHT, Ball BA. The imbalance of the Th17/Treg axis following equine ascending placental infection. J Reprod Immunol 2021; 144:103268. [PMID: 33454392 DOI: 10.1016/j.jri.2020.103268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/12/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023]
Abstract
Ascending placentitis is a leading cause of abortion in the horse, but adaptive immune response to this disease is unknown. To evaluate this, sub-acute placentitis was experimentally-induced via trans-cervical inoculation of S. zooepidemicus, and endometrium and chorioallantois was collected 8 days later (n = 6 inoculated/n = 6 control). The expression of transcripts relating to Th1, Th2, Th17, and Treg maturation was assessed via RNASeq. IHC of transcription factors relating to each subtype in the same tissues (Th1: TBX21, Th2: GATA3, Th17: IRF4, Treg: FOXp3). An immunoassay was utilized to assess circulating cytokines (Th1: IFNg, IL-2; Th2: IL-4, IL-5; Th17: IL-17, IL-6; Treg: IL-10, GM-CSF). An increase in Th1 and Th17-related transcripts were noted in the chorioallantois, although no alterations were seen in the endometrium. Th2 and Treg-related transcripts altered in a dysregulated manner, as some transcripts increased in expression while others decreased. Immunolocalization of Th1, Th2, and Th17 cells was increased in diseased chorioallantois, while no Treg cells were noted in the diseased tissue. Secreted cytokines relating to Th1 (IFNg, IL-2), Th17 (IL-6), Th2 (IL-5), and Treg (IL-10) populations increased in maternal circulation eight days after inoculation. In conclusion, the Th1/Th17 response to ascending placentitis occurs primarily in the chorioallantois, indicating the adaptive immune response to occur in fetal derived placental tissue. Additionally, ascending placentitis leads to an increase in the helper T cell populations (Th1/Th17/Th2) while decreasing the Treg response. This increase in Th17-related responses alongside a diminishing Treg-related response may precede or contribute to fetal demise, abortion, or preterm labor.
Collapse
Affiliation(s)
- C E Fedorka
- University of Kentucky, Department of Veterinary Sciences. Lexington KY, USA
| | - H El-Sheikh Ali
- University of Kentucky, Department of Veterinary Sciences. Lexington KY, USA; University of Mansoura, Department of Theriogenology, Dakahlia, Egypt
| | - O F Walker
- Lincoln Memorial University, College of Veterinary Medicine, Harrogate TN, USA
| | - K E Scoggin
- University of Kentucky, Department of Veterinary Sciences. Lexington KY, USA
| | - P Dini
- University of Kentucky, Department of Veterinary Sciences. Lexington KY, USA; University of California, Davis, Department of Population Health and Reproduction, Davis, CA, USA
| | - S C Loux
- University of Kentucky, Department of Veterinary Sciences. Lexington KY, USA
| | - M H T Troedsson
- University of Kentucky, Department of Veterinary Sciences. Lexington KY, USA
| | - B A Ball
- University of Kentucky, Department of Veterinary Sciences. Lexington KY, USA.
| |
Collapse
|
19
|
Gu XX, Jin Y, Fu T, Zhang XM, Li T, Yang Y, Li R, Zhou W, Guo JX, Zhao R, Li JJ, Dong C, Gu ZF. Relevant Characteristics Analysis Using Natural Language Processing and Machine Learning Based on Phenotypes and T-Cell Subsets in Systemic Lupus Erythematosus Patients With Anxiety. Front Psychiatry 2021; 12:793505. [PMID: 34955935 PMCID: PMC8703039 DOI: 10.3389/fpsyt.2021.793505] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/09/2021] [Indexed: 11/13/2022] Open
Abstract
Anxiety is frequently observed in patients with systemic lupus erythematosus (SLE) and the immune system could act as a trigger for anxiety. To recognize abnormal T-cell and B-cell subsets for SLE patients with anxiety, in this study, patient disease phenotypes data from electronic lupus symptom records were extracted by using natural language processing. The Hospital Anxiety and Depression Scale (HADS) was used to distinguish patients, and 107 patients were selected to meet research requirements. Then, peripheral blood was collected from two patient groups for multicolor flow cytometry experiments. The characteristics of 75 T-cell and 15 B-cell subsets were investigated between SLE patients with- (n = 23) and without-anxiety (n = 84) groups by four machine learning methods. The findings showed 13 T-cell subsets were significantly different between the two groups. Furthermore, BMI, fatigue, depression, unstable emotions, CD27+CD28+ Th/Treg, CD27-CD28- Th/Treg, CD45RA-CD27- Th, and CD45RA+HLADR+ Th cells may be important characteristics between SLE patients with- and without-anxiety groups. The findings not only point out the difference of T-cell subsets in SLE patients with or without anxiety, but also imply that T cells might play the important role in patients with anxiety disorder.
Collapse
Affiliation(s)
- Xi-Xi Gu
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong University, Nantong, China.,Joint Research Center, Affiliated Hospital of Nantong University, Nantong, China.,Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Yi Jin
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong University, Nantong, China.,Joint Research Center, Affiliated Hospital of Nantong University, Nantong, China.,Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Ting Fu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiao-Ming Zhang
- Joint Research Center, Affiliated Hospital of Nantong University, Nantong, China.,Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Teng Li
- Joint Research Center, Affiliated Hospital of Nantong University, Nantong, China.,Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Ying Yang
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Rong Li
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong University, Nantong, China.,Joint Research Center, Affiliated Hospital of Nantong University, Nantong, China.,Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Wei Zhou
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong University, Nantong, China.,Joint Research Center, Affiliated Hospital of Nantong University, Nantong, China.,Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Jia-Xin Guo
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong University, Nantong, China.,Joint Research Center, Affiliated Hospital of Nantong University, Nantong, China.,Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Rui Zhao
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Jing-Jing Li
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Chen Dong
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong University, Nantong, China.,Joint Research Center, Affiliated Hospital of Nantong University, Nantong, China.,Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Zhi-Feng Gu
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong University, Nantong, China.,Joint Research Center, Affiliated Hospital of Nantong University, Nantong, China.,Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
20
|
Jia J, Mo X, Yan F, Liu J, Ye S, Zhang Y, Lin Y, Li H, Chen D. Role of YAP-related T cell imbalance and epidermal keratinocyte dysfunction in the pathogenesis of atopic dermatitis. J Dermatol Sci 2020; 101:164-173. [PMID: 33358580 DOI: 10.1016/j.jdermsci.2020.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/16/2020] [Accepted: 12/16/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) is characterized by impaired skin barrier function and immune system dysfunction. The expression and role of Yes-associated protein (YAP) in AD are unclear. OBJECTIVE To characterize the role of the YAP in T cell imbalance and epidermal keratinocyte dysfunction in the pathogenesis of AD. METHODS We included 35 patients with AD (21 acute and 14 chronic). An AD mouse model was constructed using 2,4-dinitrofluorobenzene, and AD-like inflammatory cell model was constructed using TNF-α/IFN-γ-activated HaCaT cells. The proportion of Th1/Th2/Th17/Treg cells was detected using flow cytometry. After mononuclear cells were obtained from human peripheral blood or mouse spleen and induced to differentiate into different T cell subsets, YAP mRNA and protein expression were analyzed. Up-regulation of YAP was induced by lentivirus and down-regulation of YAP was induced by its specific inhibitor verteporfin (VP). The expression of YAP in skin lesions and infiltrating T cell subsets was detected using immunohistochemistry and double immunofluorescence staining, respectively. RESULTS We found differing degrees of Th1/Th2/Th17/Treg imbalance in acute and chronic AD. YAP expression was downregulated in Treg cells and upregulated in Th17 cells; YAP expression was downregulated in the AD epidermis. After YAP overexpression, the proportion of both Th17 and the Treg cells differentiated from mouse spleen mononuclear cells increased. There was an opposite trend after YAP inhibition. The proliferation and migration decreased and apoptosis increased after YAP inhibition in HaCaT cells. CONCLUSION Change of YAP expression may cause T cell imbalance and hamper the healing of the epidermis in AD.
Collapse
Affiliation(s)
- Jinjing Jia
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, China
| | - Xiumei Mo
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, China
| | - Fenggen Yan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, China
| | - Junfeng Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, China
| | - Siqi Ye
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, China
| | - Yu Zhang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, China
| | - Ying Lin
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, China
| | - Hongyi Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, China
| | - Dacan Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, China.
| |
Collapse
|
21
|
Lymphocyte Immunosuppression and Dysfunction Contributing to Persistent Inflammation, Immunosuppression, and Catabolism Syndrome (PICS). Shock 2020; 55:723-741. [PMID: 33021569 DOI: 10.1097/shk.0000000000001675] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
ABSTRACT Persistent Inflammation, Immune Suppression, and Catabolism Syndrome (PICS) is a disease state affecting patients who have a prolonged recovery after the acute phase of a large inflammatory insult. Trauma and sepsis are two pathologies after which such an insult evolves. In this review, we will focus on the key clinical determinants of PICS: Immunosuppression and cellular dysfunction. Currently, relevant immunosuppressive functions have been attributed to both innate and adaptive immune cells. However, there are significant gaps in our knowledge, as for trauma and sepsis the immunosuppressive functions of these cells have mostly been described in acute phase of inflammation so far, and their clinical relevance for the development of prolonged immunosuppression is mostly unknown. It is suggested that the initial immune imbalance determines the development of PCIS. Additionally, it remains unclear what distinguishes the onset of immune dysfunction in trauma and sepsis and how this drives immunosuppression in these cells. In this review, we will discuss how regulatory T cells (Tregs), innate lymphoid cells, natural killer T cells (NKT cells), TCR-a CD4- CD8- double-negative T cells (DN T cells), and B cells can contribute to the development of post-traumatic and septic immunosuppression. Altogether, we seek to fill a gap in the understanding of the contribution of lymphocyte immunosuppression and dysfunction to the development of chronic immune disbalance. Further, we will provide an overview of promising diagnostic and therapeutic interventions, whose potential to overcome the detrimental immunosuppression after trauma and sepsis is currently being tested.
Collapse
|
22
|
Amada E, Fukuda K, Kumagai K, Kawakubo H, Kitagawa Y. Soluble recombinant human thrombomodulin suppresses inflammation-induced gastrointestinal tumor growth in a murine peritonitis model. Mol Cell Biochem 2020; 475:195-203. [PMID: 32767229 DOI: 10.1007/s11010-020-03872-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 08/01/2020] [Indexed: 12/27/2022]
Abstract
Regulatory T cells (Tregs) and transforming growth factor β (TGF-β) are believed to play key roles in both postoperative pro-inflammatory and anti-inflammatory responses of malignancies. Recombinant human thrombomodulin (rTM) is implied to inhibit the interaction between TGF-β and Tregs. The aim of this study is to evaluate the antitumor effects of rTM against gastrointestinal tumors under systemic inflammation. Mice were subjected to cecal ligation and puncture and percutaneous allogeneic tumor implantation. rTM were introduced by percutaneous injection into the abdominal cavity. The effects of rTM were evaluated by weight of implanted tumor, proportion of Tregs in peripheral blood lymphocytes (PBL) and tumor infiltrating lymphocytes (TIL) and temporal evaluation of serum cytokines. The effect of rTM was also evaluated on the in vitro differentiation of naïve T cells into induced Tregs induced by TGF-β and interleukin (IL) -2. rTM significantly inhibited the proliferation of the implanted tumor cells in an inflammation-dependent manner. rTM also reduced the fractions of regulatory T cells and induced regulatory T cells among both PBL and TIL. Temporal evaluation of serum cytokine levels in the model mice showed that rTM significantly suppressed the increases in the serum levels of IL-2 and TGF-β. An in vitro differentiation assay revealed that rTM inhibited the differentiation of naïve T cells into Tregs triggered by IL-2- and TGF-β. rTM has suppressive effects on inflammation-induced gastrointestinal tumor growth by suggestively affecting differentiation of Tregs.
Collapse
Affiliation(s)
- En Amada
- Department of Surgery, School of Medicine, Keio University, 35, Shinanomachi, Shinjuku, Tokyo, Japan
| | - Kazumasa Fukuda
- Department of Surgery, School of Medicine, Keio University, 35, Shinanomachi, Shinjuku, Tokyo, Japan
| | - Koshi Kumagai
- Department of Gastroenterological Surgery, The Cancer Institute Hospital of JFCR, 3-8-31, Ariake, Koto, Tokyo, Japan.
| | - Hirofumi Kawakubo
- Department of Surgery, School of Medicine, Keio University, 35, Shinanomachi, Shinjuku, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, School of Medicine, Keio University, 35, Shinanomachi, Shinjuku, Tokyo, Japan
| |
Collapse
|
23
|
McLendon BA, Seo H, Kramer AC, Burghardt RC, Bazer FW, Johnson GA. Pig conceptuses secrete interferon gamma to recruit T cells to the endometrium during the peri-implantation period†. Biol Reprod 2020; 103:1018-1029. [PMID: 32716497 DOI: 10.1093/biolre/ioaa132] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 02/07/2023] Open
Abstract
The emerging paradigm in the immunology of pregnancy is that implantation of conceptuses does not progress in an immunologically suppressed environment. Rather, the endometrium undergoes a controlled inflammatory response during implantation as trophectoderm of elongating and implanting pig conceptuses secrete the pro-inflammatory cytokine interferon gamma (IFNG). Results of this study with pigs revealed: (1) accumulation of immune cells and apoptosis of stromal cells within the endometrium at sites of implantation during the period of IFNG secretion by conceptuses; (2) accumulation of proliferating cell nuclear antigen (PCNA)-positive T cells within the endometrium at sites of implantation; (3) significant increases in expression of T cell co-signaling receptors including programmed cell death 1 (PDCD1), CD28, cytotoxic T-lymphocyte associated protein 4 (CTLA-4), and inducible T cell co-stimulator (ICOS), as well as chemokines CXCL9, 10, and 11 within the endometrium at sites of implantation; (4) significant increases in T cell co-signaling receptors, PDCD1 and ICOS, and chemokine CXCL9 in the endometrium of cyclic gilts infused with IFNG; and (5) identification of CD4+ (22.59%) as the major T cell subpopulation, with minor subpopulations of CD8+ (1.38%), CD4+CD25+ (1.08%), and CD4+CD8+ (0.61%) T cells within the endometrium at sites of implantation. Our results provide new insights into the immunology of implantation to suggest that trophectoderm cells of pigs secrete IFNG to recruit various subpopulations of T cells to the endometrium to contribute to a controlled inflammatory environment that supports the active breakdown and restructuring of the endometrium in response to implantation of the conceptus.
Collapse
Affiliation(s)
- Bryan A McLendon
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, College Station, TX, USA
| | - Heewon Seo
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, College Station, TX, USA
| | - Avery C Kramer
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, College Station, TX, USA
| | - Robert C Burghardt
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, College Station, TX, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Greg A Johnson
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, College Station, TX, USA
| |
Collapse
|
24
|
Zimmerman KA, Hopp K, Mrug M. Role of chemokines, innate and adaptive immunity. Cell Signal 2020; 73:109647. [PMID: 32325183 DOI: 10.1016/j.cellsig.2020.109647] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/16/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023]
Abstract
Polycystic Kidney Disease (PKD) triggers a robust immune system response including changes in both innate and adaptive immunity. These changes involve immune cells (e.g., macrophages and T cells) as well as cytokines and chemokines (e.g., MCP-1) that regulate the production, differentiation, homing, and various functions of these cells. This review is focused on the role of the immune system and its associated factors in the pathogenesis of PKDs as evidenced by data from cell-based systems, animal models, and PKD patients. It also highlights relevant pre-clinical and clinical studies that point to specific immune system components as promising candidates for the development of prognostic biomarkers and therapeutic strategies to improve PKD outcomes.
Collapse
Affiliation(s)
- Kurt A Zimmerman
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Division of Nephrology, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Katharina Hopp
- Department of Medicine, Division of Renal Diseases and Hypertension, Polycystic Kidney Disease Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michal Mrug
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Department of Veterans Affairs Medical Center, Birmingham, AL 35233, USA.
| |
Collapse
|
25
|
Martinez PS, Pucheu CM, Liu CC, Carter RT. Cytokine tear film profile determination in eyes of healthy dogs and those with inflammatory periocular and skin disorders. Vet Immunol Immunopathol 2020; 221:110012. [PMID: 31978678 DOI: 10.1016/j.vetimm.2020.110012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/26/2019] [Accepted: 01/14/2020] [Indexed: 12/20/2022]
Abstract
Alterations in serum cytokine levels and profiles have been reported in association with a variety of disease conditions (e.g., allergic, immune-mediated, etc.) in both humans and animals. In comparison to serum cytokine measurements, tear cytokine measurements might be expected to more accurately reflect the inflammatory milieu associated with periocular disease. The purpose of this study was to use a multiplexed assay to compare the cytokine profile of tears in healthy dogs to those with inflammatory skin and periocular disease. We were able to detect IL-2, IL-6, IL-8, and TNF-α in >47 % of tear samples from both healthy canine patients and those with inflammatory dermatologic disease (with or without concurrent periocular involvement). In contrast, IL-7, IL-10 and IFN-γ were rarely detected. Dogs with both dermatologic and periocular disease (but not dermatologic disease alone) had higher levels of IL-8 (P < 0.001, P > 0.05, respectively) relative to healthy dogs. Patients with concurrent dermatologic and periocular disease also demonstrated significantly greater variability in IL-8 concentrations between eyes than did healthy dogs (P < 0.0001). Our findings suggest that tear cytokine analysis may prove to be a useful tool to investigate the role and interactions of the local ocular immune response in patients with inflammatory periocular disease.
Collapse
Affiliation(s)
- P S Martinez
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States
| | - C M Pucheu
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States
| | - C C Liu
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States
| | - R T Carter
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States.
| |
Collapse
|
26
|
Zhu X, Wang X, Wang Y, Zhao Y. Exosomal long non-coding RNA GAS5 suppresses Th1 differentiation and promotes Th2 differentiation via downregulating EZH2 and T-bet in allergic rhinitis. Mol Immunol 2019; 118:30-39. [PMID: 31841965 DOI: 10.1016/j.molimm.2019.11.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/19/2019] [Accepted: 11/25/2019] [Indexed: 12/18/2022]
Abstract
The imbalance of helper T cell (Th) 1/Th2 differentiation is involved in the development of allergic rhinitis (AR). Recent studies reveal the regulatory function of exosomes on Th1/Th2 differentiation. However, the key mediator in exosomes that modulate such response remains unclear. In this study, the expression of long-noncoding RNA GAS5 (LncGAS5) was detected in exosomes which were isolated from AR patient nasal mucus (AR-EXO) and ovalbumin (OVA)-stimulated nasal epithelial cells (OVA-EXO). Th1/Th2 differentiation was induced in naïve CD4+ T cells, and the percentage of IFN-γ expressing cells (Th1 cells) and IL-4 expressing cells (Th2 cells) was detected using flow cytometry. The result showed that LncGAS5 was upregulated in AR epithelial samples, AR-EXO, and OVA-EXO. The coincubation of AR-EXO and CD4+ T cells suppressed Th1 differentiation and promoted Th2 differentiation, which is mediated by LncGAS5 in AR-EXO. The LncGAS5 in AR-EXO inhibited transcription and expression of EZH2, and it also inhibited T-bet expression at mRNA and protein levels. The gain-of-function and loss-of-function experiments suggested that LncGAS5 mediates Th1/Th2 differentiation partly through downregulating T-bet and EZH2. In summary, our findings demonstrated that LncGAS5 in AR epithelium-derived exosomes is the key mediator in Th1/Th2 differentiation, providing a possible therapeutic target of AR.
Collapse
Affiliation(s)
- Xiaoyuan Zhu
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Xueping Wang
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Ying Wang
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Yulin Zhao
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
27
|
Fedorka CE, Loux SL, Scoggin KE, Adams AA, Troedsson MHT, Ball BA. Alterations in T cell-related transcripts at the feto-maternal interface throughout equine gestation. Placenta 2019; 89:78-87. [PMID: 31730925 DOI: 10.1016/j.placenta.2019.10.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 10/21/2019] [Accepted: 10/26/2019] [Indexed: 12/23/2022]
Abstract
INTRODUCTION The tolerance of pregnancy by the maternal immune system is balanced between recognition and protection. In the human this is controlled by balancing helper T cell populations (Th1, Th2) in addition to immune suppression from the regulatory arm (Tregs), but this has not been evaluated in the horse. METHODS RNA sequencing was performed on chorioallantois and endometrium of mares at 120, 180, 300 and 330 days of gestation (n = 4/stage), as well as 45-day chorioallantois (n = 4) and diestrus endometrium (n = 3). Transcripts were selected for relativity to Th1, Th2, or Treg-associated. qPCR and immunohistochemistry were used to confirm the results of select differentially expressed genes. RESULTS In the endometrium, Th1 transcripts were highest in the diestrus mare and decreased as gestational length progressed. In contrast, Th2 transcripts were upregulated in comparison to the diestrus mare and highest in mid gestation. Treg transcripts were found increased in comparison to the diestrus mare, but decreased prepartum. In the chorioallantois no Th1 transcripts changed. The majority of Th2 transcripts increased from 45 to 300 days gestation, and then decreased prepartum. Treg-related transcripts trended down in the chorioallantois from 45 days to 120 days gestation, followed by an upregulation to 300 days and a secondary decline prepartum. DISCUSSION The mare experiences a complex and evolving immune profile within the tissues of the feto-maternal interface. This consists of a balance between the Th1 and Th2 response, and a dynamic Treg response that is hypothesized to regulate overall events within the immune system.
Collapse
Affiliation(s)
- C E Fedorka
- Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - S L Loux
- Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - K E Scoggin
- Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - A A Adams
- Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - M H T Troedsson
- Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - B A Ball
- Department of Veterinary Science, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
28
|
Guo PL, Li LH, Li WL, Zhao JC, Hu FY, Zhang FC, Cai WP, Tang XP. The clinical significance of myeloid-derived suppressor cells in dengue fever patients. BMC Infect Dis 2019; 19:926. [PMID: 31675923 PMCID: PMC6824033 DOI: 10.1186/s12879-019-4574-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 10/16/2019] [Indexed: 12/30/2022] Open
Abstract
Background Myeloid-derived suppressor cells (MDSCs) play immunosuppressive roles in cancers and some infectious diseases; however, their role in dengue fever (DF) remains unknown. This study evaluated the clinical significance of MDSCs in DF patients. Methods This study comprised 178 non-severe DF patients, 20 non-dengue fever (NDF) controls, and 30 healthy donors. The DF patients were divided into the following five groups based on the fever duration from its onset to the day of sample collection: fever duration of 1–2, 3–4, 5–6, 7–8, and > 9 days. Among these DF patients, 14 were monitored for eight days, and their peripheral blood samples were collected every two days. The mononuclear cells were isolated and analyzed using flow cytometry. The correlation between the MDSCs and clinical and immunological indicators of the DF patients was evaluated using Spearman analysis. Results The count of the peripheral blood MDSCs, especially monocytic MDSCs, of the 178 DF patients were dramatically higher than those of the NDF and healthy controls, and remarkably decreased with the fever duration. Moreover, the MDSC count correlated with some indicators, including the dengue viral load (rho = 0.367, p < .001), body temperature (rho = 0.263, p = .005), prothrombin time (rho = 0.475, p < .001), CD4+ T cell number (rho = − 0.317, p < .001), CD8+ T cell number (rho = − 0.361, p < .001), “programmed cell death protein 1” (PD-1) (rho = − 0.347, p < .001), “T cell immunoglobulin domain and mucin domain-3” (Tim3) (rho = − 0.258, p = .001), interferon-α (IFN-α) (rho = 0.43, p < .001), and “regulated upon activation normal T-cell expressed and secreted” (RANTES) (rho = 0.278, p = .019). Furthermore, the level of arginase-1, but not nitric oxide, was higher in the DF patients than in the healthy controls and was closely related to the number of MDSCs (rho = 0.265, p = .024). Conclusions Our study reveals a significant correlation between MDSCs and DF clinical indicators, posing MDSCs as potential target cells for DF treatment.
Collapse
Affiliation(s)
- Peng-Le Guo
- Department of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, Guangdong Province, China
| | - Ling-Hua Li
- Department of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, Guangdong Province, China
| | - Wen-Li Li
- Department of Infectious Diseases, Guangdong Second People's Hospital, Guangzhou, 510317, Guangdong Province, China
| | - Jin-Cun Zhao
- Department of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, Guangdong Province, China.,Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, Guangdong Province, China
| | - Feng-Yu Hu
- Department of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, Guangdong Province, China
| | - Fu-Chun Zhang
- Department of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, Guangdong Province, China
| | - Wei-Ping Cai
- Department of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, Guangdong Province, China
| | - Xiao-Ping Tang
- Department of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, Guangdong Province, China.
| |
Collapse
|
29
|
Czarnowicki T, He H, Canter T, Han J, Lefferdink R, Erickson T, Rangel S, Kameyama N, Kim HJ, Pavel AB, Estrada Y, Krueger JG, Paller AS, Guttman-Yassky E. Evolution of pathologic T-cell subsets in patients with atopic dermatitis from infancy to adulthood. J Allergy Clin Immunol 2019; 145:215-228. [PMID: 31626841 DOI: 10.1016/j.jaci.2019.09.031] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 08/23/2019] [Accepted: 09/12/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND The circulating immune phenotype was defined in adults and young children with early atopic dermatitis (AD), but chronologic changes in the blood of infants and children with AD through adolescence have not been explored. OBJECTIVE We sought to compare immune activation and cytokine polarization in the blood of 0- to 5-year-old (n = 39), 6- to 11-year-old (n = 26), 12- to 17-year-old (n = 21) and 18-year-old or older (n = 43) patients with AD versus age-matched control subjects. METHODS Flow cytometry was used to measure IFN-γ, IL-9, IL-13, IL-17, and IL-22 cytokine levels in CD4+/CD8+ T cells, with inducible costimulator molecule and HLA-DR defining midterm and long-term T-cell activation, respectively, within skin-homing/cutaneous lymphocyte antigen (CLA)+ versus systemic/CLA- T cells. Unsupervised clustering differentiated patients based on their blood biomarker frequencies. RESULTS Although CLA+ TH1 frequencies were significantly lower in infants with AD versus all older patients (P < .01), frequencies of CLA+ TH2 T cells were similarly expanded across all AD age groups compared with control subjects (P < .05). After infancy, CLA- TH2 frequencies were increased in patients with AD in all age groups, suggesting systemic immune activation with disease chronicity. IL-22 frequencies serially increased from normal levels in infants to highly significant levels in adolescents and adults compared with levels in respective control subjects (P < .01). Unsupervised clustering aligned the AD profiles along an age-related spectrum from infancy to adulthood (eg, inducible costimulator molecule and IL-22). CONCLUSIONS The adult AD phenotype is achieved only in adulthood. Unique cytokine signatures characterizing individual pediatric endotypes might require age-specific therapies. Future longitudinal studies, comparing the profile of patients with cleared versus persistent pediatric AD, might define age-specific changes that predict AD clearance.
Collapse
Affiliation(s)
- Tali Czarnowicki
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Helen He
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Talia Canter
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Joseph Han
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rachel Lefferdink
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Taylor Erickson
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Stephanie Rangel
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Naoya Kameyama
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Hyun Je Kim
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ana B Pavel
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yeriel Estrada
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - James G Krueger
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Amy S Paller
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Emma Guttman-Yassky
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
30
|
Kato H, Igarashi K. To be red or white: lineage commitment and maintenance of the hematopoietic system by the "inner myeloid". Haematologica 2019; 104:1919-1927. [PMID: 31515352 PMCID: PMC6886412 DOI: 10.3324/haematol.2019.216861] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/10/2019] [Indexed: 12/21/2022] Open
Abstract
Differentiation of hematopoietic stem and progenitor cells is tightly regulated depending on environmental changes in order to maintain homeostasis. Transcription factors direct the development of hematopoietic cells, such as GATA-1 for erythropoiesis and PU.1 for myelopoiesis. However, recent findings obtained from single-cell analyses raise the question of whether these transcription factors are "initiators" or just "executors" of differentiation, leaving the initiation of hematopoietic stem and progenitor cell differentiation (i.e. lineage commitment) unclear. While a stochastic process is likely involved in commitment, it cannot fully explain the homeostasis of hematopoiesis nor "on-demand" hematopoiesis in response to environmental changes. Transcription factors BACH1 and BACH2 may regulate both commitment and on-demand hematopoiesis because they control erythroid-myeloid and lymphoid-myeloid differentiation by repressing the myeloid program, and their activities are repressed in response to infectious and inflammatory conditions. We summarize possible mechanisms of lineage commitment of hematopoietic stem and progenitor cells suggested by recent findings and discuss the erythroid and lymphoid commitment of hematopoietic stem and progenitor cells, focusing on the gene regulatory network composed of genes encoding key transcription factors. Surprising similarity exists between commitment to erythroid and lymphoid lineages, including repression of the myeloid program by BACH factors. The suggested gene regulatory network of BACH factors sheds light on the myeloid-based model of hematopoiesis. This model will help to understand the tuning of hematopoiesis in higher eukaryotes in the steady-state condition as well as in emergency conditions, the evolutional history of the system, aging and hematopoietic disorders.
Collapse
Affiliation(s)
- Hiroki Kato
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Present address, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
31
|
Liu XM, Liu Y, Yu S, Jiang LM, Song B, Chen X. Potential immunomodulatory effects of stem cells from the apical papilla on Treg conversion in tissue regeneration for regenerative endodontic treatment. Int Endod J 2019; 52:1758-1767. [DOI: 10.1111/iej.13197] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023]
Affiliation(s)
- X. M. Liu
- Department of Paediatric Dentistry School of Stomatology China Medical University ShenyangChina
- Liaoning Province Key Laboratory of Oral Disease ShenyangChina
- State Key Laboratory of Military Stomatology Xi'an China
| | - Y. Liu
- Department of Paediatric Dentistry School of Stomatology China Medical University ShenyangChina
- Liaoning Province Key Laboratory of Oral Disease ShenyangChina
- State Key Laboratory of Military Stomatology Xi'an China
| | - S. Yu
- Department of Paediatric Dentistry School of Stomatology China Medical University ShenyangChina
- Liaoning Province Key Laboratory of Oral Disease ShenyangChina
| | - L. M. Jiang
- Department of Paediatric Dentistry School of Stomatology China Medical University ShenyangChina
- Liaoning Province Key Laboratory of Oral Disease ShenyangChina
| | - B. Song
- School of Dentistry Cardiff University Cardiff UK
| | - X. Chen
- Department of Paediatric Dentistry School of Stomatology China Medical University ShenyangChina
- Liaoning Province Key Laboratory of Oral Disease ShenyangChina
| |
Collapse
|
32
|
Cormican S, Griffin MD. The Complex Role of Interleukin 6 in Regulating T-cell Responses during Acute Glomerulonephritis. J Am Soc Nephrol 2019; 30:1341-1344. [PMID: 31366693 DOI: 10.1681/asn.2019050453] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Sarah Cormican
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland; and.,Nephrology Services, Galway University Hospitals, Saolta University Health Group, Galway, Ireland
| | - Matthew D Griffin
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland; and .,Nephrology Services, Galway University Hospitals, Saolta University Health Group, Galway, Ireland
| |
Collapse
|
33
|
Shimizu T, Kamata M, Fukaya S, Hayashi K, Fukuyasu A, Tanaka T, Ishikawa T, Ohnishi T, Tada Y. Anti-IL-17A and IL-23p19 antibodies but not anti-TNFα antibody induce expansion of regulatory T cells and restoration of their suppressive function in imiquimod-induced psoriasiform dermatitis. J Dermatol Sci 2019; 95:90-98. [PMID: 31362906 DOI: 10.1016/j.jdermsci.2019.07.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 07/01/2019] [Accepted: 07/18/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Psoriasis is a chronic inflammatory skin disease. Anti-TNFα, IL-17A and IL-23p19 antibodies are effective for psoriasis. However, the contribution of regulatory T cells (Treg) in their effectiveness remains to be elucidated. OBJECTIVE We investigated the effects of TNFα, IL-17A and IL-23p19 inhibition on Tregs in imiquimod-induced psoriasiform dermatitis. METHODS Psoriasiform dermatitis was induced by imiquimod application on murine shaved back skin for six days. Mice were treated with anti-TNFα, IL-17A or IL-23p19 monoclonal antibodies every other day from one day before imiquimod application. RESULTS Administration of anti-TNFα, IL-17A or IL-23p19 antibodies improved the clinical score and downregulated Th17-related cytokines and chemokines, while IL-23p19 antibodies upregulated IL-10 mRNA expression. Anti-IL-17A or IL-23p19 antibody-treated imiquimod-applied mice showed a significant increase in the number of Foxp3+ IL-10+ Tregs. Recipient mice adoptively transferred with Tregs derived from donor mice treated with antibodies demonstrated clinical and pathological improvement in imiquimod-induced psoriasiform dermatitis. Anti-IL-17A or IL-23p19 antibody-induced Tregs significantly increased the number of Foxp3+ cells and IL-10 expression in imiquimod-induced psoriasiform dermatitis in recipient mice but anti-TNFα antibody-induced Tregs did not. CONCLUSION Anti-IL-17A or IL-23p19 antibody inhibits the IL-17/IL-23 signaling pathway, and induces expansion of Tregs and their suppressive capacity in imiquimod-induced psoriasiform dermatitis.
Collapse
Affiliation(s)
- Teruo Shimizu
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Masahiro Kamata
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan.
| | - Saki Fukaya
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Kotaro Hayashi
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Atsuko Fukuyasu
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Takamitsu Tanaka
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Takeko Ishikawa
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Takamitsu Ohnishi
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Yayoi Tada
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan.
| |
Collapse
|
34
|
Ambrée O, Ruland C, Zwanzger P, Klotz L, Baune BT, Arolt V, Scheu S, Alferink J. Social Defeat Modulates T Helper Cell Percentages in Stress Susceptible and Resilient Mice. Int J Mol Sci 2019; 20:ijms20143512. [PMID: 31319604 PMCID: PMC6678569 DOI: 10.3390/ijms20143512] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/13/2019] [Accepted: 07/16/2019] [Indexed: 12/21/2022] Open
Abstract
Altered adaptive immunity involving T lymphocytes has been found in depressed patients and in stress-induced depression-like behavior in animal models. Peripheral T cells play important roles in homeostasis and function of the central nervous system and thus modulate behavior. However, the T cell phenotype and function associated with susceptibility and resilience to depression remain largely unknown. Here, we characterized splenic T cells in susceptible and resilient mice after 10 days of social defeat stress (SDS). We found equally decreased T cell frequencies and comparably altered expression levels of genes associated with T helper (Th) cell function in resilient and susceptible mice. Interleukin (IL)-17 producing CD4+ and CD8+ T cell numbers in the spleen were significantly increased in susceptible mice. These animals further exhibited significantly reduced numbers of regulatory T cells (Treg) and decreased gene expression levels of TGF-β. Mice with enhanced Th17 differentiation induced by conditional deletion of PPARγ in CD4+ cells (CD4-PPARγKO), an inhibitor of Th17 development, were equally susceptible to SDS when compared to CD4-PPARγWT controls. These data indicate that enhanced Th17 differentiation alone does not alter stress vulnerability. Thus, SDS promotes Th17 cell and suppresses Treg cell differentiation predominantly in susceptible mice with yet unknown effects in immune responses after stress exposure.
Collapse
Affiliation(s)
- Oliver Ambrée
- Department of Psychiatry, University of Münster, 48149 Münster, Germany.
- Department of Behavioural Biology, University of Osnabrück, 49076 Osnabrück, Germany.
| | - Christina Ruland
- Department of Psychiatry, University of Münster, 48149 Münster, Germany
| | - Peter Zwanzger
- kbo-Inn-Salzach-Klinikum, 83512 Wasserburg am Inn, Germany
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| | - Luisa Klotz
- Department of Neurology, University of Münster, 49149 Münster, Germany
| | - Bernhard T Baune
- Department of Psychiatry, University of Münster, 48149 Münster, Germany
- Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Parkville, VIC 3010, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Volker Arolt
- Department of Psychiatry, University of Münster, 48149 Münster, Germany
| | - Stefanie Scheu
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Judith Alferink
- Department of Psychiatry, University of Münster, 48149 Münster, Germany.
- Cluster of Excellence EXC 1003, Cells in Motion, University of Münster, 48149 Münster, Germany.
| |
Collapse
|
35
|
Lippitsch A, Baal N, Chukovetskyi Y, Cunningham S, Michel G, Dietert K, Gurtner C, Gruber AD, Bein G, Hackstein H. Plasmacytoid dendritic cell depletion modifies FoxP3+ T cell homeostasis and the clinical course of bacterial pneumonia in mice. J Leukoc Biol 2019; 106:977-985. [PMID: 31265764 DOI: 10.1002/jlb.3ab0119-014rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 06/16/2019] [Accepted: 06/17/2019] [Indexed: 12/12/2022] Open
Abstract
Plasmacytoid dendritic cells (pDC) are critical to antiviral defense because of their high production of type I IFNs; less is known regarding their functions in bacterial infection. Moreover, pDC are involved in immunomodulation. A stable pool of regulatory T cells (Treg) is crucial for maintaining immune homeostasis. However, interactions between pDC and Treg regarding the regulation of Treg homeostasis are understudied. By using BDCA2-DTR mice as a systemic pDC depletion model, we identified increased steady-state numbers of FoxP3+ T cells with an effector Treg-like phenotype in lungs, liver, and spleen tissues. During sublethal, pulmonary Klebsiella pneumoniae infection, pDC deficiency also elevated respiratory FoxP3+ T cell numbers. Additionally, the improvement in acute pneumonia survival until day 5 post infection was accompanied by impaired proinflammatory cytokine production. In contrast, pDC-depleted mice exhibited a delayed clinical recovery during the post-acute phase. Therefore, we assume that pDC act as immunomodulators supporting the rapid onset of immune response in a proinflammatory manner and regulate inflammation or tissue regeneration in the post-acute phase. In summary, pDC assist in FoxP3+ T cell homeostasis and the regulation of Klebsiella-pneumonia progression.
Collapse
Affiliation(s)
- Anne Lippitsch
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Giessen, Germany
| | - Nelli Baal
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Giessen, Germany
| | - Yuri Chukovetskyi
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Giessen, Germany
| | - Sarah Cunningham
- Department of Transfusion Medicine and Haemostaseology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Gabriela Michel
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Giessen, Germany
| | - Kristina Dietert
- Department of Veterinary Pathology, Freie Universitaet Berlin, Berlin, Germany
| | - Corinne Gurtner
- Department of Veterinary Pathology, Freie Universitaet Berlin, Berlin, Germany
| | - Achim D Gruber
- Department of Veterinary Pathology, Freie Universitaet Berlin, Berlin, Germany
| | - Gregor Bein
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Giessen, Germany
| | - Holger Hackstein
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Giessen, Germany.,Department of Transfusion Medicine and Haemostaseology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuernberg, Erlangen, Germany
| |
Collapse
|
36
|
Geng X, Li M, Cui B, Lu C, Liu X, Zhang P, Liu B, Ma C, Shen Y, Lu Z. CD4+CD25+Foxp3+ regulatory T cells suppress NKG2D-mediated NK cell cytotoxicity in peripheral blood. Medicine (Baltimore) 2019; 98:e15722. [PMID: 31145286 PMCID: PMC6708973 DOI: 10.1097/md.0000000000015722] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Studies have shown that CD4CD25Foxp3Treg cells suppress NKG2D expression on NK cells via a cell contact-dependent mechanism and increased TGF-β and IL-10 production in some cancer models. We herein aimed to explore whether CD4CD25Foxp3Tregs suppress NKG2D-mediated NK cell cytotoxicity in peripheral blood and elucidate the exact mechanism underlying this phenomenon. METHODS To explore the function of NKG2D, NK cell cultures were treated with an NKG2D-blocking antibody to block these receptors. Additionally, TGF-β- and IL-10-blocking antibodies were added to NK and CD4CD25Foxp3Treg cell cocultures to evaluate whether the latter cells suppress NKG2D expression of NK cells via increasing the production of TGF-β and IL-10. The expression of NKG2D on NK cells was detected by 3-color flow cytometry, and NK cell activity was assessed by 3 assays: a nonradioactive cytotoxicity assay, an ELISA measuring IFN-γ production and a flow cytometry assay to evaluate CD107a expression. RESULTS Blocking NKG2D decreased NK cell cytotoxicity, IFN-γ production and CD107a expression. Moreover, blocking TGF-β and IL-10 substantially increased the NKG2D expression in NK and CD4CD25Foxp3Treg cell cocultures. Similarly, blocking TGF-β and IL-10 enhanced NK cell cytotoxicity, IFN-γ production and CD107a expression; Transwell insert assays also revealed increased IFN-γ production and CD107a and NKG2D expression. CONCLUSION CD4CD25Foxp3Tregs suppress NKG2D-mediated NK cell cytotoxicity in peripheral blood via a cell contact-dependent mechanism and increased TGF-β and IL-10 production.
Collapse
Affiliation(s)
- Xu Geng
- Department of Clinical Laboratory
| | - Ming Li
- Department of Clinical Laboratory
| | - Bin Cui
- Department of Central Laboratory
| | - Chao Lu
- Department of Clinical Laboratory
| | | | - Peng Zhang
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, PR China
| | - Bin Liu
- Department of Central Laboratory
| | | | | | | |
Collapse
|
37
|
Martinez Valenzuela L, Bordignon Draibe J, Fulladosa Oliveras X, Bestard Matamoros O, Cruzado Garrit JM, Torras Ambrós J. T-lymphocyte in ANCA-associated vasculitis: what do we know? A pathophysiological and therapeutic approach. Clin Kidney J 2019; 12:503-511. [PMID: 31384441 PMCID: PMC6671423 DOI: 10.1093/ckj/sfz029] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Indexed: 12/11/2022] Open
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is an autoimmune condition that commonly causes kidney impairment and can be fatal. The key participation of B-lymphocytes as ANCA producers and neutrophils as target of these antibodies is widely described as the mechanism of endothelial damage in this disease. There has been a rising interest in the role of T-lymphocytes in AAV in recent years. Evidence is strong from animal models, and T-lymphocytes can be found infiltrating kidney tissue and other tissue sites in AAV patients. Furthermore, the different subsets of T-lymphocytes are also key players in the aberrant immune response observed in AAV. Polarization towards a predominant Th1 and Th17 response in the acute phase of the disease has been described, along with a decline in the number of T-regulatory lymphocytes, which, in turn, show functional impairment. Interactions between different T-cell subsets, and between T-cells and neutrophils and B-cells, also enhance the inflammatory response, constituting a complex network. Novel therapies targeting T-cell immunity are emerging in this scenario and may constitute an interesting alternative to conventional therapy in selected patients. This review aims to summarize the available evidence regarding T-cell imbalances and functional impairment, especially focusing on renal involvement of AAV.
Collapse
Affiliation(s)
- Laura Martinez Valenzuela
- Bellvitge University Hospital, Nephrology Unit, Hospitalet de Llobregat, Spain.,IDIBELL Bellvitge Biomedical Research Institute, Hospitalet de Llobregat, Spain
| | - Juliana Bordignon Draibe
- Bellvitge University Hospital, Nephrology Unit, Hospitalet de Llobregat, Spain.,IDIBELL Bellvitge Biomedical Research Institute, Hospitalet de Llobregat, Spain
| | - Xavier Fulladosa Oliveras
- Bellvitge University Hospital, Nephrology Unit, Hospitalet de Llobregat, Spain.,IDIBELL Bellvitge Biomedical Research Institute, Hospitalet de Llobregat, Spain
| | - Oriol Bestard Matamoros
- Bellvitge University Hospital, Nephrology Unit, Hospitalet de Llobregat, Spain.,IDIBELL Bellvitge Biomedical Research Institute, Hospitalet de Llobregat, Spain.,Clinical Science Department, Barcelona University, Hospitalet de Llobregat, Spain
| | - Josep Maria Cruzado Garrit
- Bellvitge University Hospital, Nephrology Unit, Hospitalet de Llobregat, Spain.,IDIBELL Bellvitge Biomedical Research Institute, Hospitalet de Llobregat, Spain.,Clinical Science Department, Barcelona University, Hospitalet de Llobregat, Spain
| | - Juan Torras Ambrós
- Bellvitge University Hospital, Nephrology Unit, Hospitalet de Llobregat, Spain.,IDIBELL Bellvitge Biomedical Research Institute, Hospitalet de Llobregat, Spain.,Clinical Science Department, Barcelona University, Hospitalet de Llobregat, Spain
| |
Collapse
|
38
|
Moosbrugger-Martinz V, Gruber R, Ladstätter K, Bellutti M, Blunder S, Schmuth M, Dubrac S. Filaggrin null mutations are associated with altered circulating Tregs in atopic dermatitis. J Cell Mol Med 2018; 23:1288-1299. [PMID: 30515983 PMCID: PMC6349342 DOI: 10.1111/jcmm.14031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 10/21/2018] [Accepted: 10/26/2018] [Indexed: 01/10/2023] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease with a complex pathogenesis. Although regulatory T cells (Tregs) have previously been studied in AD, their role remains controversial, likely owing to patient heterogeneity. Thus, we recruited adult AD patients and age‐matched healthy controls, and assessed their filaggrin (FLG) genotype, serum IgE level, and eczema area and severity index (EASI). We found increased proportions of all circulating Treg subpopulations in AD patients. Moreover, we show positive correlations between circulating Tregs and serum IgE FLG null mutations limited the expansion of both memory and effector Tregs and enhanced that of recently thymus‐emigrated Tregs. Furthermore, proportions of circulating Th2‐ or Th17‐Tregs but not Th1‐Tregs were increased in AD patients, and accentuated by FLG null mutations, thereby mimicking the immune deviation observed in Th cell populations. Moreover, ICOS+ Tregs showed reduced production of interleukin‐10, suggesting impaired immunosuppression in AD. The level of demethylation of FOXP3i1, which reflects the stability of FOXP3 expression, was similar in the blood and skin of AD patients and healthy controls. Overall, these results show that Tregs may participate into AD pathogenesis and that FLG null mutations exert further modifications on specific subpopulations of circulating Tregs.
Collapse
Affiliation(s)
- Verena Moosbrugger-Martinz
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Robert Gruber
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Katharina Ladstätter
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Marion Bellutti
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Blunder
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Matthias Schmuth
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
39
|
Mitchell JM, Berzins SP, Kannourakis G. A potentially important role for T cells and regulatory T cells in Langerhans cell histiocytosis. Clin Immunol 2018; 194:19-25. [DOI: 10.1016/j.clim.2018.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/27/2018] [Accepted: 06/15/2018] [Indexed: 12/11/2022]
|
40
|
Wang Y, Merchen TD, Fang X, Lassiter R, Ho CS, Jajosky R, Kleven D, Thompson T, Mohamed E, Yu M, Waller JL, Nahman NS. Regulation of indoleamine 2,3 dioxygenase and its role in a porcine model of acute kidney allograft rejection. J Investig Med 2018; 66:1109-1117. [PMID: 30006478 DOI: 10.1136/jim-2018-000742] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2018] [Indexed: 12/21/2022]
Abstract
In kidney transplantation acute allograft rejection is the most common cause of late allograft loss. Changes in indoleamine 2,3 dioxygenase (IDO) activity, which catabolizes the degradation of tryptophan to kynurenine, may predict rejection. However, exogenous IDO is immunosuppressive in rodent kidney transplantation. Thus, the increase in IDO activity observed in acute allograft rejection is insufficient to prevent rejection. To address this question, we assessed the regulation of IDO and its role in acute rejection in a porcine model of kidney transplant. In tissue samples from rejecting kidney allografts, we showed a 13-fold increase in IDO gene transcription and 20-fold increase in IDO enzyme activity when compared with autotransplanted kidneys. Allografts also demonstrated an over fourfold increase in tissue interferon (IFN)-γ, with marked increases in tumor necrosis factor (TNF)-α, TNF-β and interleukin 1β. Gene transcription and protein levels of kynurenine 3-monooxygenase (KMO) were decreased. KMO generates the immunosuppressive kynurenine, 3-hydroxykynurenine. The results of these studies demonstrate a clear association between rejection and increased allograft IDO expression, likely driven in part by IFN-γ and facilitated by other cytokines of the allogeneic response. Moreover, the loss of downstream enzymatic activity in the IDO metabolic pathway may suggest novel mechanisms for the perpetuation of rejection.
Collapse
Affiliation(s)
- Youli Wang
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Todd D Merchen
- Department of Surgery, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Xuexiu Fang
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Randi Lassiter
- Department of Surgery, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Chak-Sum Ho
- Gift of Life Michigan, Ann Arbor, Michigan, USA
| | - Ryan Jajosky
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Daniel Kleven
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Thomas Thompson
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Eslam Mohamed
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Miao Yu
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Jennifer L Waller
- Department of Population Health Sciences, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - N Stanley Nahman
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA.,Medicine, Charlie Norwood VAMC, Augusta, Georgia, USA
| |
Collapse
|
41
|
Mast cells participate in allograft rejection: can IL-37 play an inhibitory role? Inflamm Res 2018; 67:747-755. [PMID: 29961151 DOI: 10.1007/s00011-018-1166-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/18/2018] [Accepted: 06/20/2018] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE The aim of this study was to evaluate the role of mast cells (MCs) in allograft rejection, eventually inhibited by IL-37. Immune cells including MCs participate in allograft rejection by generating IL-1, IL-33, TNF and other cytokines. METHODS We evaluated allograft rejection on the experience of our experimental data and using the relevant literature. RESULTS MCs are involved in initiation and regulation of innate and adaptive immune responses-pathways. MCs are important pro-inflammatory cells which express high-affinity receptor FceRI and can be activated by IgE and some pro-inflammatory cytokines, such as IL-1 and IL-33. The cross-linkage of high affinity IgE receptor on MCs by antigen ligation has a crucial role in allergy, asthma, anaphylaxis, cancer and allograft rejection. MCs mediate immunity in organ transplant, leading to the activation of allospecific T cells implicated in the rejection and generate pro-inflammatory cytokines/chemokines. IL-1 pro-inflammatory cytokine family members released by MCs mediate allograft rejection and inflammation. IL-37 is also an IL-1 family member generated by macrophage cell line in small amounts, which binds to IL-18Rα and produces an anti-inflammatory effect. IL-37 provokes the inhibition of TLR signaling, TLR-induced mTOR and (MyD88)-mediated responses, suppressing pro-inflammatory IL-1 family members and increasing IL-10. CONCLUSION IL-37 inhibition offers the opportunity to immunologically modulate MCs, by suppressing their production of IL-1 family members and reducing the risk of allograft rejection, resulting as a potential good therapeutic new cytokine. Here, we report the relationship between inflammatory MCs, allograft rejection and pro-inflammatory and anti-inflammatory IL-37.
Collapse
|
42
|
Regulatory Role of CD4 + T Cells in Myocarditis. J Immunol Res 2018; 2018:4396351. [PMID: 30035131 PMCID: PMC6032977 DOI: 10.1155/2018/4396351] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/21/2018] [Accepted: 05/29/2018] [Indexed: 12/13/2022] Open
Abstract
Myocarditis is an important cause of heart failure in young patients. Autoreactive, most often, infection-triggered CD4+ T cells were confirmed to be critical for myocarditis induction. Due to a defect in clonal deletion of heart-reactive CD4+ T cells in the thymus of mice and humans, significant numbers of heart-specific autoreactive CD4+ T cells circulate in the blood. Normally, regulatory T cells maintain peripheral tolerance and prevent spontaneous myocarditis development. In the presence of tissue damage and innate immune activation, however, activated self-antigen-loaded dendritic cells promote CD4+ effector T cell expansion and myocarditis. So far, a direct pathogenic role has been described for both activated Th17 and Th1 effector CD4+ T cell subsets, though Th1 effector T cell-derived interferon-gamma was shown to limit myocarditis severity and prevent transition to inflammatory dilated cardiomyopathy. Interestingly, recent observations point out that various CD4+ T cell subsets demonstrate high plasticity in maintaining immune homeostasis and modulating disease phenotypes in myocarditis. These subsets include Th1 and Th17 effector cells and regulatory T cells, despite the fact that there are still sparse and controversial data on the specific role of FOXP3-expressing Treg in myocarditis. Understanding the specific roles of these T cell populations at different stages of the disease progression might provide a key for the development of successful therapeutic strategies.
Collapse
|
43
|
Li Y, Li Y, Lu W, Li H, Wang Y, Luo H, Wu Y, Dong W, Bai G, Zhang Y. Integrated Network Pharmacology and Metabolomics Analysis of the Therapeutic Effects of Zi Dian Fang on Immune Thrombocytopenic Purpura. Front Pharmacol 2018; 9:597. [PMID: 29971001 PMCID: PMC6018083 DOI: 10.3389/fphar.2018.00597] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 05/18/2018] [Indexed: 12/25/2022] Open
Abstract
Current hormone-based treatments for immune thrombocytopenic purpura (ITP) are associated with potentially serious adverse reactions. Zi Dian Fang (ZDF) is a multi-target Traditional Chinese Medicine (TCM) used to treat both the symptoms and root causes of ITP, with fewer side effects than hormone-based treatments. This study analysis of the therapeutic effects of ZDF on ITP from three aspects: platelet proliferation, immunoregulation, and inflammation. After detection of 52 chemical constituents of ZDF by UPLC-Q-TOF/MS, The main targets and pathways affected by ZDF were screened by network pharmacology and verified by Western blot and ELISA. Meanwhile, metabolomics analysis were applied to a mouse model of ITP to identify and screen endogenous terminal metabolites differentially regulated by ZDF. Integrated network pharmacology and metabolomics analysis of the therapeutic effects of ZDF on ITP may be as follows: ZDF counteracts ITP symptoms mainly by inhibiting Ras/MAPKs (Ras/Mitogen-activated protein kinases) pathway, and the expression of upstream protein (Ras) and downstream protein (p-ERK, p-JNK, p-p38) were inhibited, which affects the content of effect index associated with proliferation (Thrombopoietin, TPO; Granulocyte-macrophage colony stimulating factor, GM-CSF), inflammation (Tumor necrosis factor-α, TNF-α; Interleukin-6, IL-6), immune (Interleukin-2, IL-2; Interferon-gamma, IFN-γ; Interleukin-4, IL-4), so that the body’s arginine, Δ12-prostaglandin j2 (Δ12-PGJ2), 9-cis-Retinoic Acid, sphingosine-1-phosphate (S1P), oleic acid amide and other 12 endogenous metabolites significantly changes. Considering the established safety profile, the present study suggests ZDF may be a useful alternative to hormone-based therapies for the treatment of ITP.
Collapse
Affiliation(s)
- Yubo Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yamei Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenliang Lu
- Tasly Institute, Tasly Pharmaceutical Group, Tianjin, China
| | - Hongbin Li
- Tasly Institute, Tasly Pharmaceutical Group, Tianjin, China
| | - Yuming Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Houmin Luo
- Tianjin State Key Laboratory of Modern Chinese Medicine, School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuanyuan Wu
- Tianjin State Key Laboratory of Modern Chinese Medicine, School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenying Dong
- Tianjin State Key Laboratory of Modern Chinese Medicine, School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Gang Bai
- College of Pharmacy, Nankai University, Tianjin, China
| | - Yanjun Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
44
|
Kawai K, Uchiyama M, Hester J, Wood K, Issa F. Regulatory T cells for tolerance. Hum Immunol 2018; 79:294-303. [DOI: 10.1016/j.humimm.2017.12.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 12/16/2017] [Accepted: 12/26/2017] [Indexed: 12/29/2022]
|
45
|
Immature Exosomes Derived from MicroRNA-146a Overexpressing Dendritic Cells Act as Antigen-Specific Therapy for Myasthenia Gravis. Inflammation 2018; 40:1460-1473. [PMID: 28523463 DOI: 10.1007/s10753-017-0589-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Myasthenia gravis (MG) is a neurological autoimmune disease characterized by fluctuating weakness of certain voluntary muscles. Current treatments for MG are largely directed at suppressing the whole immune system by using immunosuppressants or glucocorticoids and often cause several side effects. The ideal therapeutic methods for MG should suppress aberrant immunoactivation specifically, while retaining normal function of the immune system. In this study, we first produced exosomes from microRNA-146a overexpressing dendritic cells (DCs). Then, we observed suppressive effects of those exosomes in experimental autoimmune myasthenia gravis (EAMG) mice. Results showed that exosomes from microRNA-146a overexpressing DCs expressed decreased levels of CD80 and CD86. In experimental autoimmune MG, exosomes from microRNA-146a overexpressing DCs suppressed ongoing clinical MG in mice and altered T helper cell profiles from Th1/Th17 to Th2/Treg both in serum and spleen, and the therapeutic effects of those exosomes were antigen-specific and partly dose dependent. All the findings provide experimental basis for antigen-specific therapy of MG.
Collapse
|
46
|
Gołąb K, Grose R, Trzonkowski P, Wickrema A, Tibudan M, Marek-Trzonkowska N, Matosz S, Solomina J, Ostrega D, Michael Millis J, Witkowski P. Utilization of leukapheresis and CD4 positive selection in Treg isolation and the ex-vivo expansion for a clinical application in transplantation and autoimmune disorders. Oncotarget 2018; 7:79474-79484. [PMID: 27821811 PMCID: PMC5346728 DOI: 10.18632/oncotarget.13101] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 10/26/2016] [Indexed: 12/20/2022] Open
Abstract
Adoptive transfer of T regulatory cells (Tregs) is of great interest as a novel immunosuppressive therapy in autoimmune disorders and transplantation. Obtaining a sufficient number of stable and functional Tregs generated according to current Good Manufacturing Practice (cGMP) requirements has been a major challenge in introducing Tregs as a clinical therapy. Here, we present a protocol involving leukapheresis and CD4+ cell pre-enrichment prior to Treg sorting, which allows a sufficient number of Tregs for a clinical application to be obtained. With this method there is a decreased requirement for ex-vivo expansion. The protocol was validated in cGMP conditions. Our final Treg product passed all release criteria set for clinical applications. Moreover, during expansion Tregs presented their stable phenotype: percentage of CD4+CD25hiCD127− and CD4+FoxP3+ Tregs was > 95% and > 80%, respectively, and Tregs maintained proper immune suppressive function in vitro. Our results suggest that utilization of leukapheresis and CD4 positive selection during Treg isolation improves the likelihood of obtaining a sufficient number of high quality Treg cells during subsequent ex-vivo expansion and they can be applied clinically.
Collapse
Affiliation(s)
- Karolina Gołąb
- Department of Surgery, Section of Transplantation, University of Chicago, Chicago, USA
| | - Randall Grose
- South Australian Health and Medical Research Institute, University of Adelaide, Australia
| | - Piotr Trzonkowski
- Department of Clinical Immunology and Transplantology, Medical University of Gdansk, Gdansk, Poland
| | - Amittha Wickrema
- Department of Medicine, Section of Hematology-Oncology, Cancer Research Center, University of Chicago, Chicago, USA
| | - Martin Tibudan
- Department of Surgery, Section of Transplantation, University of Chicago, Chicago, USA
| | | | - Sabrina Matosz
- Department of Surgery, Section of Transplantation, University of Chicago, Chicago, USA
| | - Julia Solomina
- Department of Surgery, Section of Transplantation, University of Chicago, Chicago, USA
| | - Diane Ostrega
- Department of Surgery, Section of Transplantation, University of Chicago, Chicago, USA
| | - J Michael Millis
- Department of Surgery, Section of Transplantation, University of Chicago, Chicago, USA
| | - Piotr Witkowski
- Department of Surgery, Section of Transplantation, University of Chicago, Chicago, USA
| |
Collapse
|
47
|
Molecular adjuvants that modulate regulatory T cell function in vaccination: A critical appraisal. Pharmacol Res 2017; 129:237-250. [PMID: 29175113 DOI: 10.1016/j.phrs.2017.11.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 12/13/2022]
Abstract
Adjuvants are substances used to enhance the efficacy of vaccines. They influence the magnitude and alter the quality of the adaptive immune response to vaccine antigens by amplifying or modulating different signals involved in the innate immune response. The majority of known adjuvants have been empirically identified. The limited immunogenicity of new vaccine antigens and the need for safer vaccines have increased the importance of identifying single, well-defined adjuvants with known cellular and molecular mechanisms for rational vaccine design. Depletion or functional inhibition of CD4+CD25+FoxP3+ regulatory T cells (Tregs) by molecular adjuvants has become an emergent approach in this field. Different successful results have been obtained for specific vaccines, but there are still unresolved issues such as the risk of autoimmune disease induction, the involvement of cells other than Tregs and optimization for different conditions. This work provides a comprehensive analysis of current approaches to inhibit Tregs with molecular adjuvants for vaccine improvement, highlights the progress being made, and describes ongoing challenges.
Collapse
|
48
|
Cravedi P, Farouk S, Angeletti A, Edgar L, Tamburrini R, Duisit J, Perin L, Orlando G. Regenerative immunology: the immunological reaction to biomaterials. Transpl Int 2017; 30:1199-1208. [PMID: 28892571 DOI: 10.1111/tri.13068] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 06/29/2017] [Accepted: 09/04/2017] [Indexed: 01/09/2023]
Abstract
Regenerative medicine promises to meet two of the most urgent needs of modern organ transplantation, namely immunosuppression-free transplantation and an inexhaustible source of organs. Ideally, bioengineered organs would be manufactured from a patient's own biomaterials-both cells and the supporting scaffolding materials in which cells would be embedded and allowed to mature to eventually regenerate the organ in question. While some groups are focusing on the feasibility of this approach, few are focusing on the immunogenicity of the scaffolds that are being developed for organ bioengineering purposes. This review will succinctly discuss progress in the understanding of immunological characteristics and behavior of different scaffolds currently under development, with emphasis on the extracellular matrix scaffolds obtained decellularized animal or human organs which seem to provide the ideal template for bioengineering purposes.
Collapse
Affiliation(s)
- Paolo Cravedi
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samira Farouk
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea Angeletti
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Experimental, Diagnostic, Specialty Medicine, Nephrology, Dialysis, and Renal Transplant Unit, S. Orsola University Hospital, Bologna, Italy
| | - Lauren Edgar
- Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Riccardo Tamburrini
- Wake Forest University School of Medicine, Winston Salem, NC, USA.,Section of Transplantation, Department of Surgery, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Jerome Duisit
- Pôle de Chirurgie Expérimentale (CHEX), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Plastic and Reconstructive Surgery, Cliniques Universitaires St-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Laura Perin
- Division of Urology, GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics, Saban Research Institute, Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Giuseppe Orlando
- Wake Forest University School of Medicine, Winston Salem, NC, USA.,Section of Transplantation, Department of Surgery, Wake Forest University School of Medicine, Winston Salem, NC, USA
| |
Collapse
|
49
|
Cord blood T cell subpopulations and associations with maternal cadmium and arsenic exposures. PLoS One 2017; 12:e0179606. [PMID: 28662050 PMCID: PMC5491028 DOI: 10.1371/journal.pone.0179606] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 05/31/2017] [Indexed: 11/19/2022] Open
Abstract
Background Arsenic and cadmium are environmental pollutants, and although the evidence for adverse immune effects after prenatal arsenic and cadmium exposures is increasing, little is known about the underlying immunological mechanisms. Methods We investigated the relationship between prenatal arsenic and cadmium exposures and a variety of T cell subpopulations measured in cord blood for 63 participants in the New Hampshire Birth Cohort Study. Post-partum toenail concentrations of arsenic and cadmium were used as an estimate of maternal exposure during pregnancy. The characteristics of cord blood proportions of T lymphocytes and subpopulations (expression of markers for Th1, Th2, Th17, Th1Th17, induced and natural regulatory T cells and NKTs) are presented. Results In regression analyses, maternal arsenic exposure levels were inversely associated with cord blood T helper memory cells (-21%, 95% CI: -36%, -3%) and the association was found to be stronger in females. They were also inversely associated with activated T helper memory cells, particularly in males (-26%, 95% CI: -43%, -3%). Similarly, inverse associations were observed between cadmium exposure levels and activated T helper memory cells (-16%, 95% CI: -30%, -1%) and also for T helper memory cells in females (-20%, 95% CI: -35%, -3%). Conclusion The results suggest that prenatal exposures to relatively low levels of arsenic and cadmium may contribute to altered distribution of T cell populations at birth. These changes in theory, could have contributed to the previously reported immunosuppressive effects observed later in infancy/childhood.
Collapse
|
50
|
Cytokines affecting CD4 +T regulatory cells in transplant tolerance. III. Interleukin-5 (IL-5) promotes survival of alloantigen-specific CD4 + T regulatory cells. Transpl Immunol 2017; 43-44:33-41. [PMID: 28652007 DOI: 10.1016/j.trim.2017.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/22/2017] [Accepted: 06/22/2017] [Indexed: 12/31/2022]
Abstract
CD4+T cells mediate antigen-specific allograft tolerance, but die in culture without activated lymphocyte derived cytokines. Supplementation of the media with cytokine rich supernatant, from ConA activated spleen cells, preserves the capacity of tolerant cells to transfer tolerance and suppress rejection. rIL-2 or rIL-4 alone are insufficient to maintain these cells, however. We observed that activation of naïve CD4+CD25+FOXP3+Treg with alloantigen and the Th2 cytokine rIL-4 induces them to express interleukin-5 specific receptor alpha (IL-5Rα) suggesting that IL-5, a Th2 cytokine that is produced later in the immune response may promote tolerance mediating Treg. This study examined if recombinant IL-5(rIL-5) promoted survival of tolerant CD4+, especially CD4+CD25+T cells. CD4+T cells, from DA rats tolerant to fully allogeneic PVG heart allografts surviving over 100days without on-going immunosuppression, were cultured with PVG alloantigen and rIL-5. The ability of these cells to adoptively transfer tolerance to specific-donor allograft and suppress normal CD4+T cell mediated rejection in adoptive DA hosts was examined. Tolerant CD4+CD25+T cells' response to rIL-5 and expression of IL-5Rα was also assessed. rIL-5 was sufficient to promote transplant tolerance mediating CD4+T cells' survival in culture with specific-donor alloantigen. Tolerant CD4+T cells cultured with rIL-5 retained the capacity to transfer alloantigen-specific tolerance and inhibited naïve CD4+T cells' capacity to effect specific-donor graft rejection. rIL-5 promoted tolerant CD4+CD25+T cells' proliferation in vitro when stimulated with specific-donor but not third-party stimulator cells. Tolerant CD4+CD25+T cells expressed IL-5Rα. This study demonstrated that IL-5 promoted the survival of alloantigen-specific CD4+CD25+T cells that mediate transplant tolerance.
Collapse
|