1
|
Sun X, Hosomi K, Shimoyama A, Yoshii K, Lan H, Wang Y, Yamaura H, Nagatake T, Ishii KJ, Akira S, Kiyono H, Fukase K, Kunisawa J. TLR4 agonist activity of Alcaligenes lipid a utilizes MyD88 and TRIF signaling pathways for efficient antigen presentation and T cell differentiation by dendritic cells. Int Immunopharmacol 2023; 117:109852. [PMID: 36806039 DOI: 10.1016/j.intimp.2023.109852] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 01/18/2023] [Accepted: 02/04/2023] [Indexed: 02/22/2023]
Abstract
Alcaligenes faecalis was previously identified as an intestinal lymphoid tissue-resident commensal bacteria, and our subsequent studies showed that lipopolysaccharide and its core active element (i.e., lipid A) have a potent adjuvant activity to promote preferentially antigen-specific Th17 response and antibody production. Here, we compared A. faecalis lipid A (ALA) with monophosphoryl lipid A, a licensed lipid A-based adjuvant, to elucidate the immunological mechanism underlying the adjuvant properties of ALA. Compared with monophosphoryl lipid A, ALA induced higher levels of MHC class II molecules and costimulatory CD40, CD80, and CD86 on dendritic cells (DCs), which in turn resulted in strong T cell activation. Moreover, ALA more effectively promoted the production of IL-6 and IL-23 from DCs than did monophosphoryl lipid A, thus leading to preferential induction of Th17 and Th1 cells. As underlying mechanisms, we found that the ALA-TLR4 axis stimulated both MyD88- and TRIF-mediated signaling pathways, whereas monophosphoryl lipid A was biased toward TRIF signaling. These findings revealed the effects of ALA on DCs and T cells and its induction pattern on signaling pathways.
Collapse
Affiliation(s)
- Xiao Sun
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan; Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Atsushi Shimoyama
- Graduate School of Science, Osaka University, Osaka, Japan; Collaborative Research between NIBIOHN and Graduate School of Science, Forefront Research Center, Osaka University, Osaka, Japan
| | - Ken Yoshii
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan; Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Huangwenxian Lan
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan; Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Yunru Wang
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan; Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Haruki Yamaura
- Graduate School of Science, Osaka University, Osaka, Japan
| | - Takahiro Nagatake
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan; Laboratory of Functional Anatomy, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa, Japan
| | - Ken J Ishii
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Immunology Frontier Research Center, Osaka University, Osaka, Japan; Center for Vaccine and Adjuvant Research (CVAR), National Institute of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Shizuo Akira
- Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Hiroshi Kiyono
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Division of Gastroenterology, Department of Medicine, University of California San Diego (UCSD), San Diego, CA, United States; Chiba University (CU)-UCSD Center for Mucosal Immunology, Allergy and Vaccines (cMAV), UCSD, San Diego, CA, United States; Future Medicine Education and Research Organization, Chiba University, Chiba, Japan; Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan; Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Koichi Fukase
- Graduate School of Science, Osaka University, Osaka, Japan; Collaborative Research between NIBIOHN and Graduate School of Science, Forefront Research Center, Osaka University, Osaka, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan; Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan; Graduate School of Science, Osaka University, Osaka, Japan; Collaborative Research between NIBIOHN and Graduate School of Science, Forefront Research Center, Osaka University, Osaka, Japan; Graduate School of Medicine, Osaka University, Osaka, Japan; International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Kobe, Japan; Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan; Graduate School of Dentistry, Osaka University, Suita, Japan.
| |
Collapse
|
2
|
Qu W, Guo Y, Xu Y, Zhang J, Wang Z, Ding C, Pan Y. Advance in strategies to build efficient vaccines against tuberculosis. Front Vet Sci 2022; 9:955204. [PMID: 36504851 PMCID: PMC9731747 DOI: 10.3389/fvets.2022.955204] [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: 05/28/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
Tuberculosis is a chronic consumptive infectious disease, which can cause great damage to human and animal health all over the world. The emergence of multi-drug resistant strains, the unstable protective effect of Bacillus Calmette-Guérin (BCG) vaccine on adults, and the mixed infection with HIV all warn people to exploit new approaches for conquering tuberculosis. At present, there has been significant progress in developing tuberculosis vaccines, such as improved BCG vaccine, subunit vaccine, DNA vaccine, live attenuated vaccine and inactivated vaccine. Among these candidate vaccines, there are some promising vaccines to improve or replace BCG vaccine effect. Meanwhile, the application of adjuvants, prime-boost strategy, immunoinformatic tools and targeting components have been studied concentratedly, and verified as valid means of raising the efficiency of tuberculosis vaccines as well. In this paper, the latest advance in tuberculosis vaccines in recent years is reviewed to provide reliable information for future tuberculosis prevention and treatment.
Collapse
Affiliation(s)
- Wei Qu
- National Reference Laboratory of Veterinary Drug Residues, MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China,MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, China
| | - Yinhui Guo
- National Reference Laboratory of Veterinary Drug Residues, MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Yan Xu
- National Reference Laboratory of Veterinary Drug Residues, MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Jie Zhang
- National Reference Laboratory of Veterinary Drug Residues, MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China,MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, China
| | - Zongchao Wang
- National Reference Laboratory of Veterinary Drug Residues, MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Chaoyue Ding
- National Reference Laboratory of Veterinary Drug Residues, MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Yuanhu Pan
- National Reference Laboratory of Veterinary Drug Residues, MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China,MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, China,*Correspondence: Yuanhu Pan
| |
Collapse
|
3
|
Vaillant L, Oster P, McMillan B, Orozco Fernandez E, Velin D. GM-CSF is key in the efficacy of vaccine-induced reduction of Helicobacter pylori infection. Helicobacter 2022; 27:e12875. [PMID: 35092634 PMCID: PMC9285700 DOI: 10.1111/hel.12875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/22/2021] [Accepted: 01/16/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND Helicobacter pylori (H. pylori) colonizes the human gastric mucosa with a high worldwide prevalence. Currently, H. pylori is eradicated by the use of antibiotics. However, elevated antibiotic resistance suggests new therapeutic strategies need to be envisioned: one approach being prophylactic vaccination. Pre-clinical and clinical data show that a urease-based vaccine is efficient in decreasing H. pylori infection through the mobilization of T helper (Th) cells, especially Th17 cells. Th17 cells produce interleukins such as IL-22 and IL-17, among others, and are key players in vaccine efficacy. Recently, granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing Th17 cells have been identified. AIM This study explores the possibility that GM-CSF plays a role in the reduction of H. pylori infection following vaccination. RESULTS We demonstrate that GM-CSF+ IL-17+ Th17 cells accumulate in the stomach mucosa of H. pylori infected mice during the vaccine-induced reduction of H. pylori infection. Secondly, we provide evidence that vaccinated GM-CSF deficient mice only modestly reduce H. pylori infection. Conversely, we observe that an increase in GM-CSF availability reduces H. pylori burden in chronically infected mice. Thirdly, we show that GM-CSF, by acting on gastric epithelial cells, promotes the production of βdefensin3, which exhibits H. pylori bactericidal activities. CONCLUSION Taken together, we demonstrate a key role of GM-CSF, most probably originating from Th17 cells, in the vaccine-induced reduction of H. pylori infection.
Collapse
Affiliation(s)
- Laurie Vaillant
- Service of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversity of LausanneLausanneSwitzerland
| | - Paul Oster
- Service of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversity of LausanneLausanneSwitzerland
| | - Brynn McMillan
- Service of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversity of LausanneLausanneSwitzerland
| | - Eulalia Orozco Fernandez
- Service of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversity of LausanneLausanneSwitzerland
| | - Dominique Velin
- Service of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversity of LausanneLausanneSwitzerland
| |
Collapse
|
4
|
Immune cell and TCR/BCR repertoire profiling in systemic lupus erythematosus patients by single-cell sequencing. Aging (Albany NY) 2021; 13:24432-24448. [PMID: 34772824 PMCID: PMC8610142 DOI: 10.18632/aging.203695] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/27/2021] [Indexed: 12/27/2022]
Abstract
The immune cells and the repertoire of T cells and B cells play an important role in the pathogenesis of systemic lupus erythematosus (SLE). Exploring their expression and distribution in SLE can help us better understand this lethal autoimmune disease. In this study, we used a single-cell 5’ RNA sequence and single-cell T cell receptor (TCR)/B cell receptor (BCR) to study the immune cells and the repertoire from ten SLE patients and the paired normal controls (NC). The results showed that 9732 cells correspondence to 12 cluster immune cell types were identified in NC, whereas 11042 cells correspondence to 16 cluster immune cell types were identified in SLE. The results demonstrated that neutrophil, macrophage, and dendritic cells were accumulated in SLE by annotating the immune cell types. Besides, the bioinformatics analysis of differentially expressed genes (DEGs) in these cell types indicates their role in inflammation response. In addition, patients with SLE showed increased TCR and BCR clonotypes compared with the healthy controls. Furthermore, patients with SLE showed biased usage of TCR and BCR V(D)J genes. Taken together, we characterized the transcriptome and TCR/BCR immune repertoire profiles of SLE patients, which may provide a new avenue for the diagnosis and treatment of SLE.
Collapse
|
5
|
Tomioka H, Tatano Y, Shimizu T, Sano C. Immunoadjunctive Therapy against Bacterial Infections Using Herbal Medicines Based on Th17 Cell-mediated Protective Immunity. Curr Pharm Des 2021; 27:3949-3962. [PMID: 34102961 DOI: 10.2174/1381612827666210608143449] [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: 10/15/2020] [Accepted: 04/27/2021] [Indexed: 11/22/2022]
Abstract
One of the major health concerns in the world is the global increase in intractable bacterial infectious diseases due to the emergence of multi- and extensively drug-resistant bacterial pathogens as well as an increase in compromised hosts around the world. Particularly, in the case of mycobacteriosis, the high incidence of tuberculosis in developing countries, resurgence of tuberculosis in industrialized countries, and increase in the prevalence of Mycobacterium avium complex infections are important worldwide health concerns. However, the development of novel antimycobacterial drugs is currently making slow progress. Therefore, it is considered that devising improved administration protocols for clinical treatment against refractory mycobacteriosis using existing chemotherapeutics is more practical than awaiting the development of new antimycobacterial drugs. The regulation of host immune responses using immunoadjunctive agents may increase the efficacy of antimicrobial treatment against mycobacteriosis. The same situations also exist in cases of intractable infectious diseases due to common bacteria other than mycobacteria. The mild and long-term up-regulation of host immune reactions in hosts with intractable chronic bacterial infections, using herbal medicines and medicinal plants, may be beneficial for such immunoadjunctive therapy. This review describes the current status regarding basic and clinical studies on therapeutic regimens using herbal medicines, useful for the clinical treatment of patients with intractable bacterial infections. In particular, we focus on immunoadjunctive effects of herbal medicines on the establishment and manifestation of host antibacterial immunity related to the immunological roles of Th17 cell lineages.
Collapse
Affiliation(s)
- Haruaki Tomioka
- Department of Basic Medical Science for Nursing, Department of Contemporary Psychology, Yasuda Women's University, Hiroshima, Japan
| | - Yutaka Tatano
- Department of Pharmaceutical Sciences, International University of Health and Welfare, Fukuoka, Japan
| | - Toshiaki Shimizu
- Department of Nutrition Administration, Yasuda Women's University, Hiroshima,, Japan
| | - Chiaki Sano
- Department of Community Medicine Management, Shimane University School of Medicine, Izumo, Japan
| |
Collapse
|
6
|
Zenobia C, Herpoldt KL, Freire M. Is the oral microbiome a source to enhance mucosal immunity against infectious diseases? NPJ Vaccines 2021; 6:80. [PMID: 34078913 PMCID: PMC8172910 DOI: 10.1038/s41541-021-00341-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/24/2021] [Indexed: 12/14/2022] Open
Abstract
Mucosal tissues act as a barrier throughout the oral, nasopharyngeal, lung, and intestinal systems, offering first-line protection against potential pathogens. Conventionally, vaccines are applied parenterally to induce serotype-dependent humoral response but fail to drive adequate mucosal immune protection for viral infections such as influenza, HIV, and coronaviruses. Oral mucosa, however, provides a vast immune repertoire against specific microbial pathogens and yet is shaped by an ever-present microbiome community that has co-evolved with the host over thousands of years. Adjuvants targeting mucosal T-cells abundant in oral tissues can promote soluble-IgA (sIgA)-specific protection to confer increased vaccine efficacy. Th17 cells, for example, are at the center of cell-mediated immunity and evidence demonstrates that protection against heterologous pathogen serotypes is achieved with components from the oral microbiome. At the point of entry where pathogens are first encountered, typically the oral or nasal cavity, the mucosal surfaces are layered with bacterial cohabitants that continually shape the host immune profile. Constituents of the oral microbiome including their lipids, outer membrane vesicles, and specific proteins, have been found to modulate the Th17 response in the oral mucosa, playing important roles in vaccine and adjuvant designs. Currently, there are no approved adjuvants for the induction of Th17 protection, and it is critical that this research is included in the preparedness for the current and future pandemics. Here, we discuss the potential of oral commensals, and molecules derived thereof, to induce Th17 activity and provide safer and more predictable options in adjuvant engineering to prevent emerging infectious diseases.
Collapse
Affiliation(s)
| | | | - Marcelo Freire
- Departments of Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, USA.
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
7
|
Th17-type immunity and inflammation of aging. Aging (Albany NY) 2021; 13:13378-13379. [PMID: 34057906 PMCID: PMC8202903 DOI: 10.18632/aging.203119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 12/17/2022]
|
8
|
Cheng WT, Kantilal HK, Davamani F. The Mechanism of Bacteroides fragilis Toxin Contributes to Colon Cancer Formation. Malays J Med Sci 2020; 27:9-21. [PMID: 32863742 PMCID: PMC7444842 DOI: 10.21315/mjms2020.27.4.2] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/12/2020] [Indexed: 12/17/2022] Open
Abstract
The Bacteroides fragilis (B. fragilis) produce biofilm for colonisation in the intestinal tract can cause a series of inflammatory reactions due to B. fragilis toxin (BFT) which can lead to chronic intestinal inflammation and tissue injury and play a crucial role leading to colorectal cancer (CRC). The enterotoxigenic B. fragilis (ETBF) forms biofilm and produce toxin and play a role in CRC, whereas the non-toxigenic B. fragilis (NTBF) does not produce toxin. The ETBF triggers the expression of cyclooxygenase (COX)-2 that releases PGE2 for inducing inflammation and control cell proliferation. From chronic intestinal inflammation to cancer development, it involves signal transducers and activators of transcription (STAT)3 activation. STAT3 activates by the interaction between epithelial cells and BFT. Thus, regulatory T-cell (Tregs) will activates and reduce interleukin (IL)-2 amount. As the level of IL-2 drops, T-helper (Th17) cells are generated leading to increase in IL-17 levels. IL-17 is implicated in early intestinal inflammation and promotes cancer cell survival and proliferation and consequently triggers IL-6 production that activate STAT3 pathway. Additionally, BFT degrades E-cadherin, hence alteration of signalling pathways can upregulate spermine oxidase leading to cell morphology and promote carcinogenesis and irreversible DNA damage. Patient with familial adenomatous polyposis (FAP) disease displays a high level of tumour load in the colon. This disease is caused by germline mutation of the adenomatous polyposis coli (APC) gene that increases bacterial adherence to the mucosa layer. Mutated-APC gene genotype with ETBF increases the chances of CRC development. Therefore, the colonisation of the ETBF in the intestinal tract depicts tumour aetiology can result in risk of hostility and effect on human health.
Collapse
Affiliation(s)
- Wai Teng Cheng
- Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| | - Haresh Kumar Kantilal
- Division of Pathology, School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Fabian Davamani
- Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
9
|
Avila-Calderón ED, Flores-Romo L, Sharon W, Donis-Maturano L, Becerril-García MA, Arreola MGA, Reynoso BA, Güemes FS, Contreras-Rodríguez A. Dendritic cells and Brucella spp. interaction: the sentinel host and the stealthy pathogen. Folia Microbiol (Praha) 2020; 65:1-16. [PMID: 30783994 PMCID: PMC7224029 DOI: 10.1007/s12223-019-00691-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/06/2019] [Indexed: 01/18/2023]
Abstract
As dendritic cells (DCs) are among the first cells to encounter antigens, these cells trigger both innate and T cell responses, and are the most potent antigen-presenting cells. Brucella spp., which is an intracellular facultative and stealthy pathogen, is able to evade the bactericidal activities of professional phagocytes. Several studies have demonstrated that Brucella can survive and replicate intracellularly, thereby provoking impaired maturation of DCs. Therefore, the interaction between DCs and Brucella becomes an interesting model to study the immune response. In this review, we first will describe the most common techniques for DCs differentiation in vitro as well as general features of brucellosis. Then, the interaction of DCs and Brucella, including pathogen recognition, molecular mechanisms of bacterial pathogenesis, and intracellular trafficking of Brucella to subvert innate response, will be reviewed. Finally, we will debate diversity in immunological DC response and the controversial role of DC activation against Brucella infection.
Collapse
Affiliation(s)
- Eric Daniel Avila-Calderón
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, CINVESTAV-IPN, Av. IPN No 2508, Zacatenco, C.P 07330, Mexico city, Mexico
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Santo Tomás, 11340, Mexico city, Mexico
| | - Leopoldo Flores-Romo
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, CINVESTAV-IPN, Av. IPN No 2508, Zacatenco, C.P 07330, Mexico city, Mexico
| | - Witonsky Sharon
- Center for Molecular Medicine and Infectious Diseases/Center for One Health, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061-0442, USA
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061-0442, USA
| | - Luis Donis-Maturano
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Carretera Ensenada-Tijuana 3918, Zona Playitas, 22860, Ensenada, Baja California, Mexico
| | - Miguel Angel Becerril-García
- Departamento de Microbiología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Avenida Francisco I Madero y Dr. Aguirre Pequeño S/N Mitras Centro, 64460, Monterrey, Nuevo León, Mexico
| | - Ma Guadalupe Aguilera Arreola
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Santo Tomás, 11340, Mexico city, Mexico
| | - Beatriz Arellano Reynoso
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico city, Mexico
| | - Francisco Suarez Güemes
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, 04510, Mexico city, Mexico
| | - Araceli Contreras-Rodríguez
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Santo Tomás, 11340, Mexico city, Mexico.
| |
Collapse
|
10
|
Harnanik T, Soeroso J, Suryokusumo MG, Juliandhy T. Effects of Hyperbaric Oxygen on T helper 17/regulatory T Polarization in Antigen and Collagen-induced Arthritis: Hypoxia-inducible Factor-1α as a Target. Oman Med J 2020; 35:e90. [PMID: 31993228 PMCID: PMC6982795 DOI: 10.5001/omj.2020.08] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022] Open
Abstract
Objectives We sought to investigate and prove the effect of hyperbaric oxygen therapy (HBOT) on T helper 17 (Th17)/regulatory T (Treg) cell polarization through changes in the expression of hypoxia-inducible factor-1 alpha (HIF-1α) in rheumatoid arthritis (RA) animal model. Methods We used antigen and collagen-induced arthritis (ACIA) as a RA animal model. Sixteen male BALB/c models of ACIA mice were divided into two groups, the non-HBOT group as the control group and the HBOT group as the treatment group. Expression of HIF-1α, Th17 anti-cluster differentiation 196 (CD196), and Treg anti-interleukine 2 receptor β-chain cells (IL-2Rβ) in tissue from the left knee joint tissue were determined histologically. Oxidative stress and systemic inflammation were assessed by levels of superoxide dismutase (SOD), interleukin 17a (IL-17a), C-reactive protein (CRP), and rheumatoid factor (RF) using the enzyme-linked immune-sorbent assay. The degree of arthritis was assessed by clinical scoring of paw swelling and the diameter of paw swelling. Results We found a significant decrease (p < 0.050) in the expression of HIF-1α, Th17 (CD196), IL-17a, RF levels, and the clinical scores and the diameter of paw swelling when comparing both groups. There was no significant decrease in the level of CRP in the treatment group compared to the control group. The expression of Treg (IL-2Rβ) increased significantly (p < 0.050) and the level of SOD increased but not significantly (p > 0.050) in the treatment group compared to the control group. Conclusions HBOT has effects on the polarization of Th17 to Treg through a decrease in expression of HIF-1α in mice with ACIA. HBOT is recommended for use as a support therapy for RA in combination with drug therapy.
Collapse
Affiliation(s)
- Titut Harnanik
- Department of Hyperbaric, Drs. Med. R. Rijadi S., Phys. Naval Health Institute, Surabaya, Indonesia.,Department of Physiology, Hang Tuah University, Surabaya, Indonesia.,Department of Biochemistry, Unit of the Experimental Animal, Airlangga University, Surabaya, Indonesia
| | - Joewono Soeroso
- Department of Biochemistry, Unit of the Experimental Animal, Airlangga University, Surabaya, Indonesia
| | | | - Tedy Juliandhy
- Department of Electrical Engineering, Hang Tuah University, Surabaya, Indonesia
| |
Collapse
|
11
|
Bunte K, Beikler T. Th17 Cells and the IL-23/IL-17 Axis in the Pathogenesis of Periodontitis and Immune-Mediated Inflammatory Diseases. Int J Mol Sci 2019; 20:ijms20143394. [PMID: 31295952 PMCID: PMC6679067 DOI: 10.3390/ijms20143394] [Citation(s) in RCA: 340] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/11/2019] [Accepted: 07/08/2019] [Indexed: 12/12/2022] Open
Abstract
Innate immunity represents the semi-specific first line of defense and provides the initial host response to tissue injury, trauma, and pathogens. Innate immunity activates the adaptive immunity, and both act highly regulated together to establish and maintain tissue homeostasis. Any dysregulation of this interaction can result in chronic inflammation and autoimmunity and is thought to be a major underlying cause in the initiation and progression of highly prevalent immune-mediated inflammatory diseases (IMIDs) such as psoriasis, rheumatoid arthritis, inflammatory bowel diseases among others, and periodontitis. Th1 and Th2 cells of the adaptive immune system are the major players in the pathogenesis of IMIDs. In addition, Th17 cells, their key cytokine IL-17, and IL-23 seem to play pivotal roles. This review aims to provide an overview of the current knowledge about the differentiation of Th17 cells and the role of the IL-17/IL-23 axis in the pathogenesis of IMIDs. Moreover, it aims to review the association of these IMIDs with periodontitis and briefly discusses the therapeutic potential of agents that modulate the IL-17/IL-23 axis.
Collapse
Affiliation(s)
- Kübra Bunte
- Department of Periodontics, Preventive and Restorative Dentistry, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Thomas Beikler
- Department of Periodontics, Preventive and Restorative Dentistry, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
12
|
The Interleukin-17 Family of Cytokines in Breast Cancer. Int J Mol Sci 2018; 19:ijms19123880. [PMID: 30518157 PMCID: PMC6321268 DOI: 10.3390/ijms19123880] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/26/2018] [Accepted: 11/28/2018] [Indexed: 02/07/2023] Open
Abstract
Breast cancer (BC) is the most common cancer in women worldwide and remains a major cause of mortality with an expected 137,000 death this year in Europe. Standard management of metastatic BC comprises hormonotherapy, chemotherapy, and targeted therapies. Cyclin dependent kinase (CDK) and mammalian target of rapamycin (mTOR) inhibitors have recently proved their efficiency in hormonal receptor expressing BC. Checkpoint proteins inhibition is being evaluated in phase 3 studies. Since inflammation is constantly present in cancers, research teams have focused their attention on the interleukin-17 (IL-17) family of proinflammatory cytokines. Preclinical experiments have reported both pro and antitumor effects depending on the conditions. In the present article, we review the accumulating evidences about the roles of IL-17 in BC and discuss whether this family of cytokines could be a new target in anticancer treatments.
Collapse
|
13
|
Abstract
Psoriasis is a chronic, immune-mediated, inflammatory disease that is pathogenically driven by proinflammatory cytokines. This article reviews the immunologic role of interleukin (IL)-17, the major effector cytokine in the pathogenesis of psoriatic disease, along with the rationale for targeting the IL-17 cytokine family (IL-17A, IL-17F, and IL-17 receptor A) in the treatment of psoriasis and psoriatic arthritis. Emerging evidence indicates that major sources of IL-17A in patients with psoriatic disease are mast cells, γδ T cells, αβ T cells, and innate lymphoid cells in lesional skin and synovial fluid. Within the skin and joints, IL-17A acts on cellular targets, including keratinocytes, neutrophils, endothelial cells, fibroblasts, osteoclasts, chondrocytes, and osteoblasts, to stimulate production of various antimicrobial peptides, chemokines, and proinflammatory and proliferative cytokines, which, in turn, promote tissue inflammation and bone remodeling. The critical importance of the IL-23/IL-17A axis to the pathogenesis of psoriatic disease has resulted in many new biologic treatments targeting these cytokines. These biologics dramatically improve skin and joint symptoms in patients with moderate-to-severe psoriasis and psoriatic arthritis.
Collapse
Affiliation(s)
- Andrew Blauvelt
- Oregon Medical Research Center, 9495 SW Locust St, Suite G, Portland, OR,, 97223, USA.
| | - Andrea Chiricozzi
- Dermatology Unit, Department of Clinical and Translational Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
14
|
Pujol M, Borie C, Montoya M, Ferreira A, Vernal R. Brucella canis induces canine CD4 + T cells multi-cytokine Th1/Th17 production via dendritic cell activation. Comp Immunol Microbiol Infect Dis 2018; 62:68-75. [PMID: 30711049 DOI: 10.1016/j.cimid.2018.11.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 11/23/2018] [Accepted: 11/27/2018] [Indexed: 01/18/2023]
Abstract
Brucella canis is a small intracellular Gram-negative bacterium that frequently leads to chronic infections highly resistant to antibiotic therapy in dogs. Also, it causes mild human brucellosis compared to other zoonotic Brucella spp. Herein we characterize the cellular immune response elicited by B. canis by analysing human and canine CD4+ T cells after stimulation with autologous monocyte-derived dendritic cells (MoDCs). Human and canine B. canis-primed MoDCs stimulated autologous CD4+ T cells; however, a Th1 response was triggered by human MoDCs, whereas canine MoDCs induced Th1/Th17 responses, with increased CD4+ T cells producing IFN-γ and IL-17A simultaneously. Each pattern of cellular response may contribute to host susceptibility, helping to understand the differences in B. canis virulence between these two hosts. In addition, other aspects of canine immunology are unveiled by highlighting the participation of IL-17A-producing canine MoDCs and CD4+ T cells producing IFN-γ and IL-17A.
Collapse
Affiliation(s)
- Myriam Pujol
- Program of Immunology, Institute of Biomedical Sciences ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile; Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.
| | - Consuelo Borie
- Laboratory of Veterinary Bacteriology, Department of Animal Preventive Medicine, Faculty of Veterinary Sciences, Universidad de Chile, Santiago, Chile
| | - María Montoya
- Centro de Investigaciones Biológicas (CIB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Arturo Ferreira
- Program of Immunology, Institute of Biomedical Sciences ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile.
| | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile; Dentistry Unit, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile.
| |
Collapse
|
15
|
Lionakis MS, Levitz SM. Host Control of Fungal Infections: Lessons from Basic Studies and Human Cohorts. Annu Rev Immunol 2017; 36:157-191. [PMID: 29237128 DOI: 10.1146/annurev-immunol-042617-053318] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In the last few decades, the AIDS pandemic and the significant advances in the medical management of individuals with neoplastic and inflammatory conditions have resulted in a dramatic increase in the population of immunosuppressed patients with opportunistic, life-threatening fungal infections. The parallel development of clinically relevant mouse models of fungal disease and the discovery and characterization of several inborn errors of immune-related genes that underlie inherited human susceptibility to opportunistic mycoses have significantly expanded our understanding of the innate and adaptive immune mechanisms that protect against ubiquitous fungal exposures. This review synthesizes immunological knowledge derived from basic mouse studies and from human cohorts and provides an overview of mammalian antifungal host defenses that show promise for informing therapeutic and vaccination strategies for vulnerable patients.
Collapse
Affiliation(s)
- Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892;
| | - Stuart M Levitz
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01655;
| |
Collapse
|
16
|
The crucial roles of Th17-related cytokines/signal pathways in M. tuberculosis infection. Cell Mol Immunol 2017; 15:216-225. [PMID: 29176747 DOI: 10.1038/cmi.2017.128] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/14/2017] [Accepted: 10/15/2017] [Indexed: 12/19/2022] Open
Abstract
Interleukin-17 (IL-17), IL-21, IL-22 and IL-23 can be grouped as T helper 17 (Th17)-related cytokines because they are either produced by Th17/Th22 cells or involved in their development. Here, we review Th17-related cytokines/Th17-like cells, networks/signals and their roles in immune responses or immunity against Mycobacterium tuberculosis (Mtb) infection. Published studies suggest that Th17-related cytokine pathways may be manipulated by Mtb microorganisms for their survival benefits in primary tuberculosis (TB). In addition, there is evidence that immune responses of the signal transducer and activator of transcription 3 (STAT3) signal pathway and Th17-like T-cell subsets are dysregulated or destroyed in patients with TB. Furthermore, Mtb infection can impact upstream cytokines in the STAT3 pathway of Th17-like responses. Based on these findings, we discuss the need for future studies and the rationale for targeting Th17-related cytokines/signals as a potential adjunctive treatment.
Collapse
|
17
|
Pujol M, Castillo F, Alvarez C, Rojas C, Borie C, Ferreira A, Vernal R. Variability in the response of canine and human dendritic cells stimulated with Brucella canis. Vet Res 2017; 48:72. [PMID: 29096717 PMCID: PMC5667440 DOI: 10.1186/s13567-017-0476-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 10/16/2017] [Indexed: 01/30/2023] Open
Abstract
Brucella canis is a small intracellular Gram-negative bacterium whose primary host is the dog, but it also can cause mild human brucellosis. One of the main causes of an inefficient immune response against other species of Brucella is their interaction with dendritic cells (DCs), which affects antigen presentation and impairs the development of an effective Th1 immune response. This study analysed the cytokine pattern production, by RT-qPCR and ELISA, in human and canine DCs against whole B. canis or its purified LPS. Human and canine DCs produced different patterns of cytokines after stimulation with B. canis. In particular, while human DCs produced a Th1-pattern of cytokines (IL-1β, IL-12, and TNF-α), canine cells produced both Th1 and Th17-related cytokines (IL-6, IL-12, IL-17, and IFN-γ). Thus, differences in susceptibility and pathogenicity between these two hosts could be explained, at least partly, by the distinct cytokine patterns observed in this study, where we propose that human DCs induce an effective Th1 immune response to control the infection, while canine DCs lead to a less effective immune response, with the activation of Th17-related response ineffective to control the B. canis infection.
Collapse
Affiliation(s)
- Myriam Pujol
- Doctoral Program in Agronomy Forestry and Veterinary Sciences, Faculty of Veterinary Sciences, Universidad de Chile, Santiago, Chile.,Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Francisca Castillo
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Carla Alvarez
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Camila Rojas
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Consuelo Borie
- Laboratory of Veterinary Bacteriology, Department of Animal Preventive Medicine, Faculty of Veterinary Sciences, Universidad de Chile, Santiago, Chile
| | - Arturo Ferreira
- Program of Immunology, Institute of Biomedical Sciences ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile. .,Dentistry Unit, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile.
| |
Collapse
|
18
|
Carvajal Gonczi CM, Tabatabaei Shafiei M, East A, Martire E, Maurice-Ventouris MHI, Darlington PJ. Reciprocal modulation of helper Th1 and Th17 cells by the β2-adrenergic receptor agonist drug terbutaline. FEBS J 2017; 284:3018-3028. [PMID: 28710773 DOI: 10.1111/febs.14166] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 06/02/2017] [Accepted: 07/12/2017] [Indexed: 01/01/2023]
Abstract
Catecholamine hormones are powerful regulators of the immune system produced by the sympathetic nervous system (SNS). They regulate the adaptive immune system by altering T-cell differentiation into T helper (Th) 1 and Th2 cell subsets, but the effect on Th17 cells is not known. Th17 cells, defined, in part, by chemokine receptor CCR6 and cytokine interleukin (IL)-17A, are crucial for mediating certain pathogen-specific responses and are linked with several autoimmune diseases. We demonstrated that a proportion of human Th17 cells express beta 2-adrenergic receptor (β2AR), a G protein-coupled receptor that responds to catecholamines. Activation of peripheral blood mononuclear cells, which were obtained from venous blood drawn from healthy volunteers, with anti-cluster of differentiation 3 (CD3) and anti-CD28 and with a β2-agonist drug, terbutaline (TERB), augmented IL-17A levels (P < 0.01) in the majority of samples. TERB reduced interferon gamma (IFNγ) indicating that IL-17A and IFNγ are reciprocally regulated. Similar reciprocal regulation was observed with dbcAMP. Proliferation of Th cells was monitored by carboxyfluorescein diacetate N-succinimidyl ester labeling and flow cytometry with antibody staining for CD3 and CD4. TERB increased proliferation by a small but significant margin (P < 0.001). Next, Th17 cells (CD4+ CXCR3- CCR6+ ) were purified using an immunomagnetic positive selection kit, which removes all other mononuclear cells. TERB increased IL-17A from purified Th17 cells, which argues that TERB acts directly on Th17 cells. Thus, hormone signals from the SNS maintain a balance of Th cells subtypes through the β2AR.
Collapse
Affiliation(s)
- Catalina M Carvajal Gonczi
- The Center for Structural and Functional Genomics, Concordia University, Montreal, QC, Canada.,PERFORM Centre, Concordia University, Montreal, QC, Canada.,Department of Biology, Concordia University, Montreal, QC, Canada
| | - Mahdieh Tabatabaei Shafiei
- The Center for Structural and Functional Genomics, Concordia University, Montreal, QC, Canada.,PERFORM Centre, Concordia University, Montreal, QC, Canada.,Department of Biology, Concordia University, Montreal, QC, Canada
| | - Ashley East
- The Center for Structural and Functional Genomics, Concordia University, Montreal, QC, Canada.,PERFORM Centre, Concordia University, Montreal, QC, Canada.,Department of Exercise Science, Concordia University, Montreal, QC, Canada
| | - Erika Martire
- The Center for Structural and Functional Genomics, Concordia University, Montreal, QC, Canada.,PERFORM Centre, Concordia University, Montreal, QC, Canada.,Department of Biology, Concordia University, Montreal, QC, Canada
| | - Meagane H I Maurice-Ventouris
- The Center for Structural and Functional Genomics, Concordia University, Montreal, QC, Canada.,PERFORM Centre, Concordia University, Montreal, QC, Canada.,Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Peter J Darlington
- The Center for Structural and Functional Genomics, Concordia University, Montreal, QC, Canada.,PERFORM Centre, Concordia University, Montreal, QC, Canada.,Department of Biology, Concordia University, Montreal, QC, Canada.,Department of Exercise Science, Concordia University, Montreal, QC, Canada
| |
Collapse
|
19
|
Abstract
INTRODUCTION The IL-23/IL-17 axis is currently considered to be crucial in the pathogenesis of psoriasis. Human IL-23 is primarily produced by antigen-presenting cells and induces and maintains differentiation of Th17 cells and Th22 cells, a primary cellular source of proinflammatory cytokines such as IL-17 and IL-22, which mediate the epidermal hyperplasia, keratinocyte immune activation and tissue inflammation inherent in psoriasis. Agents that target the p40 subunit common to both IL-12 and IL-23 have shown robust clinical activity, but selectivity for IL-23p19 could offer advantages in efficacy and safety with respect to anti-p40 blockade. Areas covered: Relevant references regarding the role of the IL-23/IL-17 pathway in the pathogenesis of psoriasis/psoriatic arthritis and clinical trials with IL-23p40 and IL-23p19 blocking agents were obtained through a literature search in MEDLINE/Pubmed for articles published until November 2016. Moreover, ongoing registered clinical trials (RCTs) of moderate-to-severe psoriasis and psoriatic arthritis were searched through clinicaltrials.gov website, and a manual search was made for pertinent communications at the 2016 American Academy of Dermatology and European Academy of Dermatology and Venereology meetings. Expert commentary: There are potential advantages in selective blockade of the IL23-specific p19 subunit with respect to distal blockade of IL-17A or its receptor. Acting upstream in the IL-23/IL-17 cytokine pathway is likely to reduce the expression of multiple pro-inflammatory cytokines acting on keratinocytes -including IL-17F, IL-21 and IL-22-, in addition to IL-17A. On the other hand, safety data thus far suggest that these drugs might be devoid of some adverse effects of IL-17A blockade that seem to be class related, such as mucocutaneous Candida infections or triggering or worsening of inflammatory bowel disease. Specific IL-23p19 blockade with high-affinity monoclonal antibodies seems to be able to induce long-term remissions of the activity in psoriasis and might eventually represent a paradigm change in the treatment of psoriasis. The results of phase III and comparative head-to-head trials with these agents are eagerly awaited.
Collapse
Affiliation(s)
- Lluís Puig
- a Department of Dermatology, Hospital de la Santa Creu i Sant Pau , Universitat Autònoma de Barcelona , Barcelona , Catalonia , Spain
| |
Collapse
|
20
|
Wang L, Zhao P, Song L, Yan F, Shi C, Li Y, Han M, Lan K. Correlation of Tc17 cells at early stages after allogeneic hematopoietic stem cell transplantation with acute graft-versus-host disease. Int Immunopharmacol 2016; 41:122-126. [DOI: 10.1016/j.intimp.2016.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 11/07/2016] [Accepted: 11/07/2016] [Indexed: 01/08/2023]
|
21
|
Puig L. Interleukin-17 in skin: psoriasis and yonder. Br J Dermatol 2016; 175:859-860. [DOI: 10.1111/bjd.15014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- L. Puig
- Department of Dermatology; Hospital de la Santa Creu i Sant Pau; Universitat Autònoma de Barcelona; Barcelona Catalonia Spain
| |
Collapse
|
22
|
Bystrom J, Clanchy FI, Taher TE, Mangat P, Jawad AS, Williams RO, Mageed RA. TNFα in the regulation of Treg and Th17 cells in rheumatoid arthritis and other autoimmune inflammatory diseases. Cytokine 2016; 101:4-13. [PMID: 27639923 DOI: 10.1016/j.cyto.2016.09.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/31/2016] [Accepted: 09/02/2016] [Indexed: 01/08/2023]
Abstract
TNFα is a principal pro-inflammatory cytokine vital for immunity to infections. However, its excessive production is involved in chronic inflammation and disease pathology in autoimmune diseases. Evidence for its pathogenic role is validated by the fact that its neutralisation by therapeutic agents in vivo is beneficial in ameliorating disease and controlling symptoms. Paradoxically, however, treatment with TNFα inhibitors can either have no clinical effects, or even exacerbate disease in some patients. The explanation for such contradictory outcomes may lay in how and which downstream signalling pathways are activated and drive disease. TNFα causes its effects by binding to either or both of two membrane-bound receptors, TNFR1 and TNFR2. Engagement of the receptors can induce cell death or cell proliferation. T cells both produce and respond to TNFα and depending on whether the cytokine is membrane-bound or soluble and the level of expression of its two receptors, the biological outcome can be distinct. In addition, polymorphisms in genes encoding TNFα and T cell signalling proteins can significantly impact the outcome of TNFα receptor engagement. Early studies revealed that effector T cells in patients with rheumatoid arthritis (RA) are hyporesponsive due to chronic exposure to TNFα. However, recent evidence indicates that the relationship between TNFα and T cell responses is complex and, at times, can be paradoxical. In addition, there is controversy as to the specific effects of TNFα on different T cell subsets. This review will summarise knowledge on how TNFα modulates T cell responses and the effect of engaging either of its two receptors. Furthermore, we discuss how such interactions can dictate the outcome of treatment with TNFα inhibitors.
Collapse
Affiliation(s)
- Jonas Bystrom
- Experimental Medicine and Rheumtology, William Harvey Research Institute, Queen Mary University of London, London, UK.
| | - F I Clanchy
- Kennedy Institute of Rheumatology, Oxford University, Oxford, UK
| | - Taher E Taher
- Experimental Medicine and Rheumtology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Pam Mangat
- Department of Rheumatology, Royal Free Hospital, NHS Foundation Trust, London, UK
| | - Ali S Jawad
- Experimental Medicine and Rheumtology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | - Rizgar A Mageed
- Experimental Medicine and Rheumtology, William Harvey Research Institute, Queen Mary University of London, London, UK
| |
Collapse
|
23
|
Fang S, Huang Y, Wang S, Zhang Y, Luo X, Liu L, Zhong S, Liu X, Li D, Liang R, Miranda P, Gu P, Zhou H, Fan X, Li B. IL-17A Exacerbates Fibrosis by Promoting the Proinflammatory and Profibrotic Function of Orbital Fibroblasts in TAO. J Clin Endocrinol Metab 2016; 101:2955-65. [PMID: 27224264 DOI: 10.1210/jc.2016-1882] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
CONTEXT The development of thyroid-associated ophthalmopathy (TAO) is associated with self-immune dysfunction. Recent findings in TAO and Graves' disease indicate that IL-17A may also be involved in the autoimmunity of TAO. OBJECTIVE We sought to investigate the pathogenic function of IL-17A-producing T cells in TAO. DESIGN/SETTING/PARTICIPANTS Blood samples and orbital fibroblasts (OFs) were collected from TAO patients and healthy subjects. MAIN OUTCOME MEASURES Flow cytometry, real-time PCR, cytokine-specific ELISA, and Western blotting were performed. RESULTS Here, we showed a significantly higher proportion of IL-17A-producing T cells in TAO patients and the recruitment of both CD4(+) and CD8(+) T cells in TAO orbits. TAO orbital tissues expressed more IL-17A receptor, IL-17A, and its related cytokines, with severe fibrotic change compared with normal controls. Furthermore, we validated that IL-17A could enhance the proinflammatory function of OFs and stimulate the production of extracellular matrix proteins in OFs but not eyelid fibroblasts. The mechanisms involved in this enhancement mainly relied on MAPK activation. Finally, we observed that the deubiquitinase inhibitor vialinin A could down-regulate retinoic acid receptor-related orphan receptor-γt expression and decrease IL-17A level in TAO patients. CONCLUSION Our observations illustrate the potential pathogenic role of IL-17A-producing T cells in the inflammatory response and fibrosis of TAO. The effect of vialinin A on the reduction of retinoic acid receptor-related orphan receptor-γt level implicates its potential role as a novel therapeutic agent for TAO and other autoimmune disorders in the future.
Collapse
Affiliation(s)
- Sijie Fang
- Department of Ophthalmology (S.F., Y.H., Y.Z., S.Z., X.Li., P.G., H.Z., X.F.), Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011; and Key Laboratory of Molecular Virology and Immunology (S.F., S.W., X.Lu., L.L., D.L., R.L., P.M., B.L.), Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031
| | - Yazhuo Huang
- Department of Ophthalmology (S.F., Y.H., Y.Z., S.Z., X.Li., P.G., H.Z., X.F.), Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011; and Key Laboratory of Molecular Virology and Immunology (S.F., S.W., X.Lu., L.L., D.L., R.L., P.M., B.L.), Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031
| | - Shuaiwei Wang
- Department of Ophthalmology (S.F., Y.H., Y.Z., S.Z., X.Li., P.G., H.Z., X.F.), Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011; and Key Laboratory of Molecular Virology and Immunology (S.F., S.W., X.Lu., L.L., D.L., R.L., P.M., B.L.), Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031
| | - Yidan Zhang
- Department of Ophthalmology (S.F., Y.H., Y.Z., S.Z., X.Li., P.G., H.Z., X.F.), Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011; and Key Laboratory of Molecular Virology and Immunology (S.F., S.W., X.Lu., L.L., D.L., R.L., P.M., B.L.), Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031
| | - Xuerui Luo
- Department of Ophthalmology (S.F., Y.H., Y.Z., S.Z., X.Li., P.G., H.Z., X.F.), Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011; and Key Laboratory of Molecular Virology and Immunology (S.F., S.W., X.Lu., L.L., D.L., R.L., P.M., B.L.), Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031
| | - Luyan Liu
- Department of Ophthalmology (S.F., Y.H., Y.Z., S.Z., X.Li., P.G., H.Z., X.F.), Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011; and Key Laboratory of Molecular Virology and Immunology (S.F., S.W., X.Lu., L.L., D.L., R.L., P.M., B.L.), Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031
| | - Sisi Zhong
- Department of Ophthalmology (S.F., Y.H., Y.Z., S.Z., X.Li., P.G., H.Z., X.F.), Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011; and Key Laboratory of Molecular Virology and Immunology (S.F., S.W., X.Lu., L.L., D.L., R.L., P.M., B.L.), Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031
| | - Xingtong Liu
- Department of Ophthalmology (S.F., Y.H., Y.Z., S.Z., X.Li., P.G., H.Z., X.F.), Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011; and Key Laboratory of Molecular Virology and Immunology (S.F., S.W., X.Lu., L.L., D.L., R.L., P.M., B.L.), Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031
| | - Dan Li
- Department of Ophthalmology (S.F., Y.H., Y.Z., S.Z., X.Li., P.G., H.Z., X.F.), Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011; and Key Laboratory of Molecular Virology and Immunology (S.F., S.W., X.Lu., L.L., D.L., R.L., P.M., B.L.), Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031
| | - Rui Liang
- Department of Ophthalmology (S.F., Y.H., Y.Z., S.Z., X.Li., P.G., H.Z., X.F.), Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011; and Key Laboratory of Molecular Virology and Immunology (S.F., S.W., X.Lu., L.L., D.L., R.L., P.M., B.L.), Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031
| | - Piccioni Miranda
- Department of Ophthalmology (S.F., Y.H., Y.Z., S.Z., X.Li., P.G., H.Z., X.F.), Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011; and Key Laboratory of Molecular Virology and Immunology (S.F., S.W., X.Lu., L.L., D.L., R.L., P.M., B.L.), Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031
| | - Ping Gu
- Department of Ophthalmology (S.F., Y.H., Y.Z., S.Z., X.Li., P.G., H.Z., X.F.), Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011; and Key Laboratory of Molecular Virology and Immunology (S.F., S.W., X.Lu., L.L., D.L., R.L., P.M., B.L.), Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031
| | - Huifang Zhou
- Department of Ophthalmology (S.F., Y.H., Y.Z., S.Z., X.Li., P.G., H.Z., X.F.), Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011; and Key Laboratory of Molecular Virology and Immunology (S.F., S.W., X.Lu., L.L., D.L., R.L., P.M., B.L.), Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031
| | - Xianqun Fan
- Department of Ophthalmology (S.F., Y.H., Y.Z., S.Z., X.Li., P.G., H.Z., X.F.), Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011; and Key Laboratory of Molecular Virology and Immunology (S.F., S.W., X.Lu., L.L., D.L., R.L., P.M., B.L.), Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031
| | - Bin Li
- Department of Ophthalmology (S.F., Y.H., Y.Z., S.Z., X.Li., P.G., H.Z., X.F.), Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011; and Key Laboratory of Molecular Virology and Immunology (S.F., S.W., X.Lu., L.L., D.L., R.L., P.M., B.L.), Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences Medical School, Chinese Academy of Sciences, Shanghai 200031
| |
Collapse
|
24
|
Wang Y, Wang LL, Yang HY, Wang FF, Zhang XX, Bai YP. Interleukin-21 is associated with the severity of psoriasis vulgaris through promoting CD4+ T cells to differentiate into Th17 cells. Am J Transl Res 2016; 8:3188-3196. [PMID: 27508040 PMCID: PMC4969456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 05/11/2016] [Indexed: 06/06/2023]
Abstract
Interleukin-21 (IL-21) and T helper 17 (Th17) cells are known to be involved in the pathogenesis of psoriasis, but little is known about their relationship in psoriasis. Herein, we investigated whether IL-21 could regulate Th17 cell induction in patients with psoriasis vulgaris. 32 patients with psoriasis vulgaris and 13 healthy controls were recruited. Flow cytometry was used to detect the frequencies of cells mainly secreting IL-21 (including IL-21+CD4+ T and IL-21+ Th17 cells) and Th17 cells. An enzyme-linked immunosorbent assay (ELISA) was used to determine the serum content of IL-21. Severity of the psoriasis was evaluated by a Psoriasis Area and Severity Index (PASI) score. In addition, the differentiation of CD4+ T cells with IL-21 and the different frequencies of IL-21+CD4+ T cells, IL-21+ Th17 cells and Th17 cells were assessed, as were serum levels of IL-21 in patients with moderate to severe psoriasis before and after treatment. Our results showed that the levels of IL-21, IL-21+CD4+ T cells, IL-21+ Th17 cells and Th17 cells were significantly increased in patients and positively associated with PASI score (P < 0.01). Moreover, the levels of IL-21, IL-21+CD4+ T cells and IL-21+ Th17 cells were positively correlated with the frequency of Th17 cells (P < 0.01). In vitro experiments demonstrated that IL-21 could promote CD4+ T cells to differentiate into Th17 cells. After a 4-week treatment of acitretin and a topical therapy, all the immune markers observed in patients decreased significantly (P < 0.01), but the levels remained higher than those in healthy controls (P < 0.01). These findings indicate that IL-21 might promote Th17 cell induction in psoriasis and might be a potential immune marker for targeting this disease.
Collapse
Affiliation(s)
- Ying Wang
- Clinical Institute of China-Japan Friendship Hospital, Graduate School of Peking Union Medical CollegeBeijing, China
- Department of Dermatology & Venerology, China-Japan Friendship HospitalBeijing, China
| | - Li-Li Wang
- Clinical Institute of China-Japan Friendship Hospital, Graduate School of Peking Union Medical CollegeBeijing, China
- Department of Dermatology & Venerology, China-Japan Friendship HospitalBeijing, China
| | - Hao-Yu Yang
- Department of Dermatology & Venerology, China-Japan Friendship HospitalBeijing, China
- Beijing University of Chinese MedicineBeijing, China
| | - Fei-Fei Wang
- Department of Dermatology & Venerology, China-Japan Friendship HospitalBeijing, China
- Beijing University of Chinese MedicineBeijing, China
| | - Xue-Xiu Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou UniversityZhengzhou, Henan, China
| | - Yan-Ping Bai
- Clinical Institute of China-Japan Friendship Hospital, Graduate School of Peking Union Medical CollegeBeijing, China
- Department of Dermatology & Venerology, China-Japan Friendship HospitalBeijing, China
| |
Collapse
|
25
|
Zhou Q, Bajo MA, Del Peso G, Yu X, Selgas R. Preventing peritoneal membrane fibrosis in peritoneal dialysis patients. Kidney Int 2016; 90:515-24. [PMID: 27282936 DOI: 10.1016/j.kint.2016.03.040] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 03/13/2016] [Accepted: 03/24/2016] [Indexed: 12/11/2022]
Abstract
Long-term peritoneal dialysis causes morphologic and functional changes in the peritoneal membrane. Although mesothelial-mesenchymal transition of peritoneal mesothelial cells is a key process leading to peritoneal fibrosis, and bioincompatible peritoneal dialysis solutions (glucose, glucose degradation products, and advanced glycation end products or a combination) are responsible for altering mesothelial cell function and proliferation, mechanisms underlying these processes remain largely unclear. Peritoneal fibrosis has 2 cooperative parts, the fibrosis process itself and the inflammation. The link between these 2 processes is frequently bidirectional, with each one inducing the other. This review outlines our current understanding about the definition and pathophysiology of peritoneal fibrosis, recent studies on key fibrogenic molecular machinery in peritoneal fibrosis, such as the role of transforming growth factor-β/Smads, transforming growth factor-β β/Smad independent pathways, and noncoding RNAs. The diagnosis of peritoneal fibrosis, including effluent biomarkers and the histopathology of a peritoneal biopsy, which is the gold standard for demonstrating peritoneal fibrosis, is introduced in detail. Several interventions for peritoneal fibrosis based on biomarkers, cytology, histology, functional studies, and antagonists are presented in this review. Recent experimental trials in animal models, including pharmacology and gene therapy, which could offer novel insights into the treatment of peritoneal fibrosis in the near future, are also discussed in depth.
Collapse
Affiliation(s)
- Qin Zhou
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - M-Auxiliadora Bajo
- Nephrology Service, Hospital Universitario La Paz, IdiPAZ, REDinREN, Fibroteam, IRSIN, Madrid, Spain
| | - Gloria Del Peso
- Nephrology Service, Hospital Universitario La Paz, IdiPAZ, REDinREN, Fibroteam, IRSIN, Madrid, Spain
| | - Xueqing Yu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rafael Selgas
- Nephrology Service, Hospital Universitario La Paz, IdiPAZ, REDinREN, Fibroteam, IRSIN, Madrid, Spain
| |
Collapse
|
26
|
Bao Z, Lu G, Cui D, Yao Y, Yang G, Zhou J. IL-17A-producing T cells are associated with the progression of lung adenocarcinoma. Oncol Rep 2016; 36:641-50. [PMID: 27277161 PMCID: PMC4933549 DOI: 10.3892/or.2016.4837] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 02/20/2016] [Indexed: 12/13/2022] Open
Abstract
Accumulating evidence has shown that T cells are crucial in shaping the tumor microenvironment and regulating tumor development. However, the roles of IL-17A-producing T cells (IL-17A+CD4+ Th17, IL-17A+CD8+ Tc17 and IL-17A+ γδT17 cells) and related cytokines in the progression of lung cancer (LC) remain uncertain. Here, we found that the frequencies of both Th17 and γδT17 cells in the peripheral blood of patients with lung adenocarcinoma (LA) were higher than those in healthy controls (HCs), whereas the frequency of Tc17 cells in the patients with LA was decreased. In addition, the frequencies of circulating Th17 and γδT17 cells, but not Tc17 cells, were positively associated with tumor invasion and metastasis. Furthermore, the major source of IL-17A production was Th17 cells, followed by Tc17 and γδT17 cells, in peripheral blood from patients with LA and HCs; but the percentages of Th17 and γδT17 cells in total intracellular IL-17A+ cells obtained from the patients with LC were higher than those from HCs. Moreover, the protein and corresponding mRNA levels of IL-17A, IL-23, IL-1β, and TGF-β1 were much higher in the patients with LA than those in HCs, and the levels of IL-17A in patients were positively correlated with numbers of both Th17 and γδT17 cells, but not Tc17 cells. Finally, the frequencies of circulating Th17 and γδT17 cells, along with the levels of IL-17A, IL-23, IL-1β, and TGF-β1 were decreased in the patients with LA after tumor resection, whereas the frequency of circulating Tc17 cells was inversely increased in these patients. Our findings indicate that Th17, Tc17, γδT17 cells, and IL-17A-associated cytokines contribute to the development of LA and thus represent promising targets for therapeutic strategies.
Collapse
Affiliation(s)
- Zhang Bao
- Department of Respiratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Guohua Lu
- Department of Respiratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Dawei Cui
- Center of Clinical Laboratory, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Yinan Yao
- Department of Respiratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Guangdie Yang
- Department of Respiratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Jianying Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
27
|
Kawalkowska J, Quirke AM, Ghari F, Davis S, Subramanian V, Thompson PR, Williams RO, Fischer R, La Thangue NB, Venables PJ. Abrogation of collagen-induced arthritis by a peptidyl arginine deiminase inhibitor is associated with modulation of T cell-mediated immune responses. Sci Rep 2016; 6:26430. [PMID: 27210478 PMCID: PMC4876390 DOI: 10.1038/srep26430] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/03/2016] [Indexed: 12/15/2022] Open
Abstract
Proteins containing citrulline, a post-translational modification of arginine, are generated by peptidyl arginine deiminases (PAD). Citrullinated proteins have pro-inflammatory effects in both innate and adaptive immune responses. Here, we examine the therapeutic effects in collagen-induced arthritis of the second generation PAD inhibitor, BB-Cl-amidine. Treatment after disease onset resulted in the reversal of clinical and histological changes of arthritis, associated with a marked reduction in citrullinated proteins in lymph nodes. There was little overall change in antibodies to collagen or antibodies to citrullinated peptides, but a shift from pro-inflammatory Th1 and Th17-type responses to pro-resolution Th2-type responses was demonstrated by serum cytokines and antibody subtypes. In lymph node cells from the arthritic mice treated with BB-Cl-amidine, there was a decrease in total cell numbers but an increase in the proportion of Th2 cells. BB-Cl-amidine had a pro-apoptotic effect on all Th subsets in vitro with Th17 cells appearing to be the most sensitive. We suggest that these immunoregulatory effects of PAD inhibition in CIA are complex, but primarily mediated by transcriptional regulation. We suggest that targeting PADs is a promising strategy for the treatment of chronic inflammatory disease.
Collapse
Affiliation(s)
- Joanna Kawalkowska
- Kennedy Institute, Nuffield Department of Orthopaedics, Rheumatology & Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, UK
| | - Anne-Marie Quirke
- Kennedy Institute, Nuffield Department of Orthopaedics, Rheumatology & Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, UK
| | - Fatemeh Ghari
- Laboratory of Cancer Biology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Simon Davis
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK
| | - Venkataraman Subramanian
- Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, LRB 826, 364 Plantation Street, Worcester, MA, 01605, USA
| | - Paul R. Thompson
- Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, LRB 826, 364 Plantation Street, Worcester, MA, 01605, USA
| | - Richard O. Williams
- Kennedy Institute, Nuffield Department of Orthopaedics, Rheumatology & Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, UK
| | - Roman Fischer
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK
| | - Nicholas B. La Thangue
- Laboratory of Cancer Biology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Patrick J. Venables
- Kennedy Institute, Nuffield Department of Orthopaedics, Rheumatology & Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, UK
| |
Collapse
|
28
|
Immune Checkpoint Modulation in Colorectal Cancer: What's New and What to Expect. J Immunol Res 2015; 2015:158038. [PMID: 26605342 PMCID: PMC4641952 DOI: 10.1155/2015/158038] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/11/2015] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC), as one of the most prevalent types of cancer worldwide, is still a leading cause of cancer related mortality. There is an urgent need for more efficient therapies in metastatic disease. Immunotherapy, a rapidly expanding field of oncology, is designed to boost the body's natural defenses to fight cancer. Of the many approaches currently under study to improve antitumor immune responses, immune checkpoint inhibition has thus far been proven to be the most effective. This review will outline the treatments that take advantage of our growing understanding of the role of the immune system in cancer, with a particular emphasis on immune checkpoint molecules, involved in CRC pathogenesis.
Collapse
|