1
|
Baek H, Yang SW, Kim S, Lee Y, Park H, Park M, Jeon BJ, Park H, Hwang HS, Kim JY, Kim JH, Kang YS. Development of Anti-Inflammatory Agents Utilizing DC-SIGN Mediated IL-10 Secretion in Autoimmune and Immune-Mediated Disorders: Bridging Veterinary and Human Health. Int J Mol Sci 2025; 26:2329. [PMID: 40076949 PMCID: PMC11901132 DOI: 10.3390/ijms26052329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
DC-SIGN (dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin) is a C-type lectin receptor expressed on dendritic cells and M2 macrophages, playing a key role in immune regulation and pathogen recognition. Its ability to mediate anti-inflammatory effects by interacting with specific ligands triggers pathways that suppress pro-inflammatory responses and promote tissue repair, making it a potential therapeutic target for inflammatory and autoimmune diseases. DC-SIGN homologs in various animal species share structural similarities and perform comparable immune functions, offering valuable insights into its broader application across species. By recognizing carbohydrate ligands on pathogens, DC-SIGN facilitates immune modulation, which can be harnessed for developing therapies aimed at controlling inflammation. In veterinary medicine, autoimmune and inflammatory diseases, such as rheumatoid arthritis and inflammatory bowel disease, represent significant challenges, and the anti-inflammatory properties of DC-SIGN could provide new therapeutic options to improve disease management and enhance animal health. Future investigations should focus on the structural and functional analysis of DC-SIGN homologs in various species, as well as the development of preclinical models to translate these findings into clinical interventions bridging veterinary and human health.
Collapse
Affiliation(s)
- Hayeon Baek
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; (H.B.); (M.P.)
| | - Seung-Woo Yang
- Sanford Consortium for Regenerative Medicine, School of Medicine, University of California, San Diego, CA 92037, USA;
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, Research Institute of Medical Science, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea;
| | - Seulki Kim
- Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.K.); (Y.L.)
| | - Yunseok Lee
- Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.K.); (Y.L.)
| | - Hwi Park
- Department of Veterinary Ophthalmology, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.P.); (B.-J.J.); (H.P.); (J.-Y.K.)
| | - Min Park
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; (H.B.); (M.P.)
| | - Byung-Ju Jeon
- Department of Veterinary Ophthalmology, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.P.); (B.-J.J.); (H.P.); (J.-Y.K.)
| | - Hanwool Park
- Department of Veterinary Ophthalmology, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.P.); (B.-J.J.); (H.P.); (J.-Y.K.)
| | - Han-Sung Hwang
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, Research Institute of Medical Science, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea;
| | - Joon-Young Kim
- Department of Veterinary Ophthalmology, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.P.); (B.-J.J.); (H.P.); (J.-Y.K.)
| | - Jung-Hyun Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea;
| | - Young-Sun Kang
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; (H.B.); (M.P.)
- Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.K.); (Y.L.)
| |
Collapse
|
2
|
Sabljić L, Radulović N, Đokić J, Stojanovic DB, Radojević D, Glamočlija S, Dinić M, Golić N, Vasilev S, Uskoković P, Sofronić-Milosavljević L, Gruden-Movsesijan A, Tomić S. Biodegradable Electrospun PLGA Nanofibers-Encapsulated Trichinella Spiralis Antigens Protect from Relapsing Experimental Autoimmune Encephalomyelitis and Related Gut Microbiota Dysbiosis. Int J Nanomedicine 2025; 20:1921-1948. [PMID: 39963417 PMCID: PMC11830953 DOI: 10.2147/ijn.s499161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/18/2025] [Indexed: 02/20/2025] Open
Abstract
Purpose Trichinella spiralis has evolved complex immunomodulatory mechanisms mediated by excretory-secretory products (ESL1) that enable its survival in the host. Consequently, ESL1 antigens display excellent potential for treating autoimmune diseases such as multiple sclerosis (MS). However, whether timely controlled delivery of ESL1 antigens in vivo, as in natural infections, could enhance its therapeutic potential for MS is still unknown. Methods To test this, we encapsulated ESL1 antigens into biodegradable poly (lactide-co-glycolic) acid (PLGA) nanofibers by emulsion electrospinning as a delivery system and assessed their release dynamics in vitro, and in an animal MS model, experimental autoimmune encephalomyelitis (EAE), induced 7 days after PLGA/ESL1 subcutaneous implantation. PLGA/ESL1 effects on EAE symptoms were monitored along with multiple immune cell subsets in target organs at the peak and recovery of EAE. Gut barrier function and microbiota composition were analyzed using qPCR, 16S rRNA sequencing, and metabolomic analyses. Results ESL1 antigens, released from PLGA and drained via myeloid antigen-presenting cells through lymph nodes, protected the animals from developing EAE symptoms. These effects correlated with reduced activation of myeloid cells, increased IL-10 expression, and reduced accumulation of proinflammatory natural killer (NK) cells, T helper (Th)1 and Th17 cells in the spleen and central nervous system (CNS). Additionally, CD4+CD25hiFoxP3+ regulatory T cells and IL-10-producing B cells were expanded in PLGA/ESL1-treated animals, compared to control animals. The migration of ESL1 to the guts correlated with locally reduced inflammation and gut barrier damage. Additionally, PLGA/ESL1-treated animals displayed an unaltered microbiota characterized only by a more pronounced protective mevalonate pathway and expanded short-chain fatty acid-producing bacteria, which are known to suppress inflammation. Conclusion The delivery of T. spiralis ESL1 antigens via biodegradable electrospun PLGA nanofiber implants efficiently protected the animals from developing EAE by inducing a beneficial immune response in the spleen, gut, and CNS. This platform provides excellent grounds for further development of novel MS therapies.
Collapse
Affiliation(s)
- Ljiljana Sabljić
- Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Nataša Radulović
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Belgrade, Serbia
| | - Jelena Đokić
- Institute for Molecular Genetics and Genetical Engineering, University of Belgrade, Belgrade, Serbia
| | - Dusica B Stojanovic
- Faculty of Metallurgy and Technology, University of Belgrade, Belgrade, Serbia
| | - Dušan Radojević
- Institute for Molecular Genetics and Genetical Engineering, University of Belgrade, Belgrade, Serbia
| | - Sofija Glamočlija
- Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Miroslav Dinić
- Institute for Molecular Genetics and Genetical Engineering, University of Belgrade, Belgrade, Serbia
| | - Nataša Golić
- Institute for Molecular Genetics and Genetical Engineering, University of Belgrade, Belgrade, Serbia
| | - Saša Vasilev
- Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Petar Uskoković
- Faculty of Metallurgy and Technology, University of Belgrade, Belgrade, Serbia
| | | | | | - Sergej Tomić
- Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
3
|
Zhu Y, Chen X, Zheng H, Ma Q, Chen K, Li H. Anti-Inflammatory Effects of Helminth-Derived Products: Potential Applications and Challenges in Diabetes Mellitus Management. J Inflamm Res 2024; 17:11789-11812. [PMID: 39749005 PMCID: PMC11694023 DOI: 10.2147/jir.s493374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/15/2024] [Indexed: 01/04/2025] Open
Abstract
The global rise in diabetes mellitus (DM), particularly type 2 diabetes (T2D), has become a major public health challenge. According to the "hygiene hypothesis", helminth infections may offer therapeutic benefits for DM. These infections are known to modulate immune responses, reduce inflammation, and improve insulin sensitivity. However, they also carry risks, such as malnutrition, anemia, and intestinal obstruction. Importantly, helminth excretory/secretory products, which include small molecules and proteins, have shown therapeutic potential in treating various inflammatory diseases with minimal side effects. This review explores the anti-inflammatory properties of helminth derivatives and their potential to alleviate chronic inflammation in both type 1 diabetes and T2D, highlighting their promise as future drug candidates. Additionally, it discusses the possible applications of these derivatives in DM management and the challenges involved in translating these findings into clinical practice.
Collapse
Affiliation(s)
- Yunhuan Zhu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Xintong Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Hezheng Zheng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Qiman Ma
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Keda Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
| | - Hongyu Li
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People’s Republic of China
- Ocean College, Beibu Gulf University, Qinzhou, Guangxi, People’s Republic of China
| |
Collapse
|
4
|
Wang J, Gao M, Wang J, Zeng Y, Wang C, Cao X. LGG promotes activation of intestinal ILC3 through TLR2 receptor and inhibits salmonella typhimurium infection in mice. Virulence 2024; 15:2384553. [PMID: 39080852 PMCID: PMC11296546 DOI: 10.1080/21505594.2024.2384553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 08/04/2024] Open
Abstract
Salmonella is a foodborne pathogen that causes disruption of intestinal mucosal immunity, leading to acute gastroenteritis in the host. In this study, we found that Salmonella Typhimurium (STM) infection of the intestinal tract of mice led to a significant increase in the proportion of Lacticaseibacillus, while the secretion of IL-22 from type 3 innate lymphoid cells (ILC3) increased significantly. Feeding Lacticaseibacillus rhamnosus GG (LGG) effectively alleviated the infection of STM in the mouse intestines. TLR2-/- mice experiments found that TLR2-expressing dendritic cells (DCs) are crucial for LGG's activation of ILC3. Subsequent in vitro experiments showed that heat-killed LGG (HK-LGG) could promote DCs to secrete IL-23, which in turn further promotes the activation of ILC3 and the secretion of IL-22. Finally, organoid experiments further verified that IL-22 secreted by ILC3 can enhance the intestinal mucosal immune barrier and inhibit STM infection. This study demonstrates that oral administration of LGG is a potential method for inhibiting STM infection.
Collapse
Affiliation(s)
- Junhong Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Ming Gao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jiarui Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chunfeng Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
5
|
Fawzy EM, Selim MA, Mostafa NE, Abdelhameed RM, Darwish AM, Yousef AM, Alabiad MA, Ibrahim MN, Fawzy HM, Abdel Hamed EF. The prophylactic and therapeutic impact of Trichinella spiralis larvae excretory secretory antigens- loaded Ca-BTC metal organic frameworks on induced murine colitis. J Helminthol 2024; 98:e41. [PMID: 38785193 DOI: 10.1017/s0022149x24000191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Background: Inflammatory bowel disease is an autoimmune disease that affects the gut. T. spiralis larvae (E/S Ags) loaded on calcium-benzene-1,3,5-tricarboxylate metal-organic frameworks (Ca-BTC MOFs) were tested to determine whether they might prevent or cure acetic acid-induced murine colitis. Methods: T. spiralis larvae E/S Ags/Ca-BTC MOFs were used in prophylactic and therapeutic groups to either precede or follow the development of murine colitis. On the seventh day after colitis, mice were slaughtered. The effect of our target antigens on the progress of the colitis was evaluated using a variety of measures, including survival rate, disease activity index, colon weight/bodyweight, colon weight/length) ratios, and ratings for macroscopic and microscopic colon damage. The levels of inflammatory cytokines (interferon-γ and interleukin-4), oxidative stress marker malondialdehyde, and glutathione peroxidase in serum samples were evaluated. Foxp3 T-reg expression was carried out in colonic and splenic tissues. Results: T. spiralis larvae E/S Ags/Ca-BTC MOFs were the most effective in alleviating severe inflammation in murine colitis. The survival rate, disease activity index score, colon weight/length and colon weight/bodyweight ratios, and gross and microscopic colon damage scores have all considerably improved. A large decrease in proinflammatory cytokine (interferon-γ) and oxidative stress marker (malondialdehyde) expression and a significant increase in interleukin-4 and glutathione peroxidase expression were obtained. The expression of Foxp3+ Treg cells was elevated in colonic and splenic tissues. Conclusion: T. spiralis larvae E/S Ags/Ca-BTC MOFs had the highest anti-inflammatory, antioxidant, and cytoprotective capabilities against murine colitis and might be used to develop new preventative and treatment strategies.
Collapse
Affiliation(s)
- E M Fawzy
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, Sharkia, Egypt
| | - M A Selim
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, Sharkia, Egypt
| | - N E Mostafa
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, Sharkia, Egypt
| | - R M Abdelhameed
- Department of Applied Organic Chemistry, National Research Centre, Dokki, Giza, Egypt
| | - A M Darwish
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, Sharkia, Egypt
| | - A M Yousef
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, Sharkia, Egypt
| | - M A Alabiad
- Department of Pathology, Faculty of Medicine, Zagazig University, Egypt
| | - M N Ibrahim
- Department of Clinical Laboratories, College of applied Medical Sciences, Jouf University, Qurrayat77451, KSA
| | - H M Fawzy
- Department of Community, Faculty of Medicine, Zagazig University, Sharkia, Egypt
| | - E F Abdel Hamed
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, Sharkia, Egypt
| |
Collapse
|
6
|
Kang C, Li X, Liu P, Liu Y, Niu Y, Zeng X, Zhao H, Liu J, Qiu S. Tolerogenic dendritic cells and TLR4/IRAK4/NF-κB signaling pathway in allergic rhinitis. Front Immunol 2023; 14:1276512. [PMID: 37915574 PMCID: PMC10616250 DOI: 10.3389/fimmu.2023.1276512] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 10/05/2023] [Indexed: 11/03/2023] Open
Abstract
Dendritic cells (DCs), central participants in the allergic immune response, can capture and present allergens leading to allergic inflammation in the immunopathogenesis of allergic rhinitis (AR). In addition to initiating antigen-specific immune responses, DCs induce tolerance and modulate immune homeostasis. As a special type of DCs, tolerogenic DCs (tolDCs) achieve immune tolerance mainly by suppressing effector T cell responses and inducing regulatory T cells (Tregs). TolDCs suppress allergic inflammation by modulating immune tolerance, thereby reducing symptoms of AR. Activation of the TLR4/IRAK4/NF-κB signaling pathway contributes to the release of inflammatory cytokines, and inhibitors of this signaling pathway induce the production of tolDCs to alleviate allergic inflammatory responses. This review focuses on the relationship between tolDCs and TLR4/IRAK4/NF-κB signaling pathway with AR.
Collapse
Affiliation(s)
- Chenglin Kang
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, China
- Department of Otolaryngology, Longgang E.N.T Hospital and Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, China
- Department of Otolaryngology, Second People’s Hospital of Gansu Province, Lanzhou, China
| | - Xiaomei Li
- Department of Otolaryngology, Second People’s Hospital of Gansu Province, Lanzhou, China
| | - Peng Liu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, China
| | - Yue Liu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, China
| | - Yuan Niu
- Department of Neurology, Second People’s Hospital of Gansu Province, Lanzhou, China
| | - Xianhai Zeng
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, China
- Department of Otolaryngology, Longgang E.N.T Hospital and Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, China
| | - Hailiang Zhao
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, China
- Department of Otolaryngology, Longgang E.N.T Hospital and Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, China
| | - Jiangqi Liu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, China
- Department of Otolaryngology, Longgang E.N.T Hospital and Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, China
| | - Shuqi Qiu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, China
- Department of Otolaryngology, Longgang E.N.T Hospital and Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, China
| |
Collapse
|
7
|
Ni Y, Xiong R, Zhu Y, Luan N, Yu C, Yang K, Wang H, Xu X, Yang Y, Sun S, Shi L, Padde JR, Chen L, Chen L, Hou M, Xu Z, Lai R, Ji M. A target-based discovery from a parasitic helminth as a novel therapeutic approach for autoimmune diseases. EBioMedicine 2023; 95:104751. [PMID: 37579625 PMCID: PMC10448429 DOI: 10.1016/j.ebiom.2023.104751] [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: 03/29/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Regulatory T cells (Tregs) can alleviate the development of autoimmune and inflammatory diseases, thereby proposing their role as a new therapeutic strategy. Parasitic helminths have co-evolved with hosts to generate immunological privilege and immune tolerance through inducing Tregs. Thus, constructing a "Tregs-induction"-based discovery pipeline from parasitic helminth is a promising strategy to control autoimmune and inflammatory diseases. METHODS The gel filtration chromatography and reverse-phase high-performance liquid chromatography (RP-HPLC) were used to isolate immunomodulatory components from the egg extracts of Schistosoma japonicum. The extracted peptides were evaluated for their effects on Tregs suppressive functions using flow cytometry, ELISA and T cell suppression assay. Finally, we carried out colitis and psoriasis models to evaluate the function of Tregs induced by helminth-derived peptide in vivo. FINDINGS Here, based on target-driven discovery strategy, we successfully identified a small 3 kDa peptide (SjDX5-53) from egg extracts of schistosome, which promoted both human and murine Tregs production. SjDX5-53 presented immunosuppressive function by arresting dendritic cells (DCs) at an immature state and augmenting the proportion and suppressive capacity of Tregs. In mouse models, SjDX5-53 protected mice against autoimmune-related colitis and psoriasis through inducing Tregs and inhibiting inflammatory T-helper (Th) 1 and Th17 responses. INTERPRETATION SjDX5-53 exhibited the promising therapeutic effects in alleviating the phenotype of immune-related colitis and psoriasis. This study displayed a screening and validation pipeline of the inducer of Tregs from helminth eggs, highlighting the discovery of new biologics inspired by co-evolution of hosts and their parasites. FUNDING This study was supported by the Natural Science Foundation of China (82272368) and Natural Science Foundation of Jiangsu Province (BK20211586).
Collapse
Affiliation(s)
- Yangyue Ni
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Ruiyan Xiong
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yuxiao Zhu
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Ning Luan
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, China
| | - Chuanxin Yu
- Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Kun Yang
- Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Huiquan Wang
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Xuejun Xu
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yuxuan Yang
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Siyu Sun
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Liyun Shi
- Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, China
| | - Jon Rob Padde
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Lin Chen
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Lu Chen
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Min Hou
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Zhipeng Xu
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, China.
| | - Minjun Ji
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
8
|
Abou-El-Naga IF, Mogahed NMFH. Potential roles of Toxocara canis larval excretory secretory molecules in immunomodulation and immune evasion. Acta Trop 2023; 238:106784. [PMID: 36502886 DOI: 10.1016/j.actatropica.2022.106784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/15/2022] [Accepted: 11/27/2022] [Indexed: 12/13/2022]
Abstract
Toxocara canis larvae invade various tissues of different vertebrate species without developing into adults in paratenic host. The long-term survival of the larvae despite exposure to the well-armed immune response is a notable achievement. The larvae modulate the immune response to help the survival of both the host and the larvae. They skew the immune response to type 2/regulatory phenotype. The outstanding ability of the larvae to modulate the host immune response and to evade the immune arms is attributed to the secretion of Toxocara excretory-secretory products (TESPs). TESPs are complex mixture of differing molecules. The present review deals with the molecular composition of the TESPs, their interaction with the host molecules, their effect on the innate immune response, the receptor recognition, the downstream signals the adaptive immunity and the repair of tissues. This review also addresses the role of TESPs molecules in the immune evasion strategy and the potential effect of the induced immunomodulation in some diseases. Identification of parasite components that influence the nematode-host interactions could enhance understanding the molecular basis of nematode pathogenicity. Furthermore, the identification of helminths molecules with immunomodulatory potential could be used in immunotherapies for some diseases.
Collapse
Affiliation(s)
- Iman F Abou-El-Naga
- Medical Parasitology Department, Faculty of Medicine, Alexandria University, 12 Abdel Hamid El Deeb Street, Tharwat, Alexandria, Egypt.
| | - Nermine M F H Mogahed
- Medical Parasitology Department, Faculty of Medicine, Alexandria University, 12 Abdel Hamid El Deeb Street, Tharwat, Alexandria, Egypt
| |
Collapse
|
9
|
Puricelli C, Boggio E, Gigliotti CL, Stoppa I, Sutti S, Rolla R, Dianzani U. Cutting-Edge Delivery Systems and Adjuvants in Tolerogenic Vaccines: A Review. Pharmaceutics 2022; 14:1782. [PMID: 36145531 PMCID: PMC9501480 DOI: 10.3390/pharmaceutics14091782] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Conventional therapies for immune-mediated diseases, including autoimmune disorders, transplant reactions, and allergies, have undergone a radical evolution in the last few decades; however, they are still not specific enough to avoid widespread immunosuppression. The idea that vaccine usage could be extended beyond its traditional immunogenic function by encompassing the ability of vaccines to induce antigen-specific tolerance may revolutionize preventive and therapeutic strategies in several clinical fields that deal with immune-mediated disorders. This approach has been supported by improved data relating to the several mechanisms involved in controlling unwanted immune responses and allowing peripheral tolerance. Given these premises, several approaches have been developed to induce peripheral tolerance against the antigens that are involved in the pathological immune response, including allergens, autoantigens, and alloantigens. Technological innovations, such as nucleic acid manipulation and the advent of micro- and nanoparticles, have further supported these novel preventive and therapeutic approaches. This review focuses on the main strategies used in the development of tolerogenic vaccines, including the technological issues used in their design and the role of "inverse adjuvants". Even though most studies are still limited to the preclinical field, the enthusiasm generated by their results has prompted some initial clinical trials, and they show great promise for the future management of immune-mediated pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | | | - Roberta Rolla
- Department of Health Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy
| | | |
Collapse
|
10
|
Bruschi F, Ashour D, Othman A. Trichinella-induced immunomodulation: Another tale of helminth success. Food Waterborne Parasitol 2022; 27:e00164. [PMID: 35615625 PMCID: PMC9125654 DOI: 10.1016/j.fawpar.2022.e00164] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 01/01/2023] Open
Abstract
Trichinella spiralis is a unique parasite in that both the adults and larvae survive in two different intracellular niches in the same host. The immune response, albeit intense, is highly modulated to ensure the survival of both the host and the parasite. It is skewed to T helper 2 and regulatory arms. Diverse cells from both the innate and adaptive compartments of immunity, including dendritic cells, T regulatory cells, and alternatively activated macrophages are thought to mediate such immunomodulation. The parasite has also an outstanding ability to evade the immune system by several elaborate processes. The molecules derived from the parasites including Trichinella, particularly the components of the excretory-secretory products, are being continually identified and explored for the potential of ameliorating the immunopathology in animal models of diverse inflammatory and autoimmune human diseases. Herein we discuss the various aspects of Trichinella-induced immunomodulation with a special reference to the practical implications of the immune system manipulation in alleviating or possibly curing human diseases.
Collapse
Key Words
- AAM, alternatively activated macrophage
- AW, adult worm
- Allergy
- Autoimmune diseases
- Breg, regulatory B cell
- CAM, classically activated macrophage
- Cancer
- ES L1, ES product of T. spiralis muscle larva
- ES, excretory–secretory
- IFN- γ, interferon-γ
- IIL, intestinal infective larva
- IL, interleukin
- Immune evasion
- Immunomodulation
- ML, muscle larva
- NBL, newborn larva
- NOS, nitric oxide synthase
- TGF-β, transforming growth factor-β
- TLR, toll-like receptor
- TNF- α, tumor necrosis factor-α
- Th, T helper
- Tol-DC, tolerogenic dendritic cell
- Treg, regulatory T cell
- Trichinella
- Trichinella-derived molecules
- Ts-AES, ES from adult T. spiralis
Collapse
Affiliation(s)
- F. Bruschi
- School of Medicine, Department of Translational Research, N.T.M.S., Università di Pisa, Pisa, Italy
| | - D.S. Ashour
- Department of Medical Parasitology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - A.A. Othman
- Department of Medical Parasitology, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
11
|
Yang J, Kuang H, Li N, Hamdy AM, Song J. The modulation and mechanism of probiotic-derived polysaccharide capsules on the immune response in allergic diseases. Crit Rev Food Sci Nutr 2022; 63:8768-8780. [PMID: 35400262 DOI: 10.1080/10408398.2022.2062294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Allergic diseases, derived from the dysregulation of immune tolerance mechanisms, have been rising in the last two decades. Recently, increasing evidence has shown that probiotic-derived polysaccharide capsules exhibit a protective effect against allergic diseases, involving regulation of Th1/Th2 balance, induction of differentiation of T regulatory cells and activation of dendritic cells (DCs). DCs have a central role in controlling the immune response through their interaction with gut microbiota via their pattern recognition receptors, including Toll-like receptors and C-type-lectin receptors. This review discusses the effects and critical mechanism of probiotic-derived polysaccharide capsules in regulating the immune system to alleviate allergic diseases. We first describe the development of immune response in allergic diseases and recent relevant findings. Particular emphasis is placed on the effects of probiotic-derived polysaccharide capsules on allergic immune response. Then, we discuss the underlying mechanism of the impact of probiotic-derived polysaccharide capsules on DCs-mediated immune tolerance induction.
Collapse
Affiliation(s)
- Jing Yang
- Chongqing Engineering Research Center for Processing & Storage of Distinct Agricultural Products, Chongqing Technology and Business University, Chongqing, China
| | - Hong Kuang
- Chongqing Engineering Research Center for Processing & Storage of Distinct Agricultural Products, Chongqing Technology and Business University, Chongqing, China
| | - Ning Li
- Chongqing Engineering Research Center for Processing & Storage of Distinct Agricultural Products, Chongqing Technology and Business University, Chongqing, China
| | - Ahmed Mahmoud Hamdy
- Dairy Science Department, Faculty of Agriculture, Assiut University, Assiut, Egypt
| | - Jiajia Song
- College of Food Science, Southwest University, Chongqing, China
| |
Collapse
|
12
|
Shi W, Xu N, Wang X, Vallée I, Liu M, Liu X. Helminth Therapy for Immune-Mediated Inflammatory Diseases: Current and Future Perspectives. J Inflamm Res 2022; 15:475-491. [PMID: 35087284 PMCID: PMC8789313 DOI: 10.2147/jir.s348079] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/11/2022] [Indexed: 12/17/2022] Open
Affiliation(s)
- Wenjie Shi
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, People’s Republic of China
| | - Ning Xu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, People’s Republic of China
| | - Xuelin Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, People’s Republic of China
| | - Isabelle Vallée
- UMR BIPAR, Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, Laboratoire de Santé Animale, Maisons-Alfort, France
| | - Mingyuan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, People’s Republic of China
| | - Xiaolei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, People’s Republic of China
- Correspondence: Xiaolei Liu; Mingyuan Liu, Tel +86-15943092280; +86-13019125996, Email ;
| |
Collapse
|
13
|
Willment JA. Fc-conjugated C-type lectin receptors: Tools for understanding host-pathogen interactions. Mol Microbiol 2021; 117:632-660. [PMID: 34709692 DOI: 10.1111/mmi.14837] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022]
Abstract
The use of soluble fusion proteins of pattern recognition receptors (PRRs) used in the detection of exogenous and endogenous ligands has helped resolve the roles of PRRs in the innate immune response to pathogens, how they shape the adaptive immune response, and function in maintaining homeostasis. Using the immunoglobulin (Ig) crystallizable fragment (Fc) domain as a fusion partner, the PRR fusion proteins are soluble, stable, easily purified, have increased affinity due to the Fc homodimerization properties, and consequently have been used in a wide range of applications such as flow cytometry, screening of protein and glycan arrays, and immunofluorescent microscopy. This review will predominantly focus on the recognition of pathogens by the cell membrane-expressed glycan-binding proteins of the C-type lectin receptor (CLR) subgroup of PRRs. PRRs bind to conserved pathogen-associated molecular patterns (PAMPs), such as glycans, usually located within or on the outer surface of the pathogen. Significantly, many glycans structures are identical on both host and pathogen (e.g. the Lewis (Le) X glycan), allowing the use of Fc CLR fusion proteins with known endogenous and/or exogenous ligands as tools to identify pathogen structures that are able to interact with the immune system. Screens of highly purified pathogen-derived cell wall components have enabled identification of many unique PAMP structures recognized by CLRs. This review highlights studies using Fc CLR fusion proteins, with emphasis on the PAMPs found in fungi, bacteria, viruses, and parasites. The structure and unique features of the different CLR families is presented using examples from a broad range of microbes whenever possible.
Collapse
Affiliation(s)
- Janet A Willment
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| |
Collapse
|
14
|
Novel probiotic yeast from Miso promotes regulatory dendritic cell IL-10 production and attenuates DSS-induced colitis in mice. J Gastroenterol 2021; 56:829-842. [PMID: 34213612 DOI: 10.1007/s00535-021-01804-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/19/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Yeasts are a type of fungi thought to have probiotic functions. In this study, we isolated a novel probiotic yeast (Zygosaccharomyces sapae strain I-6) from Miso (a traditional Japanese fermented food). We examined its effects on phenotypic changes in intestinal dendritic cells (DCs), and evaluated its anti-inflammatory effects in dextran sulfate sodium (DSS)-induced colitis. METHODS A single colony was selected from homogenized Miso, based on its ability to produce interleukin (IL)-10 in CD11c+ bone marrow DCs (BMDCs) in vitro. The anti-inflammatory effects of strain I-6 on CD11c+ BMDCs and CD11c+ CD103+ DCs were analyzed in mouse mesenteric lymph nodes in vitro and in a DSS mouse model. RESULTS The IL-10 concentrations in the co-culture BMDC supernatants treated with I-6 were dramatically higher than in those treated with Saccharomyces cerevisiae (Sc). IL-10 production is mediated by both TLR2 and Dectin-1. β-Glucan extracted from I-6 also induced higher levels of IL-10 production in BMDCs than β-glucan from Sc. The number of mesenteric lymph node CD11c+ CD103+ DCs was significantly increased by I-6 administration, compared with Sc administration. Strain I-6 showed strong anti-inflammatory effects on DSS-induced colitis compared to Sc. Moreover, the adoptive transfer of I-6-treated BMDCs showed anti-inflammatory effects on DSS-induced colitis in mice without oral administration of I-6 cells. CONCLUSIONS Strain I-6 induced phenotypic changes in intestinal CD11c+ DCs characterized by high IL-10 production and exerted strong anti-inflammatory effects on DSS-induced colitis. Traditional Japanese fermented foods may be a valuable source of probiotic yeasts for effective IBD therapy and treatment.
Collapse
|
15
|
Ilić N, Kosanović M, Gruden-Movsesijan A, Glamočlija S, Sofronić-Milosavljević L, Čolić M, Tomić S. Harnessing immunomodulatory mechanisms of Trichinella spiralis to design novel nanomedical approaches for restoring self-tolerance in autoimmunity. Immunol Lett 2021; 238:57-67. [PMID: 34363897 DOI: 10.1016/j.imlet.2021.04.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/28/2021] [Accepted: 04/28/2021] [Indexed: 01/13/2023]
Abstract
The rapid increase in the prevalence of autoimmune diseases in recent decades, especially in developed countries, coincided with improved living conditions and healthcare. Part of this increase could be ascribed to the lack of exposure to infectious agents like helminths that co-evolved with us and display potent immune regulatory actions. In this review we discussed many investigations, including our own, showing that Trichinella spiralis via its excretory-secretory products attenuate Th1/Th17 immunopathological response in autoimmunity and potentiate the protective Th2 and or regulatory T cell response, acting as an effective induction of tolerogenic dendritic cells (DCs), and probably mimicking the autoantigen in some diseases. A recent discovery of T. spiralis extracellular vesicles (TsEVs) suggested that inducing a complex regulation of the immune response requires simultaneous delivery of different signals in nano-sized packages. Indeed, different artificial nanomedical approaches discussed here suggested that co-delivery of multiple signals via nanoparticles is the most promising strategy for the treatment of autoimmune diseases. Although a long way is ahead of us before we could completely replicate natural nano-delivery systems which are both safe and potent in restoring self-tolerance, a clear path is being opened from a careful examination of parasite-host interactions.
Collapse
Affiliation(s)
- Nataša Ilić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University in Belgrade, Serbia
| | - Maja Kosanović
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University in Belgrade, Serbia
| | - Alisa Gruden-Movsesijan
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University in Belgrade, Serbia
| | - Sofija Glamočlija
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University in Belgrade, Serbia
| | - Ljiljana Sofronić-Milosavljević
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University in Belgrade, Serbia
| | - Miodrag Čolić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University in Belgrade, Serbia; Medical Faculty Foča, University of East Sarajevo, Bosnia and Hercegovina; Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Sergej Tomić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University in Belgrade, Serbia.
| |
Collapse
|
16
|
Keumatio Doungstop BC, van Vliet SJ, van Ree R, de Jong EC, van Kooyk Y. Carbohydrates in allergy: from disease to novel immunotherapies. Trends Immunol 2021; 42:635-648. [PMID: 34052120 DOI: 10.1016/j.it.2021.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/01/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022]
Abstract
Respiratory allergic disorders are a global public health problem that are responsible for substantial morbidity and healthcare expenditure. Despite the availability of allergen immunotherapy (AIT), its efficacy is suboptimal and regimens are lengthy, with a significant risk of potentially severe side effects. Studies on the recognition of allergens by immune cells through carbohydrate-lectin interactions, which play a crucial role in immune modulation and pathogenesis of allergy, have paved the way for improvements in AIT. We highlight innovative approaches for more effective and safer AIT, including the use of allergens conjugated to specific carbohydrates that bind to C-type lectins (CLRs) and sialic acid-binding immunoglobulin-type lectins (Siglecs) on immune cells to induce suppressive responses.
Collapse
Affiliation(s)
- B C Keumatio Doungstop
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center (UMC), location Vrije Universiteit Medical Center (VUmc), Amsterdam, The Netherlands; Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - S J van Vliet
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center (UMC), location Vrije Universiteit Medical Center (VUmc), Amsterdam, The Netherlands; Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - R van Ree
- Department of Experimental Immunology, Amsterdam UMC, location Academic Medical Center (AMC), Amsterdam, The Netherlands; Department of Otorhinolaryngology, Amsterdam UMC, location AMC, Amsterdam, The Netherlands; Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - E C de Jong
- Department of Experimental Immunology, Amsterdam UMC, location Academic Medical Center (AMC), Amsterdam, The Netherlands; Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - Y van Kooyk
- Department of Experimental Immunology, Amsterdam UMC, location Academic Medical Center (AMC), Amsterdam, The Netherlands; Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands.
| |
Collapse
|
17
|
Castenmiller C, Keumatio-Doungtsop BC, van Ree R, de Jong EC, van Kooyk Y. Tolerogenic Immunotherapy: Targeting DC Surface Receptors to Induce Antigen-Specific Tolerance. Front Immunol 2021; 12:643240. [PMID: 33679806 PMCID: PMC7933040 DOI: 10.3389/fimmu.2021.643240] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/02/2021] [Indexed: 12/11/2022] Open
Abstract
Dendritic cells (DCs) are well-established as major players in the regulation of immune responses. They either induce inflammatory or tolerogenic responses, depending on the DC-subtype and stimuli they receive from the local environment. This dual capacity of DCs has raised therapeutic interest for their use to modify immune-activation via the generation of tolerogenic DCs (tolDCs). Several compounds such as vitamin D3, retinoic acid, dexamethasone, or IL-10 and TGF-β have shown potency in the induction of tolDCs. However, an increasing interest exists in defining tolerance inducing receptors on DCs for new targeting strategies aimed to develop tolerance inducing immunotherapies, on which we focus particular in this review. Ligation of specific cell surface molecules on DCs can result in antigen presentation to T cells in the presence of inhibitory costimulatory molecules and tolerogenic cytokines, giving rise to regulatory T cells. The combination of factors such as antigen structure and conformation, delivery method, and receptor specificity is of paramount importance. During the last decades, research provided many tools that can specifically target various receptors on DCs to induce a tolerogenic phenotype. Based on advances in the knowledge of pathogen recognition receptor expression profiles in human DC subsets, the most promising cell surface receptors that are currently being explored as possible targets for the induction of tolerance in DCs will be discussed. We also review the different strategies that are being tested to target DC receptors such as antigen-carbohydrate conjugates, antibody-antigen fusion proteins and antigen-adjuvant conjugates.
Collapse
Affiliation(s)
- Charlotte Castenmiller
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection & Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Brigitte-Carole Keumatio-Doungtsop
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection & Immunity, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ronald van Ree
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection & Immunity, University of Amsterdam, Amsterdam, Netherlands.,Department of Otorhinolaryngology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Esther C de Jong
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection & Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection & Immunity, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|