1
|
Liu Y, Chen Z, Cheng H, Zheng R, Huang W. Mucosal immunotherapy targeting APC in lung disease. J Inflamm (Lond) 2025; 22:15. [PMID: 40229816 PMCID: PMC11998460 DOI: 10.1186/s12950-025-00432-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 01/28/2025] [Indexed: 04/16/2025] Open
Abstract
Several studies have demonstrated that the pulmonary immune response is primarily facilitated by antigen-presenting cells (APCs), and that both professional and non-professional APCs contribute to overall pulmonary immunity. APCs play unique roles and mechanisms in pathogen elimination and immunomodulation. Mucosal immunity exhibits potential advantages over traditional parenteral immunity in that it stimulates immune defenses in mucosal and systemic tissues, which is important for reducing the burden of lung disease. However, obtaining a comprehensive understanding of the crosstalk between mucosal immunity and APC in the context of various lung diseases remains challenging. This mini-review aimed to elucidate the mechanisms of novel mucosal immunity, targeting APC action during lung infections, allergies, and malignant tumorigenesis. This minreview provides important insights into more effective therapeutic approaches for various lung diseases.
Collapse
Affiliation(s)
- Yangqi Liu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zijian Chen
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Hanchang Cheng
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Runzhi Zheng
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Weizhe Huang
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China.
| |
Collapse
|
2
|
Weitzman ER, Alegria M, Caplan A, Dowling D, Evans J, Fisher CE, Jordan A, Kossowsky J, Landau M, Larson H, Levy O, Levy S, Mnookin S, Reif S, Ross J, Sherman AC. Social complexity of a fentanyl vaccine to prevent opioid overdose conference proceedings: Radcliffe Institute for Advanced Study conference proceedings. Vaccine 2025; 44:126324. [PMID: 39317618 DOI: 10.1016/j.vaccine.2024.126324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/04/2024] [Indexed: 09/26/2024]
Abstract
Despite significant public health attention and investment, hundreds of thousands of individuals have suffered fatal opioid overdose since the onset of the opioid crisis. Risk of opioid overdose has been exacerbated by the influx of fentanyl, a powerful synthetic opioid, into the drug supply. The National Institutes of Health Helping End Addiction Long-term (HEAL) Initiative is supporting the development of vaccines targeting fentanyl to protect against overdose. If successful, a vaccine would induce anti-fentanyl antibodies to sequester fentanyl (but not other opioids) in the blood, preventing fentanyl from crossing into the brain and reaching the central nervous system where it can cause overdose. Introduction of an overdose preventing strategy that relies on a vaccine to confer passive protection may be impactful. However, vaccines are poorly understood by the public and politicized. Moreover, the overdose ecosystem is complex and extends across numerous social, economic, medical, and cultural systems. As such, optimal use of a vaccine strategy to address overdose may benefit from multidisciplinary consideration of the social, ethical, and systemic factors that influence substance use and overdose that may also impact the acceptability of a fentanyl vaccine and related implementation strategies. In March 2022, Dr. Elissa Weitzman convened a two-day conference at the Harvard Radcliffe Institute for Advanced Study on the Social Complexity of a Fentanyl Vaccine to Prevent Opioid Overdose. In all, 19 professionals from diverse disciplines (medicine, psychology, history, ethics, immunology, vaccinology, communications, policy) attended the conference and led discussions that centered on population health and epidemiology, history of medicine and frameworks for understanding substance use, ethics, decision-making and attitudes, and operational issues to the question of a novel immunotherapy targeting fentanyl overdose. Participants also debated the risks and benefits of vaccine administration in response to fictional clinical case vignettes. A summary of the conference presentations and discussions follows.
Collapse
Affiliation(s)
- Elissa R Weitzman
- Division of Addiction Medicine, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, United States; Division of Adolescent and Young Adult Medicine, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, United States; Computational Health Informatics Program, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, United States; Department of Pediatrics, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, United States.
| | - Margarita Alegria
- Department of Medicine, Massachusetts General Hospital, 50 Staniford St, Boston, MA 02114, United States; Department of Medicine, Harvard Medical School, 25 Shattuck St, Boston, MA, United States; Department of Psychiatry, Harvard Medical School, 25 Shattuck St, Boston, MA, United States
| | - Arthur Caplan
- New York University Grossman School of Medicine, 550 1(st) Ave, New York, NY 10016, United States
| | - David Dowling
- Precision Vaccines Program, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, United States; Department of Pediatrics, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, United States
| | - Jay Evans
- Inimmune Corporation, 1121 E Broadway St, Missoula, MT 59802, United States
| | - Carl Erik Fisher
- Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, New York 10032, United States
| | - Ayana Jordan
- New York University Grossman School of Medicine, 550 1(st) Ave, New York, NY 10016, United States
| | - Joe Kossowsky
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, United States; Department of Anesthesia, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, United States
| | | | - Heidi Larson
- Department of Infectious Disease Epidemiology and Dynamics, London School of Hygiene and Tropical Medicine, Keppel St, London WC1E 7HT, UK
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, United States; The Broad Institute, 415 Main St, Cambridge, MA 02142, United States; Department of Pediatrics, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, United States
| | - Sharon Levy
- Division of Addiction Medicine, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, United States; Department of Pediatrics, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, United States
| | - Seth Mnookin
- School of Humanities, Arts, and Social Sciences, Massachusetts Institute of Technology, 160 Memorial Dr, Cambridge, MA 02139, United States
| | - Sharon Reif
- Heller School for Social Policy and Management, Brandeis University, 415 South St, Waltham, MA 02453, United States
| | - Jennifer Ross
- Division of Addiction Medicine, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, United States; Department of Pediatrics, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, United States
| | - Amy Caryn Sherman
- Precision Vaccines Program, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, United States; Division of Infectious Disease, Brigham and Women's Hospital, 15 Francis St., Boston, MA 02115, United States; Department of Pediatrics, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, United States
| |
Collapse
|
3
|
Zhang W, Park HB, Yadav D, An EK, Kim SJ, Ryu D, Agrawal R, Ryu JH, Kwak M, Lee PCW, Jin JO. P-type pilus PapG protein elicits toll-like receptor 2-mediated immune activation during cancer immunotherapy. Int J Biol Macromol 2024; 282:137061. [PMID: 39481736 DOI: 10.1016/j.ijbiomac.2024.137061] [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: 06/12/2024] [Revised: 10/14/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
The immune activation ability of FimH, an adhesion protein in pili of Escherichia coli (E. coli), has been recently reported. However, studies on the immune activity of PapG, another major pili terminal protein, have not been well explored. In this study, the immune stimulatory effect of purified recombinant PapG was evaluated. PapG treatment promoted dramatic changes in dendritic morphology of the bone marrow-derived dendritic cells (BMDCs) and induced upregulation of co-stimulatory molecule levels, major histocompatibility complex (MHC) I and II expression, and pro-inflammatory cytokine production in BMDCs. To identify the stimulatory receptor of PapG, an in silico study was performed. PapG exhibited strong binding affinity with murine toll-like receptor 2 (TLR2). In addition, PapG-induced activation of splenic DC and its subsets was unsuccessful in TLR2-knock out mice. Combination of PapG and ovalbumin (OVA) elicited OVA-specific T cell proliferation and cytokine production and cytotoxicity that consequently promoted anti-cancer immune responses against OVA-expressing B16 melanoma. Furthermore, PapG treatment induced activation of peripheral blood DCs and its subsets in humans in a TLR2 dependent manner. PapG-stimulated human conventional DC2 promoted syngeneic T cell proliferation and activation. The findings of this study demonstrated that PapG could be a useful immune stimulator for immunotherapy against cancer.
Collapse
Affiliation(s)
- Wei Zhang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Hae-Bin Park
- Department of Microbiology, Brain Korea 21 project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Dhananjay Yadav
- Department of Life Science, Yeungnam University, Gyeongsan 38541, South Korea; USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, Florida 33613, USA
| | - Eun-Koung An
- Department of Microbiology, Brain Korea 21 project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - So-Jung Kim
- Department of Microbiology, Brain Korea 21 project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Dayoung Ryu
- Department of Microbiology, Brain Korea 21 project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Richa Agrawal
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, The University of Chicago, 929 E 57(th) street, Chicago, IL 60637, USA
| | - Ja-Hyoung Ryu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, South Korea
| | - Minseok Kwak
- Department of Chemistry, Pukyong National University, Busan 48513, South Korea
| | - Peter C W Lee
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea.
| | - Jun-O Jin
- Department of Microbiology, Brain Korea 21 project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea.
| |
Collapse
|
4
|
Shi C, Tang Z, Jin Z, Huang S, Xu X, Qu C, Lin TH. Characterization of DmToll and DmToll7 homologue in Litopenaeus vannamei based on structure analysis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 158:105209. [PMID: 38838948 DOI: 10.1016/j.dci.2024.105209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 06/07/2024]
Abstract
Toll-like receptors (TLRs) are a family of pattern recognition receptors (PRRs) that recognize invading pathogens and activate downstream signaling pathways. The number of 10 Tolls is found in Litopenaeus vannamei but have not yet been identified as the corresponding Toll homologue of model animal. In this study, we predicted the three-dimensional (3D) structures of 10 LvTolls (LvToll1-10) with AlphaFold2 program. The per-residue local distance difference test (pLDDT) scores of LvTolls showed the predicted structure of LvTolls had high accuracy (pLDDT>70). By structural analysis, 3D structures of LvToll2 and LvToll3 had high similarity with Drosophila melanogaster Toll and Toll7, respectively. 3D structure of LvToll7 and LvToll10 were not similar to that of other LvTolls. Moreover, we also predicted that LvSpätzle4 had high structural similarity to DmSpätzle. There were 9 potential hydrogen bonds in LvToll2-LvSpätzle4 complex. Importantly, co-immunoprecipitation assay showed that LvToll2 could bind with LvSpätzle4. Collectively, this study provides new insight for researching invertebrate immunity by identifying the protein of model animal homologue.
Collapse
Affiliation(s)
- Chenchen Shi
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zhuyun Tang
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China; National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Zhixin Jin
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Shan Huang
- Fujian Provincial Key Laboratory of Functional and Clinical Translational Medicine, Xiamen Medical College, Xiamen, Fujian, 361023, China; Department of Basic Medical Science, Xiamen Medical College, Xiamen, Fujian, 361023, China
| | - Xiuyue Xu
- Fujian Provincial Key Laboratory of Functional and Clinical Translational Medicine, Xiamen Medical College, Xiamen, Fujian, 361023, China; Department of Clinical Medicine, Xiamen Medical College, Xiamen, Fujian, 361023, China
| | - Chen Qu
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
| | - Ta-Hui Lin
- Fujian Provincial Key Laboratory of Functional and Clinical Translational Medicine, Xiamen Medical College, Xiamen, Fujian, 361023, China; State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian, 361102, China; Department of Basic Medical Science, Xiamen Medical College, Xiamen, Fujian, 361023, China.
| |
Collapse
|
5
|
Xia X, Zhang JW, Zhao B, Zhang M, Chen ZR, Zhang BF, Ji YL, Wang X, Xiong WM, Li JW, Lv QL. Progress of engineered bacteria for tumour therapy. Int Immunopharmacol 2024; 132:111935. [PMID: 38599096 DOI: 10.1016/j.intimp.2024.111935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/14/2024] [Accepted: 03/24/2024] [Indexed: 04/12/2024]
Abstract
Finding novel therapeutic modalities, improving drug delivery efficiency and targeting, and reducing the immune escape of tumor cells are currently hot topics in the field of tumor therapy. Bacterial therapeutics have proven highly effective in preventing tumor spread and recurrence, used alone or in combination with traditional therapies. In recent years, a growing number of researchers have significantly improved the targeting and penetration of bacteria by using genetic engineering technology, which has received widespread attention in the field of tumor therapy. In this paper, we provide an overview and assessment of the advancements made in the field of tumor therapy using genetically engineered bacteria. We cover three major aspects: the development of engineered bacteria, their integration with other therapeutic techniques, and the current state of clinical trials. Lastly, we discuss the limitations and challenges that are currently being faced in the utilization of engineered bacteria for tumor therapy.
Collapse
Affiliation(s)
- Xue Xia
- Jiangxi Key Laboratory of Translational Cancer Research, NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi 330029, PR China; College of Chemistry and Bio-engineering, Yichun University, Yichun 336000, PR China
| | - Jing-Wen Zhang
- Jiangxi Key Laboratory of Translational Cancer Research, NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi 330029, PR China; College of Chemistry and Bio-engineering, Yichun University, Yichun 336000, PR China
| | - Bing Zhao
- Jiangxi Key Laboratory of Translational Cancer Research, NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi 330029, PR China; College of Chemistry and Bio-engineering, Yichun University, Yichun 336000, PR China
| | - Min Zhang
- Nanchang Inspection and Testing Center, Nanchang Key Laboratory for Quality and Safety Risk Assessment of Health Food and its Contact Materials, Nanchang 330012, PR China
| | - Zhang-Ren Chen
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330000, PR China
| | - Bing-Feng Zhang
- College of Chemistry and Bio-engineering, Yichun University, Yichun 336000, PR China
| | - Yu-Long Ji
- Jiangxi Key Laboratory of Translational Cancer Research, NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi 330029, PR China
| | - Xia Wang
- Jiangxi Key Laboratory of Translational Cancer Research, NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi 330029, PR China
| | - Wen-Min Xiong
- Jiangxi Key Laboratory of Translational Cancer Research, NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi 330029, PR China
| | - Jia-Wei Li
- Department of Cardiovascular, The First Affiliated Hospital of Nanchang University, Jiangxi, PR China.
| | - Qiao-Li Lv
- Jiangxi Key Laboratory of Translational Cancer Research, NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital, Jiangxi Clinical Research Center for Cancer, Nanchang, Jiangxi 330029, PR China; College of Chemistry and Bio-engineering, Yichun University, Yichun 336000, PR China.
| |
Collapse
|
6
|
DaMata JP, Zelkoski AE, Nhan PB, Ennis KHE, Kim JS, Lu Z, Malloy AMW. Dissociation protocols influence the phenotypes of lymphocyte and myeloid cell populations isolated from the neonatal lymph node. Front Immunol 2024; 15:1368118. [PMID: 38756770 PMCID: PMC11097666 DOI: 10.3389/fimmu.2024.1368118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
Frequencies and phenotypes of immune cells differ between neonates and adults in association with age-specific immune responses. Lymph nodes (LN) are critical tissue sites to quantify and define these differences. Advances in flow cytometry have enabled more multifaceted measurements of complex immune responses. Tissue processing can affect the immune cells under investigation that influence key findings. To understand the impact on immune cells in the LN after processing for single-cell suspension, we compared three dissociation protocols: enzymatic digestion, mechanical dissociation with DNase I treatment, and mechanical dissociation with density gradient separation. We analyzed cell yields, viability, phenotypic and maturation markers of immune cells from the lung-draining LN of neonatal and adult mice two days after intranasal respiratory syncytial virus (RSV) infection. While viability was consistent across age groups, the protocols influenced the yield of subsets defined by important phenotypic and activation markers. Moreover, enzymatic digestion did not show higher overall yields of conventional dendritic cells and macrophages from the LN. Together, our findings show that the three dissociation protocols have similar impacts on the number and viability of cells isolated from the neonatal and adult LN. However, enzymatic digestion impacts the mean fluorescence intensity of key lineage and activation markers that may influence experimental findings.
Collapse
Affiliation(s)
- Jarina P. DaMata
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Amanda E. Zelkoski
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
| | - Paula B. Nhan
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Katherine H. E. Ennis
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Ji Sung Kim
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Zhongyan Lu
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Allison M. W. Malloy
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
| |
Collapse
|
7
|
Zhang T, Aipire A, Li Y, Guo C, Li J. Antigen cross-presentation in dendric cells: From bench to bedside. Biomed Pharmacother 2023; 168:115758. [PMID: 37866002 DOI: 10.1016/j.biopha.2023.115758] [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: 08/30/2023] [Revised: 10/14/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023] Open
Abstract
Cross-presentation (XPT) is an adaptation of the cellular process in which dendritic cells (DCs) present exogenous antigens on major histocompatibility complex (MHC) class I molecules for recognition of the cytotoxic T lymphocytes (CTL) and natural killer (NK) cells, resulting in immunity or tolerance. Recent advances in DCs have broadened our understanding of the underlying mechanisms of XPT and strengthened their application in tumor immunotherapy. In this review, we summarized the known mechanisms of XPT, including the receptor-mediated internalization of exogenous antigens, endosome escape, engagement of the other XPT-related proteins, and adjuvants, which significantly enhance the XPT capacity of DCs. Consequently, various strategies to enhance XPT can be adopted and optimized to improve outcomes of DC-based therapy.
Collapse
Affiliation(s)
- Tingting Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Adila Aipire
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Yijie Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Changying Guo
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China.
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China.
| |
Collapse
|
8
|
Zhang W, An EK, Kim SJ, Park HB, Lee PCW, Jin JO. Escherichia coli adhesion protein FimH exacerbates colitis via CD11b +CD103 - dendritic cell activation. Front Immunol 2023; 14:1284770. [PMID: 38077339 PMCID: PMC10703180 DOI: 10.3389/fimmu.2023.1284770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Introduction Immune stimulators are used to improve vaccine efficiency; however, they are accompanied by various side effects. In previous studies, we reported that the Escherichia coli adhesion protein, FimH, induces immune activity; however, we did not examine any side effects in colon inflammation. Methods FimH was administered orally or intraperitoneally (i.p.) to mice with dextran sulfate sodium (DSS)-induced colitis, and changes in symptoms were observed. Immune cells infiltrated into the colon after the induction of colon inflammation were analyzed using a flow cytometer. Changes in Th1 and Th17 cells that induce colitis were analyzed. Further, mesenteric lymph node (mLN) dendritic cells (DCs) activated by FimH were identified and isolated to examine their ability to induce T-cell immunity. Results FimH oral and i.p. administration in C57BL/6 mice did not induce inflammation in the colon; however, DSS-induced colitis was exacerbated by oral and i.p. FimH administration. FimH treatment increased immune cell infiltration in the colon compared to that in DSS colitis. Th1 and Th17 cells, which are directly related to colitis, were increased in the colon by FimH; however, FimH did not directly affect the differentiation of these T cells. FimH upregulated the CD11b+CD103- DC activity in the mLNs, which produced the signature cytokines required for Th1 and Th17. In addition, isolated CD11b+CD103- DCs, after stimulation with FimH, directly induced Th1 and Th17 differentiation in a co-culture of CD4 T cells. Conclusion This study demonstrated the side effects of FimH and indicated that the use of FimH can aggravate the disease in patients with colitis.
Collapse
Affiliation(s)
- Wei Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Eun-Koung An
- Department of Microbiology, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - So-Jung Kim
- Department of Microbiology, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hae-Bin Park
- Department of Microbiology, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Peter C. W. Lee
- Department of Biochemistry and Molecular Biology, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jun-O Jin
- Department of Microbiology, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
9
|
Li M, Chen C, Wang X, Guo P, Feng H, Zhang X, Zhang W, Gu C, Zhu J, Wen G, Feng Y, Xiao L, Peng G, Rao VB, Tao P. T4 bacteriophage nanoparticles engineered through CRISPR provide a versatile platform for rapid development of flu mucosal vaccines. Antiviral Res 2023; 217:105688. [PMID: 37516153 DOI: 10.1016/j.antiviral.2023.105688] [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: 05/04/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 07/31/2023]
Abstract
Vaccines that trigger mucosal immune responses at the entry portals of pathogens are highly desired. Here, we showed that antigen-decorated nanoparticle generated through CRISPR engineering of T4 bacteriophage can serve as a universal platform for the rapid development of mucosal vaccines. Insertion of Flu viral M2e into phage T4 genome through fusion to Soc (Small Outer Capsid protein) generated a recombinant phage, and the Soc-M2e proteins self-assembled onto phage capsids to form 3M2e-T4 nanoparticles during propagation of T4 in E. coli. Intranasal administration of 3M2e-T4 nanoparticles maintains antigen persistence in the lungs, resulting in increased uptake and presentation by antigen-presenting cells. M2e-specific secretory IgA, effector (TEM), central (TCM), and tissue-resident memory CD4+ T cells (TRM) were efficiently induced in the local mucosal sites, which mediated protections against divergent influenza viruses. Our studies demonstrated the mechanisms of immune protection following 3M2e-T4 nanoparticles vaccination and provide a versatile T4 platform that can be customized to rapidly develop mucosal vaccines against future emerging epidemics.
Collapse
Affiliation(s)
- Mengling Li
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Cen Chen
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Xialin Wang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Pengju Guo
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Helong Feng
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, Hubei, 430070, China
| | - Xueqi Zhang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Wanpo Zhang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Changqin Gu
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Jingen Zhu
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC, 20064, USA
| | - Guoyuan Wen
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, Hubei, 430070, China
| | - Yaoyu Feng
- Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Lihua Xiao
- Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Guiqing Peng
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Venigalla B Rao
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC, 20064, USA
| | - Pan Tao
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China.
| |
Collapse
|
10
|
Yang Q, Yang XD, Liu MQ, Zeng C, Zhao HK, Xiang KW, Hou ZS, Wen HS, Li JF. Transcriptome analysis of liver, gill and intestine in rainbow trout (Oncorhynchus mykiss) symptomatically or asymptomatically infected with Vibrio anguillarum. FISH & SHELLFISH IMMUNOLOGY 2023; 135:108643. [PMID: 36871630 DOI: 10.1016/j.fsi.2023.108643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/16/2023] [Accepted: 02/23/2023] [Indexed: 06/18/2023]
Abstract
Rainbow trout (Oncorhynchus mykiss), an important economic cold-water fish worldwide, is severely threatened by viruses and bacteria in the farming industry. The vibriosis outbreak has caused a significant setback to aquaculture. Vibrio anguillarum, one of the common disease-causing vibriosis associated with severe lethal vibriosis in aquaculture, infects fish mainly by adsorption and invasion of the skin, gills, lateral line and intestine. To investigate the defense mechanism of rainbow trout against the pathogen after infection with Vibrio anguillarum, trout were intraperitoneally injected by Vibrio anguillarum and divided into symptomatic group (SG) and asymptomatic group (AG) according to the phenotype. RNA-Seq technology was used to evaluate the transcriptional signatures of liver, gill and intestine of trout injected with Vibrio anguillarum (SG and AG) and corresponding control groups (CG(A) and CG(B)). The GO and KEGG enrichment analyses were used to investigate the mechanisms underlying the differences in susceptibility to Vibrio anguillarum. Results showed that in SG, immunomodulatory genes in the cytokine network were activated and tissue function-related genes were down-regulated, while apoptosis mechanisms were activated. However, AG responded to Vibrio anguillarum infection by activating complement related immune defenses, while metabolism and function related genes were up-regulated. Conclusively, a rapid and effective immune and inflammatory response can successfully defend Vibrio anguillarum infection. However, a sustained inflammatory response can lead to tissue and organ damage and cause death. Our results may provide a theoretical basis for breeding rainbow trout for disease resistance.
Collapse
Affiliation(s)
- Qian Yang
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education (KLMME), Ocean University of China, Qingdao, China
| | - Xiao-Dong Yang
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education (KLMME), Ocean University of China, Qingdao, China
| | - Meng-Qun Liu
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education (KLMME), Ocean University of China, Qingdao, China
| | - Chu Zeng
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education (KLMME), Ocean University of China, Qingdao, China
| | - Hong-Kui Zhao
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education (KLMME), Ocean University of China, Qingdao, China
| | - Kai-Wen Xiang
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education (KLMME), Ocean University of China, Qingdao, China
| | - Zhi-Shuai Hou
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education (KLMME), Ocean University of China, Qingdao, China
| | - Hai-Shen Wen
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education (KLMME), Ocean University of China, Qingdao, China
| | - Ji-Fang Li
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education (KLMME), Ocean University of China, Qingdao, China.
| |
Collapse
|
11
|
Liu P, Kang C, Zhang J, Liu Y, Liu J, Hu T, Zeng X, Qiu S. The role of dendritic cells in allergic diseases. Int Immunopharmacol 2022; 113:109449. [DOI: 10.1016/j.intimp.2022.109449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/06/2022] [Accepted: 11/09/2022] [Indexed: 11/18/2022]
|
12
|
Díaz-Dinamarca DA, Salazar ML, Castillo BN, Manubens A, Vasquez AE, Salazar F, Becker MI. Protein-Based Adjuvants for Vaccines as Immunomodulators of the Innate and Adaptive Immune Response: Current Knowledge, Challenges, and Future Opportunities. Pharmaceutics 2022; 14:1671. [PMID: 36015297 PMCID: PMC9414397 DOI: 10.3390/pharmaceutics14081671] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/05/2022] [Accepted: 08/07/2022] [Indexed: 12/03/2022] Open
Abstract
New-generation vaccines, formulated with subunits or nucleic acids, are less immunogenic than classical vaccines formulated with live-attenuated or inactivated pathogens. This difference has led to an intensified search for additional potent vaccine adjuvants that meet safety and efficacy criteria and confer long-term protection. This review provides an overview of protein-based adjuvants (PBAs) obtained from different organisms, including bacteria, mollusks, plants, and humans. Notably, despite structural differences, all PBAs show significant immunostimulatory properties, eliciting B-cell- and T-cell-mediated immune responses to administered antigens, providing advantages over many currently adopted adjuvant approaches. Furthermore, PBAs are natural biocompatible and biodegradable substances that induce minimal reactogenicity and toxicity and interact with innate immune receptors, enhancing their endocytosis and modulating subsequent adaptive immune responses. We propose that PBAs can contribute to the development of vaccines against complex pathogens, including intracellular pathogens such as Mycobacterium tuberculosis, those with complex life cycles such as Plasmodium falciparum, those that induce host immune dysfunction such as HIV, those that target immunocompromised individuals such as fungi, those with a latent disease phase such as Herpes, those that are antigenically variable such as SARS-CoV-2 and those that undergo continuous evolution, to reduce the likelihood of outbreaks.
Collapse
Affiliation(s)
- Diego A. Díaz-Dinamarca
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7750000, Chile
| | - Michelle L. Salazar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
| | - Byron N. Castillo
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
| | - Augusto Manubens
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Biosonda Corporation, Santiago 7750000, Chile
| | - Abel E. Vasquez
- Sección de Biotecnología, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7750000, Chile
- Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Providencia, Santiago 8320000, Chile
| | - Fabián Salazar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter EX4 4QD, UK
| | - María Inés Becker
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago 7750000, Chile
- Biosonda Corporation, Santiago 7750000, Chile
| |
Collapse
|
13
|
Fan JY, Huang Y, Li Y, Muluh TA, Fu SZ, Wu JB. Bacteria in cancer therapy: A new generation of weapons. Cancer Med 2022; 11:4457-4468. [PMID: 35522104 PMCID: PMC9741989 DOI: 10.1002/cam4.4799] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 12/15/2022] Open
Abstract
Tumors are presently a major threat to human life and health. Malignant tumors are conventionally treated through radiotherapy and chemotherapy. However, traditional therapies yield unsatisfactory results due to high toxicity to the normal cells, inability to treat deep tumor tissues, and the possibility of inducing drug resistance in the tumor cells. This has caused immunotherapy to emerge as an effective and alternate treatment strategy. To overcome the limitations of the conventional treatments as well as to avert the risk of various drug resistance and cytotoxicity, bacterial anti-tumor immunotherapy has raised the interest of researchers. This therapeutic strategy employs bacteria to specifically target and colonize the tumor tissues with preferential accumulation and proliferation. Such bacterial accumulation initiates a series of anti-tumor immune responses, effectively eliminating the tumor cells. This immunotherapy can use the bacteria alone or concomitantly with the other methods. For example, the bacteria can deliver the anti-cancer effect mediators by regulating the expression of the bacterial genes or by synthesizing the bioengineered bacterial complexes. This review will discuss the mechanism of utilizing bacteria in treating tumors, especially in terms of immune mechanisms. This could help in better integrating the bacterial method with other treatment options, thereby, providing a more effective, reliable, and unique treatment therapy for tumors.
Collapse
Affiliation(s)
- Jun Ying Fan
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China
| | - Yuan Huang
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China
| | - Yi Li
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China
| | - Tobias Achu Muluh
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China
| | - Shao Zhi Fu
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China,Department of Nuclear MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China
| | - Jing Bo Wu
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China,Department of Nuclear MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China,Academician (Expert) Workstation of Sichuan ProvinceLuzhouSichuanP.R. China
| |
Collapse
|
14
|
Duan T, Du Y, Xing C, Wang HY, Wang RF. Toll-Like Receptor Signaling and Its Role in Cell-Mediated Immunity. Front Immunol 2022. [PMID: 35309296 DOI: 10.3389/fimmu.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Innate immunity is the first defense system against invading pathogens. Toll-like receptors (TLRs) are well-defined pattern recognition receptors responsible for pathogen recognition and induction of innate immune responses. Since their discovery, TLRs have revolutionized the field of immunology by filling the gap between the initial recognition of pathogens by innate immune cells and the activation of the adaptive immune response. TLRs critically link innate immunity to adaptive immunity by regulating the activation of antigen-presenting cells and key cytokines. Furthermore, recent studies also have shown that TLR signaling can directly regulate the T cell activation, growth, differentiation, development, and function under diverse physiological conditions. This review provides an overview of TLR signaling pathways and their regulators and discusses how TLR signaling, directly and indirectly, regulates cell-mediated immunity. In addition, we also discuss how TLR signaling is critically important in the host's defense against infectious diseases, autoimmune diseases, and cancer.
Collapse
Affiliation(s)
- Tianhao Duan
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Yang Du
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Changsheng Xing
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Helen Y Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Rong-Fu Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
15
|
Duan T, Du Y, Xing C, Wang HY, Wang RF. Toll-Like Receptor Signaling and Its Role in Cell-Mediated Immunity. Front Immunol 2022; 13:812774. [PMID: 35309296 PMCID: PMC8927970 DOI: 10.3389/fimmu.2022.812774] [Citation(s) in RCA: 378] [Impact Index Per Article: 126.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
Innate immunity is the first defense system against invading pathogens. Toll-like receptors (TLRs) are well-defined pattern recognition receptors responsible for pathogen recognition and induction of innate immune responses. Since their discovery, TLRs have revolutionized the field of immunology by filling the gap between the initial recognition of pathogens by innate immune cells and the activation of the adaptive immune response. TLRs critically link innate immunity to adaptive immunity by regulating the activation of antigen-presenting cells and key cytokines. Furthermore, recent studies also have shown that TLR signaling can directly regulate the T cell activation, growth, differentiation, development, and function under diverse physiological conditions. This review provides an overview of TLR signaling pathways and their regulators and discusses how TLR signaling, directly and indirectly, regulates cell-mediated immunity. In addition, we also discuss how TLR signaling is critically important in the host's defense against infectious diseases, autoimmune diseases, and cancer.
Collapse
Affiliation(s)
- Tianhao Duan
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Yang Du
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Changsheng Xing
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Helen Y. Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Rong-Fu Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
16
|
Brennan K, Craven S, Cheung M, Kane D, Noone E, O'Callaghan J, Molloy EJ, Walsh PT, McAuliffe FM, Doyle SL. Cytosolic dsRNA improves neonatal innate immune responses to adjuvants in use in pediatric vaccines. J Leukoc Biol 2022; 112:523-537. [PMID: 35098572 PMCID: PMC9542317 DOI: 10.1002/jlb.5a0521-242r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 11/17/2021] [Accepted: 01/06/2022] [Indexed: 11/12/2022] Open
Abstract
Pattern recognition receptors (PRRs) of the innate immune system represent the critical front‐line defense against pathogens, and new vaccine formulations target these PRR pathways to boost vaccine responses, through activation of cellular/Th1 immunity. The majority of pediatric vaccines contain aluminum (ALUM) or monophosphoryl lipid A (MPLA) as adjuvants to encourage immune activation. Evidence suggests that elements of the innate immune system, currently being targeted for vaccine adjuvanticity do not fully develop until puberty and it is likely that effective adjuvants for the neonatal and pediatric populations are being overlooked due to modeling of responses in adult systems. We recently reported that the activity of the cytosolic nucleic acid (CNA) sensing family of PRRs is strong in cord blood and peripheral blood of young children. This study investigates the function of CNA sensors in subsets of neonatal innate immune cells and shows that myeloid cells from cord blood can be activated to express T cell costimulatory markers, and also to produce Th1 promoting cytokines. CD80 and CD86 were consistently up‐regulated in response to cytosolic Poly(I:C) stimulation in all cell types examined and CNA activation also induced robust Type I IFN and low levels of TNFα in monocytes, monocyte‐derived macrophages, and monocyte‐derived dendritic cells. We have compared CNA activation to adjuvants currently in use (MPLA or ALUM), either alone or in combination and found that cytosolic Poly(I:C) in combination with MPLA or ALUM can improve expression of activation marker levels above those observed with either adjuvant alone. This may prove particularly promising in the context of improving the efficacy of existing ALUM‐ or MPLA‐containing vaccines, through activation of T cell‐mediated immunity.
Collapse
Affiliation(s)
- Kiva Brennan
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Crumlin Dublin Ireland
- Department of Clinical Medicine, School of Medicine Trinity College Dublin Dublin Ireland
| | - Simon Craven
- UCD Perinatal Research Centre, Obstetrics & Gynaecology School of Medicine, University College Dublin, National Maternity Hospital Dublin Ireland
| | - Maria Cheung
- UCD Perinatal Research Centre, Obstetrics & Gynaecology School of Medicine, University College Dublin, National Maternity Hospital Dublin Ireland
| | - Daniel Kane
- UCD Perinatal Research Centre, Obstetrics & Gynaecology School of Medicine, University College Dublin, National Maternity Hospital Dublin Ireland
| | - Eleanor Noone
- Department of Clinical Medicine, School of Medicine Trinity College Dublin Dublin Ireland
| | - Joseph O'Callaghan
- Department of Clinical Medicine, School of Medicine Trinity College Dublin Dublin Ireland
| | - Eleanor J Molloy
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Crumlin Dublin Ireland
- Department of Paediatrics School of Medicine, Trinity College Dublin Dublin Ireland
- Coombe Women and Infants University Hospital Dublin Ireland
| | - Patrick T Walsh
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Crumlin Dublin Ireland
- Department of Clinical Medicine, School of Medicine Trinity College Dublin Dublin Ireland
| | - Fionnuala M McAuliffe
- UCD Perinatal Research Centre, Obstetrics & Gynaecology School of Medicine, University College Dublin, National Maternity Hospital Dublin Ireland
| | - Sarah L Doyle
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Crumlin Dublin Ireland
- Department of Clinical Medicine, School of Medicine Trinity College Dublin Dublin Ireland
| |
Collapse
|
17
|
Gao Q, Yi S, Li Y, Luo J, Xing Q, Yang X, Zhao M, Min M, Wang Q, Wang Y, Ma L, Peng S. The Role of Flagellin B in Vibrio anguillarum-Induced Intestinal Immunity and Functional Domain Identification. Front Immunol 2021; 12:774233. [PMID: 34912344 PMCID: PMC8667730 DOI: 10.3389/fimmu.2021.774233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
Vibrio anguillarum, an opportunistic pathogen of aquatic animals, moves using a filament comprised of polymerised flagellin proteins. Flagellins are essential virulence factors for V. anguillarum infection. Herein, we investigated the effects of flagellins (flaA, flaB, flaC, flaD and flaE) on cell apoptosis, TLR5 expression, and production of IL-8 and TNF-α. FlaB exhibited the strongest immunostimulation effects. To explore the functions of flaB in infection, we constructed a flaB deletion mutant using a two-step recombination method, and in vitro experiments showed a significant decrease in the expression of TLR5 and inflammatory cytokines compared with wild-type cells. However in the in vivo study, expression of inflammatory cytokines and intestinal mucosal structure showed no significant differences between groups. Additionally, flaB induced a significant increase in TLR5 expression based on microscopy analysis of fluorescently labelled TLR5, indicating interactions between the two proteins, which was confirmed by native PAGE and yeast two-hybrid assay. Molecular simulation of interactions between flaB and TLR5 was performed to identify the residues involved in binding, revealing two binding sites. Then, based on molecular dynamics simulations, we carried out thirteen site-directed mutations occurring at the amino acid sites of Q57, N83, N87, R91, D94, E122, D152, N312, R313, N320, L97, H316, I324 in binding regions of flaB protein by TLR5, respectively. Surface plasmon resonance (SPR) was employed to compare the affinities of flaB mutants for TLR5, and D152, D94, I324, N87, R313, N320 and H316 were found to mediate interactions between flaB and TLR5. Our comprehensive and systematic analysis of V. anguillarum flagellins establishes the groundwork for future design of flagellin-based vaccines.
Collapse
Affiliation(s)
- Quanxin Gao
- Zhejiang Provincial Key Laboratory of Aquatic Resources, Conservation and Development, Key Laboratory of Aquatic Animal Genetic Breeding and Nutrition, Chinese Academy of Fishery Sciences, College of Life Science, Huzhou University, Huzhou, China
| | - Shaokui Yi
- Zhejiang Provincial Key Laboratory of Aquatic Resources, Conservation and Development, Key Laboratory of Aquatic Animal Genetic Breeding and Nutrition, Chinese Academy of Fishery Sciences, College of Life Science, Huzhou University, Huzhou, China
| | - Yang Li
- Zhejiang Provincial Key Laboratory of Aquatic Resources, Conservation and Development, Key Laboratory of Aquatic Animal Genetic Breeding and Nutrition, Chinese Academy of Fishery Sciences, College of Life Science, Huzhou University, Huzhou, China
| | - Jinping Luo
- Zhejiang Provincial Key Laboratory of Aquatic Resources, Conservation and Development, Key Laboratory of Aquatic Animal Genetic Breeding and Nutrition, Chinese Academy of Fishery Sciences, College of Life Science, Huzhou University, Huzhou, China
| | - Qianqian Xing
- Zhejiang Provincial Key Laboratory of Aquatic Resources, Conservation and Development, Key Laboratory of Aquatic Animal Genetic Breeding and Nutrition, Chinese Academy of Fishery Sciences, College of Life Science, Huzhou University, Huzhou, China
| | - Xia Yang
- Zhejiang Provincial Key Laboratory of Aquatic Resources, Conservation and Development, Key Laboratory of Aquatic Animal Genetic Breeding and Nutrition, Chinese Academy of Fishery Sciences, College of Life Science, Huzhou University, Huzhou, China
| | - Ming Zhao
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Minghua Min
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Qian Wang
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Yabing Wang
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Lingbo Ma
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Shiming Peng
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| |
Collapse
|
18
|
Liu L, Hu J, Wang Y, Lei H, Xu D. The role and research progress of the balance and interaction between regulatory T cells and other immune cells in obesity with insulin resistance. Adipocyte 2021; 10:66-79. [PMID: 33472506 PMCID: PMC7834085 DOI: 10.1080/21623945.2021.1876375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Metabolic homoeostasis in adipose tissue plays a major role in obesity-related insulin resistance (IR). Regulatory T (Treg) cells have been recorded to regulate metabolic homoeostasis in adipose tissue. However, their specific mechanism is not yet known. This review aims to present the role of Treg cells and other immune cells in obesity-associated IR, focusing on the balance of numbers and functions of Treg cells and other immune cells as well as the crucial role of their interactions in maintaining adipose tissue homoeostasis. Th1 cells, Th17 cells, CD8+ T cells, and pro-inflammatory macrophages mediate the occurrence of obesity and IR by antagonizing Treg cells, while anti-inflammatory dendritic cells, eosinophils and type 2 innate lymphoid cells (ILC2s) regulate the metabolic homoeostasis of adipose tissue by promoting the proliferation and differentiation of Treg cells. γ δ T cells and invariant natural killer T (iNKT) cells have complex effects on Treg cells, and their roles in obesity-associated IR are controversial. The balance of Treg cells and other immune cells can help maintain the metabolic homoeostasis of adipose tissue. Further research needs to explore more specific molecular mechanisms, thus providing more precise directions for the treatment of obesity with IR.
Collapse
Affiliation(s)
- Leiling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiahui Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yating Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Lei
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
19
|
Human macrophages utilize a wide range of pathogen recognition receptors to recognize Legionella pneumophila, including Toll-Like Receptor 4 engaging Legionella lipopolysaccharide and the Toll-like Receptor 3 nucleic-acid sensor. PLoS Pathog 2021; 17:e1009781. [PMID: 34280250 PMCID: PMC8321404 DOI: 10.1371/journal.ppat.1009781] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/29/2021] [Accepted: 07/03/2021] [Indexed: 12/15/2022] Open
Abstract
Cytokines made by macrophages play a critical role in determining the course of Legionella pneumophila infection. Prior murine-based modeling indicated that this cytokine response is initiated upon recognition of L. pneumophila by a subset of Toll-like receptors, namely TLR2, TLR5, and TLR9. Through the use of shRNA/siRNA knockdowns and subsequently CRISPR/Cas9 knockouts (KO), we determined that TRIF, an adaptor downstream of endosomal TLR3 and TLR4, is required for full cytokine secretion by human primary and cell-line macrophages. By characterizing a further set of TLR KO's in human U937 cells, we discerned that, contrary to the viewpoint garnered from murine-based studies, TLR3 and TLR4 (along with TLR2 and TLR5) are in fact vital to the macrophage response in the early stages of L. pneumophila infection. This conclusion was bolstered by showing that i) chemical inhibitors of TLR3 and TLR4 dampen the cytokine output of primary human macrophages and ii) transfection of TLR3 and TLR4 into HEK cells conferred an ability to sense L. pneumophila. TLR3- and TLR4-dependent cytokines promoted migration of human HL-60 neutrophils across an epithelial layer, pointing to the biological importance for the newfound signaling pathway. The response of U937 cells to L. pneumophila LPS was dependent upon TLR4, a further contradiction to murine-based studies, which had concluded that TLR2 is the receptor for Legionella LPS. Given the role of TLR3 in sensing nucleic acid (i.e., dsRNA), we utilized newly-made KO U937 cells to document that DNA-sensing by cGAS-STING and DNA-PK are also needed for the response of human macrophages to L. pneumophila. Given the lack of attention given them in the bacterial field, C-type lectin receptors were similarly examined; but, they were not required. Overall, this study arguably represents the most extensive, single-characterization of Legionella-recognition receptors within human macrophages.
Collapse
|
20
|
Zhao Y, Pu M, Zhang J, Wang Y, Yan X, Yu L, He Z. Recent advancements of nanomaterial-based therapeutic strategies toward sepsis: bacterial eradication, anti-inflammation, and immunomodulation. NANOSCALE 2021; 13:10726-10747. [PMID: 34165483 DOI: 10.1039/d1nr02706a] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Sepsis is a life threatening disease that is caused by a dysregulated host immune response to infection, resulting in tissue damage and organ dysfunction, which account for a high in-hospital mortality (approximately 20%). However, there are still no effective and specific therapeutics for clinical sepsis management. Nanomaterial-based strategies have emerged as promising tools for improving the therapeutic efficacy of sepsis by combating lethal bacterial infection, modulating systemic inflammatory response, preventing multiple organ failure, etc. This review has comprehensively summarized the recent advancements in nanomaterial-based strategies for the management of sepsis and severe complications, in which those nanosystems act either as inherent therapeutics or as nanocarriers for the precise delivery of agents. These formulations mechanically possess antibacterial, anti-inflammatory, immunomodulatory, and anti-oxidative effects, achieving multifunctional synergistic treatment efficacy against sepsis. Furthermore, several cell membrane-derived biomimetic nanoplatforms have been used as decoys to trap and neutralize the pathogenic toxins. The critical role of other adjuvant therapies in sepsis management, including the combination of nanotechnology and stem cell therapy, is also highlighted. Overall, this review provides insights into innovative nanotechnology-based strategies applied in sepsis treatment.
Collapse
Affiliation(s)
- Yi Zhao
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Minju Pu
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Jingwen Zhang
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Yanan Wang
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Xuefeng Yan
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Liangmin Yu
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Zhiyu He
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| |
Collapse
|
21
|
Feng M, Zhou S, Yu Y, Su Q, Li X, Lin W. Regulation of the Migration of Distinct Dendritic Cell Subsets. Front Cell Dev Biol 2021; 9:635221. [PMID: 33681216 PMCID: PMC7933215 DOI: 10.3389/fcell.2021.635221] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/01/2021] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DCs), a class of antigen-presenting cells, are widely present in tissues and apparatuses of the body, and their ability to migrate is key for the initiation of immune activation and tolerogenic immune responses. The importance of DCs migration for their differentiation, phenotypic states, and immunologic functions has attracted widespread attention. In this review, we discussed and compared the chemokines, membrane molecules, and migration patterns of conventional DCs, plasmocytoid DCs, and recently proposed DC subgroups. We also review the promoters and inhibitors that affect DCs migration, including the hypoxia microenvironment, tumor microenvironment, inflammatory factors, and pathogenic microorganisms. Further understanding of the migration mechanisms and regulatory factors of DC subgroups provides new insights for the treatment of diseases, such as infection, tumors, and vaccine preparation.
Collapse
Affiliation(s)
- Meng Feng
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shuping Zhou
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yong Yu
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qinghong Su
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaofan Li
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wei Lin
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
22
|
Environmental signals rather than layered ontogeny imprint the function of type 2 conventional dendritic cells in young and adult mice. Nat Commun 2021; 12:464. [PMID: 33469015 PMCID: PMC7815729 DOI: 10.1038/s41467-020-20659-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 12/13/2020] [Indexed: 01/29/2023] Open
Abstract
Conventional dendritic cells (cDC) are key activators of naive T cells, and can be targeted in adults to induce adaptive immunity, but in early life are considered under-developed or functionally immature. Here we show that, in early life, when the immune system develops, cDC2 exhibit a dual hematopoietic origin and, like other myeloid and lymphoid cells, develop in waves. Developmentally distinct cDC2 in early life, despite being distinguishable by fate mapping, are transcriptionally and functionally similar. cDC2 in early and adult life, however, are exposed to distinct cytokine environments that shape their transcriptional profile and alter their ability to sense pathogens, secrete cytokines and polarize T cells. We further show that cDC2 in early life, despite being distinct from cDC2 in adult life, are functionally competent and can induce T cell responses. Our results thus highlight the potential of harnessing cDC2 for boosting immunity in early life.
Collapse
|
23
|
Vijayan A, Van Maele L, Fougeron D, Cayet D, Sirard JC. The GM-CSF Released by Airway Epithelial Cells Orchestrates the Mucosal Adjuvant Activity of Flagellin. THE JOURNAL OF IMMUNOLOGY 2020; 205:2873-2882. [PMID: 33008952 DOI: 10.4049/jimmunol.2000746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/09/2020] [Indexed: 01/15/2023]
Abstract
The TLR5 agonist flagellin is a potent adjuvant and is currently being developed for use in vaccines. The mechanisms that drive flagellin's activity are influenced by its administration route. Previous studies showed that lung structural cells (especially epithelial cells lining the conducting airways) are pivotal for the efficacy of intranasally administered flagellin-containing vaccines. In this study, we looked at how the airway epithelial cells (AECs) regulate the flagellin-dependent stimulation of Ag-specific CD4+ T cells and the Ab response in mice. Our results demonstrate that after sensing flagellin, AECs trigger the release of GM-CSF in a TLR5-dependent fashion and the doubling of the number of activated type 2 conventional dendritic cells (cDC2s) in draining lymph nodes. Furthermore, the neutralization of GM-CSF reduced cDC2s activation. This resulted in lower of Ag-specific CD4+ T cell count and Ab titers in mice. Our data indicate that during pulmonary immunization, the GM-CSF released by AECs orchestrates the cross-talk between cDC2s and CD4+ T cells and thus drives flagellin's adjuvant effect.
Collapse
Affiliation(s)
- Aneesh Vijayan
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Laurye Van Maele
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Delphine Fougeron
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Delphine Cayet
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Jean-Claude Sirard
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| |
Collapse
|
24
|
Alexander-Miller MA. Challenges for the Newborn Following Influenza Virus Infection and Prospects for an Effective Vaccine. Front Immunol 2020; 11:568651. [PMID: 33042150 PMCID: PMC7524958 DOI: 10.3389/fimmu.2020.568651] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/18/2020] [Indexed: 01/10/2023] Open
Abstract
Newborns are at significantly increased risk of severe disease following infection with influenza virus. This is the collective result of their naïve status, altered immune responsiveness, and the lack of a vaccine that is effective in these individuals. Numerous studies have revealed impairments in both the innate and adaptive arms of the immune system of newborns. The consequence of these alterations is a quantitative and qualitative decrease in both antibody and T cell responses. This review summarizes the hurdles newborns experience in mounting an effective response that can clear influenza virus and limit disease following infection. In addition, the challenges, as well as the opportunities, for developing vaccines that can elicit protective responses in these at risk individuals are discussed.
Collapse
Affiliation(s)
- Martha A Alexander-Miller
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
25
|
Golonka RM, Saha P, Yeoh BS, Chattopadhyay S, Gewirtz AT, Joe B, Vijay-Kumar M. Harnessing innate immunity to eliminate SARS-CoV-2 and ameliorate COVID-19 disease. Physiol Genomics 2020; 52:217-221. [PMID: 32275178 DOI: 10.1152/physiolgenomics.00033.2020] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Rachel M Golonka
- UT Microbiome Consortium, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Piu Saha
- UT Microbiome Consortium, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Beng San Yeoh
- UT Microbiome Consortium, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Saurabh Chattopadhyay
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Andrew T Gewirtz
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Bina Joe
- UT Microbiome Consortium, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Matam Vijay-Kumar
- UT Microbiome Consortium, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio.,Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| |
Collapse
|