1
|
Cao Y, Du X, Yu J, Wang Y, Jin X, Gu B, Yin Q. Seno-antigen-pulsed dendritic cell vaccine induce anti-aging immunity to improve adipose tissue senescence and metabolic abnormalities. Biomed Pharmacother 2024; 179:117433. [PMID: 39260327 DOI: 10.1016/j.biopha.2024.117433] [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: 05/21/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024] Open
Abstract
Anti-aging immunity induced by vaccines was recently reported to enable the elimination of senescent cells. However, the initial immune response to vaccination declines with age, and there is evidence that elderly dendritic cells (DCs) have a reduced capacity to stimulate T cells. Identification of alternative anti-aging vaccine is therefore warranted. Here, we developed a DC vaccine that delivers a cationic protein (CP) fused with the seno-antigen peptides Gpnmb (Gpnmb-CP) into DCs. The Gpnmb-CP-pulsed DC vaccine (Gpnmb-CP-DC) efficiently presented antigens and activated CD8+ T cells, leading to enhanced immune cytotoxicity and memory responses in CD8+ T cells. Thus, the targeted anti-aging immunity triggered by Gpnmb-CP-DC has the ability to selectively eliminate senescent adipocytes and effectively improve age-related metabolic abnormalities in both high-fat diet (HFD)-induced young and aged mice models, as well as in natural aging mouse model. In contrast, the Gpnmb-CP protein vaccine exhibits minimal efficacy in aged mice model. Furthermore, we observed a decreased phagocytic capacity for antigens in aging DCs, accompanied by an upregulation of the immune checkpoint PDL1 expression and a noticeable decline in activated CD8+ T cell. Hence, Gpnmb-CP-DC emerges as a promising vaccine candidate, demonstrating the capacity to induce potent anti-aging immunity, mitigating adipose tissue senescence and metabolic abnormalities, while resilient to the senescent environment of the organism.
Collapse
Affiliation(s)
- Yin Cao
- Department of Andrology, The First Hospital of Jilin University, Changchun, China
| | - Xiaoxue Du
- Departments of Laboratory Medicine, Lequn Branch, The First Hospital of Jilin University, Changchun, China
| | - Jiahong Yu
- Departments of Laboratory Medicine, Lequn Branch, The First Hospital of Jilin University, Changchun, China
| | - Ying Wang
- Academy of Health Management, Changchun University of Chinese Medicine, Changchun, China
| | - Xinliang Jin
- Department of General Surgery, First Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Baijian Gu
- Department of Anorectal, First Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Qiliang Yin
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
2
|
Swain SL. CD4 memory has a hierarchical structure created by requirements for infection-derived signals at an effector checkpoint. Front Immunol 2023; 14:1306433. [PMID: 38152398 PMCID: PMC10751922 DOI: 10.3389/fimmu.2023.1306433] [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: 10/03/2023] [Accepted: 11/28/2023] [Indexed: 12/29/2023] Open
Abstract
Our recent studies reveal that the persistence, location, and amount of both antigen and signals that induce pathogen recognition responses determine the number of CD4 memory cells, the subsets that develop, their location, and hence their protective efficacy. Non-replicating vaccines provide antigen that is short-lived and generate low levels of only some memory subsets that are mostly restricted to secondary lymphoid tissue. In contrast, exposure to long-lived replicating viruses and bacteria provides high levels of diverse antigens in sites of infection and induces strong pathogen recognition signals for extended periods of time, resulting in much higher levels of memory cells of diverse subsets in both lymphoid and nonlymphoid sites. These include memory subsets with highly potent functions such as T follicular helpers and cytotoxic CD4 effectors at sites of infection, where they can most effectively combat the pathogen early after re-infection. These effectors also do not develop without antigen and pathogen recognition signals at the effector stage, and both subsets must receive these signals in the tissue sites where they will become resident. We postulate that this leads to a hierarchical structure of memory, with the strongest memory induced only by replicating pathogens. This paradigm suggests a likely roadmap for markedly improving vaccine design.
Collapse
Affiliation(s)
- Susan L. Swain
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| |
Collapse
|
3
|
Devarajan P, Vong AM, Castonguay CH, Silverstein NJ, Kugler-Umana O, Bautista BL, Kelly KA, Luban J, Swain SL. Cytotoxic CD4 development requires CD4 effectors to concurrently recognize local antigen and encounter type I IFN-induced IL-15. Cell Rep 2023; 42:113182. [PMID: 37776519 PMCID: PMC10842051 DOI: 10.1016/j.celrep.2023.113182] [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: 01/13/2023] [Revised: 07/30/2023] [Accepted: 09/13/2023] [Indexed: 10/02/2023] Open
Abstract
Cytotoxic CD4 T cell effectors (ThCTLs) kill virus-infected major histocompatibility complex (MHC) class II+ cells, contributing to viral clearance. We identify key factors by which influenza A virus infection drives non-cytotoxic CD4 effectors to differentiate into lung tissue-resident ThCTL effectors. We find that CD4 effectors must again recognize cognate antigen on antigen-presenting cells (APCs) within the lungs. Both dendritic cells and B cells are sufficient as APCs, but CD28 co-stimulation is not needed. Optimal generation of ThCTLs requires signals induced by the ongoing infection independent of antigen presentation. Infection-elicited type I interferon (IFN) induces interleukin-15 (IL-15), which, in turn, supports CD4 effector differentiation into ThCTLs. We suggest that these multiple spatial, temporal, and cellular requirements prevent excessive lung ThCTL responses when virus is already cleared but ensure their development when infection persists. This supports a model where continuing infection drives the development of multiple, more differentiated subsets of CD4 effectors by distinct pathways.
Collapse
Affiliation(s)
| | - Allen M Vong
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Catherine H Castonguay
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Noah J Silverstein
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Olivia Kugler-Umana
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Bianca L Bautista
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Karen A Kelly
- Department of Animal Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jeremy Luban
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Susan L Swain
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
4
|
Linterman MA. Age-dependent changes in T follicular helper cells shape the humoral immune response to vaccination. Semin Immunol 2023; 69:101801. [PMID: 37379670 DOI: 10.1016/j.smim.2023.101801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023]
Abstract
Vaccination is an excellent strategy to limit the morbidity and mortality associated with infectious disease. Vaccination creates protective, long-lived antibody-mediated immunity by inducing the germinal centre response, an intricate immune reaction that produces memory B cells and long-lived antibody-secreting plasma cells that provide protection against (re)infection. The magnitude and quality of the germinal centre response declines with age, contributing to poor vaccine-induced immunity in older individuals. T follicular helper cells are essential for the formation and function of the germinal centre response. This review will discuss how age-dependent changes in T follicular helper cells influence the germinal centre response, and the evidence that age-dependent changes need not be a barrier to successful vaccination in the later years of life.
Collapse
Affiliation(s)
- Michelle A Linterman
- Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom.
| |
Collapse
|
5
|
Jones MC, Castonguay C, Nanaware PP, Weaver GC, Stadinski B, Kugler-Umana OA, Huseby ES, Stern LJ, McKinstry KK, Strutt TM, Devarajan P, Swain SL. CD4 Effector TCR Avidity for Peptide on APC Determines the Level of Memory Generated. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1950-1961. [PMID: 37093656 PMCID: PMC10247507 DOI: 10.4049/jimmunol.2200337] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 03/30/2023] [Indexed: 04/25/2023]
Abstract
Initial TCR affinity for peptide Ag is known to impact the generation of memory; however, its contributions later, when effectors must again recognize Ag at 5-8 d postinfection to become memory, is unclear. We examined whether the effector TCR affinity for peptide at this "effector checkpoint" dictates the extent of memory and degree of protection against rechallenge. We made an influenza A virus nucleoprotein (NP)-specific TCR transgenic mouse strain, FluNP, and generated NP-peptide variants that are presented by MHC class II to bind to the FluNP TCR over a broad range of avidity. To evaluate the impact of avidity in vivo, we primed naive donor FluNP in influenza A virus-infected host mice, purified donor effectors at the checkpoint, and cotransferred them with the range of peptides pulsed on activated APCs into second uninfected hosts. Higher-avidity peptides yielded higher numbers of FluNP memory cells in spleen and most dramatically in lung and draining lymph nodes and induced better protection against lethal influenza infection. Avidity determined memory cell number, not cytokine profile, and already impacted donor cell number within several days of transfer. We previously found that autocrine IL-2 production at the checkpoint prevents default effector apoptosis and supports memory formation. Here, we find that peptide avidity determines the level of IL-2 produced by these effectors and that IL-2Rα expression by the APCs enhances memory formation, suggesting that transpresentation of IL-2 by APCs further amplifies IL-2 availability. Secondary memory generation was also avidity dependent. We propose that this regulatory pathway selects CD4 effectors of highest affinity to progress to memory.
Collapse
Affiliation(s)
- Michael C. Jones
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Catherine Castonguay
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Padma P. Nanaware
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Grant C. Weaver
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Brian Stadinski
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Olivia A. Kugler-Umana
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Eric S. Huseby
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Lawrence J. Stern
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Karl Kai McKinstry
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL. 32827,USA
| | - Tara M. Strutt
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL. 32827,USA
| | - Priyadharshini Devarajan
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Susan L. Swain
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
6
|
Lv R, Liu X, Zhang Y, Dong N, Wang X, He Y, Yue H, Yin Q. Pathophysiological mechanisms and therapeutic approaches in obstructive sleep apnea syndrome. Signal Transduct Target Ther 2023; 8:218. [PMID: 37230968 DOI: 10.1038/s41392-023-01496-3] [Citation(s) in RCA: 80] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
Obstructive sleep apnea syndrome (OSAS) is a common breathing disorder in sleep in which the airways narrow or collapse during sleep, causing obstructive sleep apnea. The prevalence of OSAS continues to rise worldwide, particularly in middle-aged and elderly individuals. The mechanism of upper airway collapse is incompletely understood but is associated with several factors, including obesity, craniofacial changes, altered muscle function in the upper airway, pharyngeal neuropathy, and fluid shifts to the neck. The main characteristics of OSAS are recurrent pauses in respiration, which lead to intermittent hypoxia (IH) and hypercapnia, accompanied by blood oxygen desaturation and arousal during sleep, which sharply increases the risk of several diseases. This paper first briefly describes the epidemiology, incidence, and pathophysiological mechanisms of OSAS. Next, the alterations in relevant signaling pathways induced by IH are systematically reviewed and discussed. For example, IH can induce gut microbiota (GM) dysbiosis, impair the intestinal barrier, and alter intestinal metabolites. These mechanisms ultimately lead to secondary oxidative stress, systemic inflammation, and sympathetic activation. We then summarize the effects of IH on disease pathogenesis, including cardiocerebrovascular disorders, neurological disorders, metabolic diseases, cancer, reproductive disorders, and COVID-19. Finally, different therapeutic strategies for OSAS caused by different causes are proposed. Multidisciplinary approaches and shared decision-making are necessary for the successful treatment of OSAS in the future, but more randomized controlled trials are needed for further evaluation to define what treatments are best for specific OSAS patients.
Collapse
Affiliation(s)
- Renjun Lv
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Xueying Liu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Yue Zhang
- Department of Geriatrics, the 2nd Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Na Dong
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Xiao Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Yao He
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Hongmei Yue
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
| |
Collapse
|
7
|
Shuai L, She J, Diao R, Zhao H, Liu X, Hu Q, Li D, Su D, Ye X, Guo Y, Zhong M, Wang L. Hydroxychloroquine protects against autoimmune premature ovarian insufficiency by modulating the Treg/Th17 cell ratio in BALB/c mice. Am J Reprod Immunol 2023; 89:e13686. [PMID: 36752682 DOI: 10.1111/aji.13686] [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: 11/21/2022] [Revised: 01/16/2023] [Accepted: 01/28/2023] [Indexed: 02/09/2023] Open
Abstract
AIMS The role of hydroxychloroquine (HCQ) in premature ovarian insufficiency (POI) remains unclear. The purpose of this study was to evaluate the effect of HCQ on ovarian function in mice with POI and to clarify its potential mechanisms. METHODS POI was induced in mice by injection with zona pellucida 3 peptide (pZP3), and HCQ was administered intragastrically. Stages of the estrous cycle were determined using vaginal cytology. The ovarian structure was observed under a microscope after hematoxylin-eosin staining. The levels of serum hormones and anti-ZP antibody (aZPAb) were measured using enzyme-linked immunosorbent assay (ELISA). The expression levels of CD4, CD45, and ZP2, ZP3 were determined using immunofluorescence and immunohistochemistry, respectively. The T regulatory (Treg)/ T helper 17 (Th17) cell ratio was analyzed using flow cytometry analysis. Western blotting was performed to assess the expression levels of proteins, transcription factors and cytokines. RESULTS Administration of HCQ to mice with POI greatly restored their estrus cycle. In the treatment group compared to the POI group, estradiol (E2 ) levels were higher, and follicle stimulating hormone (FSH) levels were lower. In addition, following pZP3, HCQ treatment increased ZP2 and ZP3 expression. Additionally, by inhibiting the activation of the TLR7 signaling pathway, HCQ attenuated the infiltration of inflammatory cells and prevented the activated naive CD4+ T cells from developing into Th17 cells. CONCLUSION Our findings showed that HCQ effectively restored ovarian function by altering the Treg/Th17 cell ratio in mice with POI, indicating that HCQ maybe a promising therapeutic method for patients with POI.
Collapse
Affiliation(s)
- Ling Shuai
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First Affiliated Hospital of Shenzhen University, Reproductive Medicine Centre, Shenzhen Second People's Hospital, Shenzhen, China
| | - Jiajie She
- The First Affiliated Hospital of Shenzhen University, Reproductive Medicine Centre, Shenzhen Second People's Hospital, Shenzhen, China
| | - Ruiying Diao
- The First Affiliated Hospital of Shenzhen University, Reproductive Medicine Centre, Shenzhen Second People's Hospital, Shenzhen, China
| | - Huihui Zhao
- Department of Obstetrics and Gynecology, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Xuemin Liu
- The First Affiliated Hospital of Shenzhen University, Reproductive Medicine Centre, Shenzhen Second People's Hospital, Shenzhen, China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, China
| | - Qingyu Hu
- The First Affiliated Hospital of Shenzhen University, Reproductive Medicine Centre, Shenzhen Second People's Hospital, Shenzhen, China
- Department of Medicine, Shenzhen University, Shenzhen, China
| | - Dongdong Li
- The First Affiliated Hospital of Shenzhen University, Reproductive Medicine Centre, Shenzhen Second People's Hospital, Shenzhen, China
- Department of Gynecology and Obstetrics, Graduate College of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Danna Su
- The First Affiliated Hospital of Shenzhen University, Reproductive Medicine Centre, Shenzhen Second People's Hospital, Shenzhen, China
- Shantou University Medical College, Shantou, China
| | - Xiaofeng Ye
- The First Affiliated Hospital of Shenzhen University, Reproductive Medicine Centre, Shenzhen Second People's Hospital, Shenzhen, China
| | - Yan Guo
- The First Affiliated Hospital of Shenzhen University, Reproductive Medicine Centre, Shenzhen Second People's Hospital, Shenzhen, China
- Guangdong Medical University, Zhanjiang, China
| | - Mei Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liping Wang
- The First Affiliated Hospital of Shenzhen University, Reproductive Medicine Centre, Shenzhen Second People's Hospital, Shenzhen, China
| |
Collapse
|
8
|
Ananya A, Holden KG, Gu Z, Nettleton D, Mallapragada SK, Wannemuehler MJ, Kohut ML, Narasimhan B. "Just right" combinations of adjuvants with nanoscale carriers activate aged dendritic cells without overt inflammation. Immun Ageing 2023; 20:10. [PMID: 36895007 PMCID: PMC9996592 DOI: 10.1186/s12979-023-00332-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/05/2023] [Indexed: 03/11/2023]
Abstract
BACKGROUND The loss in age-related immunological markers, known as immunosenescence, is caused by a combination of factors, one of which is inflammaging. Inflammaging is associated with the continuous basal generation of proinflammatory cytokines. Studies have demonstrated that inflammaging reduces the effectiveness of vaccines. Strategies aimed at modifying baseline inflammation are being developed to improve vaccination responses in older adults. Dendritic cells have attracted attention as an age-specific target because of their significance in immunization as antigen presenting cells that stimulate T lymphocytes. RESULTS In this study, bone marrow derived dendritic cells (BMDCs) were generated from aged mice and used to investigate the effects of combinations of adjuvants, including Toll-like receptor, NOD2, and STING agonists with polyanhydride nanoparticles and pentablock copolymer micelles under in vitro conditions. Cellular stimulation was characterized via expression of costimulatory molecules, T cell-activating cytokines, proinflammatory cytokines, and chemokines. Our results indicate that multiple TLR agonists substantially increase costimulatory molecule expression and cytokines associated with T cell activation and inflammation in culture. In contrast, NOD2 and STING agonists had only a moderate effect on BMDC activation, while nanoparticles and micelles had no effect by themselves. However, when nanoparticles and micelles were combined with a TLR9 agonist, a reduction in the production of proinflammatory cytokines was observed while maintaining increased production of T cell activating cytokines and enhancing cell surface marker expression. Additionally, combining nanoparticles and micelles with a STING agonist resulted in a synergistic impact on the upregulation of costimulatory molecules and an increase in cytokine secretion from BMDCs linked with T cell activation without excessive secretion of proinflammatory cytokines. CONCLUSIONS These studies provide new insights into rational adjuvant selection for vaccines for older adults. Combining appropriate adjuvants with nanoparticles and micelles may lead to balanced immune activation characterized by low inflammation, setting the stage for designing next generation vaccines that can induce mucosal immunity in older adults.
Collapse
Affiliation(s)
- Ananya Ananya
- Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Kaitlyn G Holden
- Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA
| | - Zhiling Gu
- Department of Statistics, Iowa State University, Ames, IA, 50011, USA
| | - Dan Nettleton
- Department of Statistics, Iowa State University, Ames, IA, 50011, USA
| | - Surya K Mallapragada
- Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| | | | - Marian L Kohut
- Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA
- Department of Kinesiology, Iowa State University, Ames, IA, 50011, USA
| | - Balaji Narasimhan
- Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA.
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA.
| |
Collapse
|
9
|
Kugler‐Umana O, Zhang W, Kuang Y, Liang J, Castonguay CH, Tonkonogy SL, Marshak‐Rothstein A, Devarajan P, Swain SL. IgD + age-associated B cells are the progenitors of the main T-independent B cell response to infection that generates protective Ab and can be induced by an inactivated vaccine in the aged. Aging Cell 2022; 21:e13705. [PMID: 36056604 PMCID: PMC9577953 DOI: 10.1111/acel.13705] [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: 01/26/2022] [Revised: 07/18/2022] [Accepted: 08/18/2022] [Indexed: 01/25/2023] Open
Abstract
Age-associated B cells (ABC) accumulate with age and are associated with autoimmunity and chronic infection. However, their contributions to acute infection in the aged and their developmental pathways are unclear. We find that the response against influenza A virus infection in aged mice is dominated by a Fas+ GL7- effector B cell population we call infection-induced ABC (iABC). Most iABC express IgM and include antibody-secreting cells in the spleen, lung, and bone marrow. We find that in response to influenza, IgD+ CD21- CD23- ABC are the precursors of iABC and become memory B cells. These IgD+ ABC develop in germ-free mice, so are independent of foreign antigen recognition. The response of ABC to influenza infection, resulting in iABC, is T cell independent and requires both extrinsic TLR7 and TLR9 signals. In response to influenza infection, IgD+ ABC can induce a faster recovery of weight and higher total anti-influenza IgG and IgM titers that can neutralize virus. Immunization with whole inactivated virus also generates iABC in aged mice. Thus, in unimmunized aged mice, whose other B and T cell responses have waned, IgD+ ABC are likely the naive B cells with the potential to become Ab-secreting cells and to provide protection from infection in the aged.
Collapse
Affiliation(s)
- Olivia Kugler‐Umana
- Department of PathologyUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Wenliang Zhang
- Department of PathologyUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Yi Kuang
- Department of PathologyUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Jialing Liang
- Department of PathologyUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Catherine H. Castonguay
- Department of PathologyUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Susan L. Tonkonogy
- College of Veterinary MedicineNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Ann Marshak‐Rothstein
- Department of MedicineUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | | | - Susan L. Swain
- Department of PathologyUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| |
Collapse
|
10
|
Lee JL, Linterman MA. Mechanisms underpinning poor antibody responses to vaccines in ageing. Immunol Lett 2022; 241:1-14. [PMID: 34767859 PMCID: PMC8765414 DOI: 10.1016/j.imlet.2021.11.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 12/13/2022]
Abstract
Vaccines are a highly effective intervention for conferring protection against infections and reducing the associated morbidity and mortality in vaccinated individuals. However, ageing is often associated with a functional decline in the immune system that results in poor antibody production in older individuals after vaccination. A key contributing factor of this age-related decline in vaccine efficacy is the reduced size and function of the germinal centre (GC) response. GCs are specialised microstructures where B cells undergo affinity maturation and diversification of their antibody genes, before differentiating into long-lived antibody-secreting plasma cells and memory B cells. The GC response requires the coordinated interaction of many different cell types, including B cells, T follicular helper (Tfh) cells, T follicular regulatory (Tfr) cells and stromal cell subsets like follicular dendritic cells (FDCs). This review discusses how ageing affects different components of the GC reaction that contribute to its limited output and ultimately impaired antibody responses in older individuals after vaccination. An understanding of the mechanisms underpinning the age-related decline in the GC response is crucial in informing strategies to improve vaccine efficacy and extend the healthy lifespan amongst older people.
Collapse
Affiliation(s)
- Jia Le Lee
- Immunology Program, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| | - Michelle A Linterman
- Immunology Program, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| |
Collapse
|
11
|
Zhang L, Ko CY, Zeng YM. Immunoregulatory Effect of Short-Chain Fatty Acids from Gut Microbiota on Obstructive Sleep Apnea-Associated Hypertension. Nat Sci Sleep 2022; 14:393-405. [PMID: 35299627 PMCID: PMC8922759 DOI: 10.2147/nss.s354742] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/27/2022] [Indexed: 12/12/2022] Open
Abstract
The intestine is the largest bacterial ecosystem and immune response organ of the human body. The microbiota regulates the metabolic and immune functions of the host through their metabolites. Short-chain fatty acids (SCFAs) are part of the metabolites of the gut microbiota (GM), providing energy to intestinal epithelial cells and regulating the immune system. A decrease in SCFA-producing bacteria, imbalanced effector T-helper cells (Th cells), and increasing corresponding inflammatory cytokine were found in both animal models and clinical patients with obstructive sleep apnea (OSA) and hypertension (HTN). Intervention with probiotics, prebiotics, or postbiotics in animal models simulating OSA-associated HTN restored blood pressure to normal, which allows the hypothesis that GM are involved in the pathophysiology of OSA-induced HTN patients through their metabolites' SCFAs; however, the exact regulatory mechanism is not completely clear. This review describes the potential mechanisms of SCFAs, a major metabolite of the GM, in the pathology of OSA-induced HTN, from the perspective of immune system regulation in the available studies.
Collapse
Affiliation(s)
- Li Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, People's Republic of China.,Respiratory Medicine Center of Fujian Province, Quanzhou, 362000, People's Republic of China
| | - Chih-Yuan Ko
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, People's Republic of China.,Respiratory Medicine Center of Fujian Province, Quanzhou, 362000, People's Republic of China.,Department of Clinical Nutrition, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, People's Republic of China.,School of Public Health, Fujian Medical University, Fuzhou, Fujian, 350122, People's Republic of China
| | - Yi-Ming Zeng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, People's Republic of China.,Respiratory Medicine Center of Fujian Province, Quanzhou, 362000, People's Republic of China
| |
Collapse
|
12
|
Swain SL, Jones MC, Devarajan P, Xia J, Dutton RW, Strutt TM, McKinstry KK. Durable CD4 T-Cell Memory Generation Depends on Persistence of High Levels of Infection at an Effector Checkpoint that Determines Multiple Fates. Cold Spring Harb Perspect Biol 2021; 13:a038182. [PMID: 33903157 PMCID: PMC8559547 DOI: 10.1101/cshperspect.a038182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We have discovered that the determination of CD4 effector and memory fates after infection is regulated not only by initial signals from antigen and pathogen recognition, but also by a second round of such signals at a checkpoint during the effector response. Signals to effectors determine their subsequent fate, inducing further progression to tissue-restricted follicular helpers, cytotoxic CD4 effectors, and long-lived memory cells. The follicular helpers help the germinal center B-cell responses that give rise to high-affinity long-lived antibody responses and memory B cells that synergize with T-cell memory to provide robust long-lived protection. We postulate that inactivated vaccines do not provide extended signals from antigen and pathogen beyond a few days, and thus elicit ineffective CD4 T- and B-cell effector responses and memory. Defining the mechanisms that underlie effective responses should provide insights necessary to develop vaccine strategies that induce more effective and durable immunity.
Collapse
Affiliation(s)
- Susan L Swain
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Ave, Worcester, Massachusetts 01655, USA
| | - Michael C Jones
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Ave, Worcester, Massachusetts 01655, USA
| | - Priyadharshini Devarajan
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Ave, Worcester, Massachusetts 01655, USA
| | - Jingya Xia
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Ave, Worcester, Massachusetts 01655, USA
| | - Richard W Dutton
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Ave, Worcester, Massachusetts 01655, USA
| | - Tara M Strutt
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32827, USA
| | - K Kai McKinstry
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32827, USA
| |
Collapse
|
13
|
Abstract
Dendritic cells (DCs) are efficient antigen-presenting cells that serve as a link between the innate and adaptive immune systems. These cells are broadly involved in cellular and humoral immune responses by presenting antigens to initiate T cell reactions, cytokine and chemokine secretion, T cell differentiation and expansion, B cell activation and regulation, and the mediation of immune tolerance. The functions of DCs depend on their activation status, which is defined by the stages of maturation, phenotype differentiation, and migration ability, among other factors. IL-6 is a soluble mediator mainly produced by a variety of immune cells, including DCs, that exerts pleiotropic effects on immune and inflammatory responses through interaction with specific receptors expressed on the surface of target cells. Here, we review the role of IL-6, when generated in an inflammatory context or as derived from DCs, in modulating the biologic function and activation status of DCs and emphasize the importance of searching for novel strategies to target the IL-6/IL-6 signaling pathway as a means to diminish the inflammatory activity of DCs in immune response or to prime the immunogenic activity of DCs in immunosuppressive conditions.
Collapse
Affiliation(s)
- Yu-Dong Xu
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mi Cheng
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pan-Pan Shang
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong-Qing Yang
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
14
|
Ghosh D, Jiang W, Mukhopadhyay D, Mellins ED. New insights into B cells as antigen presenting cells. Curr Opin Immunol 2021; 70:129-137. [PMID: 34242927 DOI: 10.1016/j.coi.2021.06.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 01/06/2023]
Abstract
In addition to their role as antibody producing cells, B cells make a critical contribution to adaptive immune responses by functioning as professional antigen-presenting cells (APC). Distinctive features of B cells as APC include the expression of the B cell receptor (BCR) for antigen and regulated expression of HLA-DO. Here, we discuss recent progress in investigation of B cells as APC. We start with an update on the canonical MHC class II antigen presentation pathway in B cells and alternative pathways, including generation of extracellular vesicles. Turning to APC function, we highlight the roles of B cells as thymic APC, as APC for T follicular helper (TFH), as APC for CD4 memory T cells and as presenters of idiotypic BCR determinants. We also note recent examples that link B cell Ag-presentation to disease. Emerging evidence indicates that, in addition to unique features of B cells compared to other professional APC, there is appreciable heterogeneity among B cells, arising from, for example, B cell activation state or the microenvironment.
Collapse
Affiliation(s)
- Debopam Ghosh
- Department of Pediatrics, Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wei Jiang
- Department of Pediatrics, Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dhriti Mukhopadhyay
- Department of Surgery, University of Arizona, Tucson, AZ 85724, USA; Tuba City Regional Health Care, Tuba City, AZ 86045, USA
| | - Elizabeth D Mellins
- Department of Pediatrics, Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
15
|
Swain SL, Kugler-Umana O, Tonkonogy SL. "An Intrinsic Program Determines Key Age-Associated Changes in Adaptive Immunity that Limit Response to Non-Pathogens.". FRONTIERS IN AGING 2021; 2:701900. [PMID: 35382063 PMCID: PMC8979546 DOI: 10.3389/fragi.2021.701900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/09/2021] [Indexed: 01/14/2023]
Abstract
As mice age their adaptive immune system changes dramatically, leading to weakened responses to newly encountered antigens and poor efficacy of vaccines. A shared pattern emerges in the aged, with both CD4 T and B cell responses requiring higher levels of pathogen recognition. Moreover, in aged germ-free mice we find accumulation of the same novel age-associated T and B cell subsets that we and others have previously identified using mice maintained in normal laboratory animal housing conditions, suggesting that their development follows an intrinsic program.
Collapse
Affiliation(s)
- Susan L. Swain
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Olivia Kugler-Umana
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Susan L. Tonkonogy
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
16
|
The impact of immuno-aging on SARS-CoV-2 vaccine development. GeroScience 2021; 43:31-51. [PMID: 33569701 PMCID: PMC7875765 DOI: 10.1007/s11357-021-00323-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
The SARS-CoV-2 pandemic has almost 56 million confirmed cases resulting in over 1.3 million deaths as of November 2020. This infection has proved more deadly to older adults (those >65 years of age) and those with immunocompromising conditions. The worldwide population aged 65 years and older is increasing, and the total number of aged individuals will outnumber those younger than 65 years by the year 2050. Aging is associated with a decline in immune function and chronic activation of inflammation that contributes to enhanced viral susceptibility and reduced responses to vaccination. Here we briefly review the pathogenicity of the virus, epidemiology and clinical response, and the underlying mechanisms of human aging in improving vaccination. We review current methods to improve vaccination in the older adults using novel vaccine platforms and adjuvant systems. We conclude by summarizing the existing clinical trials for a SARS-CoV-2 vaccine and discussing how to address the unique challenges for vaccine development presented with an aging immune system.
Collapse
|
17
|
Saleh R, Sasidharan Nair V, Al-Dhaheri M, Khawar M, Abu Nada M, Alajez NM, Elkord E. RNA-Seq Analysis of Colorectal Tumor-Infiltrating Myeloid-Derived Suppressor Cell Subsets Revealed Gene Signatures of Poor Prognosis. Front Oncol 2020; 10:604906. [PMID: 33312958 PMCID: PMC7703275 DOI: 10.3389/fonc.2020.604906] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022] Open
Abstract
Elevated levels of myeloid-derived suppressor cells (MDSCs), including polymorphonuclear MDSCs (PMN-MDSCs) and immature MDSCs (I-MDSCs), are usually associated with disease progression in cancer patients, including colorectal cancer (CRC). However, biological mechanisms and molecular pathways regulated by MDSC subpopulations in the CRC tumor microenvironment (TME) have not been fully investigated. In this study, we performed transcriptomic analysis of tumor-infiltrating I-MDSCs and PMN-MDSCs isolated from tumor tissues of six CRC patients, compared to antigen-presenting cells (APCs). We also compared the transcriptomic profiles of tumor-infiltrating PMN-MDSCs to I-MDSCs. Our results showed different molecular pathways regulated by each MDSC subset, potentially reflecting their phenotypical/molecular/functional characteristics in the CRC TME. Moreover, we identified gene signatures in PMN-MDSC and I-MDSC of poor overall survival (OS) and disease-free survival (DFS) using the Cancer Genome Atlas (TCGA) dataset from patients with colon adenocarcinoma (COAD). However, functional studies are required to validate these findings.
Collapse
Affiliation(s)
- Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Varun Sasidharan Nair
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | | | - Mahwish Khawar
- Department of Surgery, Hamad Medical Corporation, Doha, Qatar
| | | | - Nehad M Alajez
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.,Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, Manchester, United Kingdom
| |
Collapse
|
18
|
Xia J, Kuang Y, Liang J, Jones M, Swain SL. Influenza Vaccine-Induced CD4 Effectors Require Antigen Recognition at an Effector Checkpoint to Generate CD4 Lung Memory and Antibody Production. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:2077-2090. [PMID: 32929040 PMCID: PMC8525320 DOI: 10.4049/jimmunol.2000597] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023]
Abstract
Previously, we discovered that influenza-generated CD4 effectors must recognize cognate Ag at a defined effector checkpoint to become memory cells. Ag recognition was also required for efficient protection against lethal influenza infection. To extend these findings, we investigated if vaccine-generated effectors would have the same requirement. We compared live infection with influenza to an inactivated whole influenza vaccine. Live infection provided strong, long-lasting Ag presentation that persisted through the effector phase. It stimulated effector generation, long-lived CD4 memory generation, and robust generation of Ab-producing B cells. In contrast, immunization with an inactivated virus vaccine, even when enhanced by additional Ag-pulsed APC, presented Ag for 3 d or less and generated few CD4 memory cells or long-lived Ab-producing B cells. To test if checkpoint Ag addition would enhance this vaccine response, we immunized mice with inactivated vaccine and injected Ag-pulsed activated APC at the predicted effector checkpoint to provide Ag presentation to the effector CD4 T cells. This enhanced generation of CD4 memory, especially tissue-resident memory in the lung, long-lived bone marrow Ab-secreting cells, and influenza-specific IgG Ab. All responses increased as we increased the density of peptide Ag on the APC to high levels. This suggests that CD4 effectors induced by inactivated vaccine require high levels of cognate Ag recognition at the effector checkpoint to most efficiently become memory cells. Thus, we suggest that nonlive vaccines will need to provide high levels of Ag recognition throughout the effector checkpoint to optimize CD4 memory generation.
Collapse
Affiliation(s)
- Jingya Xia
- Department of Pathology, 368 Plantation Ave, University of Massachusetts Medical School, Worcester, MA 01655
| | - Yi Kuang
- Department of Pathology, 368 Plantation Ave, University of Massachusetts Medical School, Worcester, MA 01655,Merck Exploratory Science Center, Cambridge, MA 02141
| | - Jialing Liang
- Department of Pathology, 368 Plantation Ave, University of Massachusetts Medical School, Worcester, MA 01655
| | - Michael Jones
- Department of Pathology, 368 Plantation Ave, University of Massachusetts Medical School, Worcester, MA 01655
| | - Susan L. Swain
- Department of Pathology, 368 Plantation Ave, University of Massachusetts Medical School, Worcester, MA 01655
| |
Collapse
|
19
|
Fukuyama H, Shinnakasu R, Kurosaki T. Influenza vaccination strategies targeting the hemagglutinin stem region. Immunol Rev 2020; 296:132-141. [PMID: 32542739 PMCID: PMC7323124 DOI: 10.1111/imr.12887] [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: 03/30/2020] [Revised: 05/03/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022]
Abstract
Influenza is one of the best examples of highly mutable viruses that are able to escape immune surveillance. Indeed, in response to influenza seasonal infection or vaccination, the majority of the induced antibodies are strain‐specific. Current vaccine against the seasonal strains with the strategy of surveillance‐prediction‐vaccine does not cover an unmet virus strain leading to pandemic. Recently, antibodies targeting conserved epitopes on the hemagglutinin (HA) protein have been identified, albeit rarely, and they often showed broad protection. These antibody discoveries have brought the feasibility to develop a universal vaccine. Most of these antibodies bind the HA stem domain and accumulate in the memory B cell compartment. Broadly reactive stem‐biased memory responses were induced by infection with antigenically divergent influenza strains and were able to eradicate these viruses, together indicating the importance of generating memory B cells expressing high‐quality anti‐stem antibodies. Here, we emphasize recent progress in our understanding of how such memory B cells can be generated and discuss how these advances may be relevant to the quest for a universal influenza vaccine.
Collapse
Affiliation(s)
- Hidehiro Fukuyama
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.,Cellular Systems Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan.,INSERM EST, Strasbourg Cedex 2, France
| | - Ryo Shinnakasu
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Tomohiro Kurosaki
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.,Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| |
Collapse
|
20
|
Stebegg M, Bignon A, Hill DL, Silva-Cayetano A, Krueger C, Vanderleyden I, Innocentin S, Boon L, Wang J, Zand MS, Dooley J, Clark J, Liston A, Carr E, Linterman MA. Rejuvenating conventional dendritic cells and T follicular helper cell formation after vaccination. eLife 2020; 9:52473. [PMID: 32204792 PMCID: PMC7093110 DOI: 10.7554/elife.52473] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 03/12/2020] [Indexed: 12/22/2022] Open
Abstract
Germinal centres (GCs) are T follicular helper cell (Tfh)-dependent structures that form in response to vaccination, producing long-lived antibody secreting plasma cells and memory B cells that protect against subsequent infection. With advancing age the GC and Tfh cell response declines, resulting in impaired humoral immunity. We sought to discover what underpins the poor Tfh cell response in ageing and whether it is possible to correct it. Here, we demonstrate that older people and aged mice have impaired Tfh cell differentiation upon vaccination. This deficit is preceded by poor activation of conventional dendritic cells type 2 (cDC2) due to reduced type 1 interferon signalling. Importantly, the Tfh and cDC2 cell response can be boosted in aged mice by treatment with a TLR7 agonist. This demonstrates that age-associated defects in the cDC2 and Tfh cell response are not irreversible and can be enhanced to improve vaccine responses in older individuals.
Collapse
Affiliation(s)
- Marisa Stebegg
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom
| | - Alexandre Bignon
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom
| | - Danika Lea Hill
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom
| | - Alyssa Silva-Cayetano
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom
| | - Christel Krueger
- Epigenetics Programme, Babraham Institute, Cambridge, United Kingdom
| | - Ine Vanderleyden
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom
| | - Silvia Innocentin
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom
| | | | - Jiong Wang
- Division of Nephrology, Department of Medicine and Clinical and Translational Science Institute, University of Rochester Medical Center, Rochester, United States
| | - Martin S Zand
- Division of Nephrology, Department of Medicine and Clinical and Translational Science Institute, University of Rochester Medical Center, Rochester, United States
| | - James Dooley
- Autoimmune Genetics Laboratory, VIB and University of Leuven, Leuven, Belgium
| | - Jonathan Clark
- Biological Chemistry, Babraham Institute, Cambridge, United Kingdom
| | - Adrian Liston
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom
| | - Edward Carr
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom.,Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Michelle A Linterman
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom
| |
Collapse
|
21
|
Kugler-Umana O, Devarajan P, Swain SL. Understanding the Heterogeneous Population of Age-Associated B Cells and Their Contributions to Autoimmunity and Immune Response to Pathogens. Crit Rev Immunol 2020; 40:297-309. [PMID: 33426819 PMCID: PMC8118092 DOI: 10.1615/critrevimmunol.2020034934] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In humans and mice, susceptibility to infections and autoimmunity increases with age due to age-associated changes in innate and adaptive immune responses. Aged innate cells are also less active, leading to decreased naive T- and B-cell responses. Aging innate cells contribute to an overall heightened inflammatory environment. Naive T and B cells undergo cell-intrinsic age-related changes that result in reduced effector and memory responses. However, previously established B- and T-cell memory responses persist with age. One dramatic change is the appearance of a newly recognized population of age-associated B cells (ABCs) that has a unique cluster of differentiation (CD)21-CD23- phenotype. Here, we discuss the discovery and origins of the naive phenotype immunoglobulin (Ig)D+ versus activated CD11c+T-bet+ ABCs, with a focus on protective and pathogenic properties. In humans and mice, antigen-experienced CD11c+T-bet+ ABCs increase with autoimmunity and appear in response to bacterial and viral infections. However, our analyses indicate that CD21-CD23- ABCs include a resting, naive, progenitor ABC population that expresses IgD. Similar to generation of CD11c+T-bet+ ABCs, naive ABC response to pathogens depends on toll-like receptor stimulation, making this a key feature of ABC activation. Here, we put forward a potential developmental map of distinct subsets from putative naive ABCs. We suggest that defining signals that can harness the naive ABC response may contribute to protection against pathogens in the elderly. CD11c+T-bet+ ABCs may be useful targets for therapeutic strategies to counter autoimmunity.
Collapse
Affiliation(s)
- Olivia Kugler-Umana
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | - Susan L. Swain
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
22
|
Sun W, Li Q, Ning X, Yang Y, Guo J, Zhu Q, Guo Y, Li H, Wang Y, Zhou Y, Kou Z. TFPR1 acts as an immune regulator and an efficient adjuvant for proteins and peptides by activating immune cells, primarily through TLR2. Vaccine 2019; 38:288-297. [PMID: 31629567 DOI: 10.1016/j.vaccine.2019.10.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 09/03/2019] [Accepted: 10/05/2019] [Indexed: 02/08/2023]
Abstract
Triflin, a non-toxic protein found in the venom of the Habu snake, belongs to the CRISP (cysteine-rich secretory protein) family, which comprises two domains: a C-terminal cysteine-rich domain (CRD) and an N-terminal pathogenesis-related-1 (PR-1) domain. The function of the highly structurally conserved PR-1 domain is unknown. Here, we successfully expressed the PR-1 domain of triflin (hereafter called TFPR1) in E. coli. Animal experiments showed that TFPR1 augmented Th1-biased antibody- and cell-mediated immune responses in mice immunized with two protein antigens (OVA and HBsAg) or a peptide antigen (HIV-1 pep). A flow cytometry-based binding assay and in vitro stimulation with TFPR1 showed that it triggered Th1-biased proinflammatory and immunoregulatory cytokine secretion primarily by binding to B cells and macrophages within the mouse splenocyte population. Quantitative RT-PCR, antibody blocking assays using a specific anti-mTLR2 antibody, and stimulatory experiments in vitro using splenocytes from TLR2-KO mice demonstrated that TFPR1 activated murine immune cells, primarily by stimulating toll-like receptor 2 (TLR2). These results suggest that TFPR1 acts as a novel immune modulator and potent adjuvant primarily by activating TLR2. Thus, the PR-1-based core domain might play a role in immune regulation.
Collapse
Affiliation(s)
- Weilai Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Qiao Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; Graduate School, Anhui Medical University, Hefei 230032, China
| | - Xiuzhe Ning
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yi Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Jingjing Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Qing Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yan Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Hao Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yuepeng Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; Graduate School, Anhui Medical University, Hefei 230032, China
| | - Yusen Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; Graduate School, Anhui Medical University, Hefei 230032, China
| | - Zhihua Kou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; Graduate School, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
23
|
Chen X, Chen X, Gao J, Yang H, Duan Y, Feng Y, He X, Gong X, Wang H, Wu X, Chang J. Astragaloside III Enhances Anti-Tumor Response of NK Cells by Elevating NKG2D and IFN-γ. Front Pharmacol 2019; 10:898. [PMID: 31456687 PMCID: PMC6701288 DOI: 10.3389/fphar.2019.00898] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 07/16/2019] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells play an irreplaceable role in the development of colon cancer, in which antitumor function of NK cells was impaired. Astragaloside III is a natural compound from Astragalus that has been shown to have immunomodulatory effects in various systems. However, few studies have evaluated the antitumor effects of Astragaloside III through stimulating systemic immunity and regulating NK cells. In this study, flow cytometry, immunohistochemical analysis, and immunofunctional assays were performed to elucidate the functions of Astragaloside III in restoring antitumor function of NK cells. We demonstrated that Astragaloside III significantly elevated the expression of natural killer group 2D (NKG2D), Fas, and interferon-γ (IFN-γ) production in NK cells, leading to increased tumor-killing ability. Experiments in cell co-culture assays and CT26-bearing mice model further confirmed that Astragaloside III could effectively impede tumor growth by increasing infiltration of NK cells into tumor and upregulating the antitumor response of NK cells. We further revealed that Astragaloside III increased IFN-γ secretion of NK cells by enhancing the expression of transcription factor T-bet. In conclusion, the effective anti-tumor function of Astragaloside III was achieved through up-regulation of the immune response of NK cells and elevation of NKG2D, Fas, and IFN-γ production.
Collapse
Affiliation(s)
- Xingmeng Chen
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Xi Chen
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junxiao Gao
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Han Yang
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Yue Duan
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Yuxin Feng
- Tianjin Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xin He
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaoqun Gong
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Hanjie Wang
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Xiaoli Wu
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| | - Jin Chang
- Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, School of Life Sciences, Tianjin University, Tianjin, China
| |
Collapse
|
24
|
Qiu CC, Atencio AE, Gallucci S. Inhibition of fatty acid metabolism by etomoxir or TOFA suppresses murine dendritic cell activation without affecting viability. Immunopharmacol Immunotoxicol 2019; 41:361-369. [PMID: 31155984 PMCID: PMC10724852 DOI: 10.1080/08923973.2019.1616754] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/24/2019] [Accepted: 05/01/2019] [Indexed: 02/08/2023]
Abstract
Objective: Dendritic cells (DCs) are important players in immunity against pathogens, but overactive DCs have been implicated in autoimmune diseases, like lupus, in which a paucity of targeted therapies remains. Recent research shows that DCs upregulate their immunometabolism when activating. We explored whether modulating fatty acid (FA) metabolism needed for oxidative phosphorylation can affect the activation of two main DC subsets. Material and methods: Sorted murine plasmacytoid DCs (pDCs) and conventional DCs (cDCs), generated in FLT3-L medium, were treated with etomoxir, an inhibitor of FA oxidation, or TOFA, an inhibitor of FA synthesis, then stimulated with TLR9 agonist CpGA. Surface activation markers and viability were analyzed by flow cytometry, cytokine, and chemokine production and were measured by ELISA. Results: Modulation of FA metabolism suppressed the upregulation of costimulatory molecules and the production of proinflammatory cytokine IL-6 and type I Interferon-dependent chemokine CXCL10 by both subsets of DCs, without affecting DC viability, neither of resting DCs or upon activation. Etomoxir inhibited pDCs at lower doses than cDCs, suggesting that pDCs may be more susceptible to FA metabolic modulation. Conclusions: Both cDCs, the primary antigen presenting cell, and pDCs, the primary type I IFN producer, exhibit a suppressed ability to activate but normal viability when their FA metabolism is inhibited by etomoxir or TOFA. Our findings indicate that FA metabolism plays an important role in the activation of both pDCs and cDCs and suggest that its modulation is an exploitable therapeutic target to suppress DC activation in inflammation or autoimmunity.
Collapse
Affiliation(s)
- Connie C Qiu
- a Laboratory of Dendritic Cell Biology, Department of Microbiology & Immunology , Lewis Katz School of Medicine at Temple University , Philadelphia , PA , USA
| | - Atilio E Atencio
- a Laboratory of Dendritic Cell Biology, Department of Microbiology & Immunology , Lewis Katz School of Medicine at Temple University , Philadelphia , PA , USA
| | - Stefania Gallucci
- a Laboratory of Dendritic Cell Biology, Department of Microbiology & Immunology , Lewis Katz School of Medicine at Temple University , Philadelphia , PA , USA
| |
Collapse
|
25
|
Liu ZF, Chen JL, Li WY, Fan MW, Li YH. FimH as a mucosal adjuvant enhances persistent antibody response and protective efficacy of the anti-caries vaccine. Arch Oral Biol 2019; 101:122-129. [PMID: 30927661 DOI: 10.1016/j.archoralbio.2019.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/07/2019] [Accepted: 02/16/2019] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To investigate whether the recombinant FimH-S.T protein could modulate immune response to anti-caries vaccine in vitro and in vivo. DESIGN Recombinant FimH protein derived from Salmonella was constructed and purified. The expression of dendritic cell maturation markers and cytokines release were performed by flow cytometry, Real-time PCR and ELISA. In addition, BALB/c mice were administered with anti-caries PAc vaccine plus FimH-S.T, antibody responses were evaluated by ELISA. Splenocytes of immunized mice were detected for their proliferative ability in response to in vitro retreatment with PAc antigen by flow cytometry. Caries protection against dental caries formation was also investigated. RESULTS The purified FimH-S.T induced phenotypic maturation of DC2.4 by up-regulating the expression of costimulatory molecules and MHC II, provoked the production and secretion of cytokines via TLR4-dependent signaling pathway in vitro. Furthermore, the mice immunized with the mixture of FimH-S.T and PAc significantly enhanced the PAc-specific antibodies in the serum along with saliva and promoted splenocyte proliferation. Our results also confirmed that PAc+FimH-S.T decreased the caries lesions formation which provided high protective efficacy against dental caries. CONCLUSION Our study demonstrates that recombinant FimH-S.T could enhance specific IgA responses and protection of anti-caries vaccine, possessing mucosal adjuvant ability by activating DC2.4 via TLR4 signaling pathway.
Collapse
Affiliation(s)
- Zhong-Fang Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan 430079,China
| | - Jun-Lan Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan 430079,China
| | - Wu-You Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan 430079,China
| | - Ming-Wen Fan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan 430079,China.
| | - Yu-Hong Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan 430079,China.
| |
Collapse
|
26
|
Devarajan P, Swain SL. Original Antigenic Sin: Friend or Foe in Developing a Broadly Cross-Reactive Vaccine to Influenza? Cell Host Microbe 2019; 25:354-355. [DOI: 10.1016/j.chom.2019.02.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
27
|
Yang J, Sakai J, Siddiqui S, Lee RC, Ireland DDC, Verthelyi D, Akkoyunlu M. IL-6 Impairs Vaccine Responses in Neonatal Mice. Front Immunol 2018; 9:3049. [PMID: 30619375 PMCID: PMC6307459 DOI: 10.3389/fimmu.2018.03049] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/10/2018] [Indexed: 12/24/2022] Open
Abstract
The inability of infants to mount proper follicular helper T (TFH) cell response renders this age group susceptible to infectious diseases. Initial instruction of T cells by antigen presenting cells and subsequent differentiation into TFH cells are controlled by T cell receptor signal strength, co-stimulatory molecules and cytokines such as IL-6 and IL-21. In immunized adults, IL-6 promotes TFH development by increasing the expression of CXCR5 and the TFH master transcription factor, B cell lymphoma 6. Underscoring the importance of IL-6 in TFH generation, we found improved antibody responses accompanied by increased TFH cells and decreased follicular regulatory helper T (TFR) cells, a Foxp3 expressing inhibitory CD4+ T cell occupying the germinal center (GC), when a tetanus toxoid conjugated pneumococcal polysaccharide type 14 vaccine was injected in adult mice together with IL-6. Paradoxically, in neonates IL-6 containing PPS14-TT vaccine suppressed the already impaired TFH development and antibody responses in addition to increasing TFR cell population. Supporting the diminished TFH development, we detected lower frequency of phospho-STAT-3+ TFH in immunized neonatal T cells after IL-6 stimulation than adult cells. Moreover, IL-6 induced more phospho-STAT-3+ TFR in neonatal cells than adult cells. We also measured lower expression of IL-6R on TFH cells and higher expression on TFR cells in neonatal cells than adult cells, a possible explanation for the difference in IL-6 induced signaling in different age groups. Supporting the flow cytometry findings, microscopic examination revealed the localization of Treg cells in the splenic interfollicular niches of immunized adult mice compared to splenic follicles in neonatal mice. In addition to the limitations in the formation of IL-21 producing TFH cells, neonatal mice GC B cells also expressed lower levels of IL-21R in comparison to the adult mice cells. These findings point to diminished IL-6 activity on neonatal TFH cells as an underlying mechanism of the increased TFR: TFH ratio in immunized neonatal mice.
Collapse
Affiliation(s)
- Jiyeon Yang
- Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Jiro Sakai
- Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Shafiuddin Siddiqui
- Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Robert C Lee
- Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Derek D C Ireland
- Office of Biotechnology Products, Division of Biotechnology Review and Research III, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Daniela Verthelyi
- Office of Biotechnology Products, Division of Biotechnology Review and Research III, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Mustafa Akkoyunlu
- Division of Bacterial Allergenic and Parasitic Diseases, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
28
|
Carabelli J, Prato CA, Sanmarco LM, Aoki MP, Campetella O, Tribulatti MV. Interleukin-6 signalling mediates Galectin-8 co-stimulatory activity of antigen-specific CD4 T-cell response. Immunology 2018; 155:379-386. [PMID: 29972692 DOI: 10.1111/imm.12980] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/31/2018] [Accepted: 06/22/2018] [Indexed: 12/21/2022] Open
Abstract
Galectin-8 (Gal-8) is a mammalian lectin endowed with the ability to co-stimulate antigen-specific immune responses. We have previously demonstrated that bone-marrow-derived dendritic cells produce high levels of interleukin-6 (IL-6) in response to Gal-8 stimulation. As IL-6 is a pleiotropic cytokine that has a broad effect on cells of the immune system, we aimed to elucidate whether IL-6 was involved in Gal-8-dependent co-stimulatory signals during antigen recognition by specific CD4 T cells. With this aim, splenocytes from DO11.10 mice were incubated with a low dose of the cognate ovalbumin peptide in combination with Gal-8. Interleukin-6 was found significantly increased in cultures stimulated with Gal-8 alone or Gal-8 plus cognate peptide. Moreover, IL-6 signalling was triggered during Gal-8-induced co-stimulation, as determined by phosphorylation of signal transducer and activator of transcription 3. Interleukin-6 blockade by neutralizing monoclonal antibody precluded Gal-8 co-stimulatory activity but did not affect the antigen-specific T-cell receptor activation. Different subsets of dendritic cells, as well as macrophages and B cells, were identified as the cellular source of IL-6 during Gal-8-induced co-stimulation. To confirm that IL-6 mediated the Gal-8 co-stimulatory effect, antigen-presenting cells from IL-6-deficient or wild-type mice were co-cultured with purified CD4 T cells from OTII mice in the presence of cognate peptide and Gal-8. Notably, Gal-8-induced co-stimulation, but not the antigen-specific response, was significantly impaired in the presence of IL-6-deficient antigen-presenting cells. In addition, exogenous IL-6 fully restored Gal-8-induced co-stimulation. Taken together, our results demonstrate that IL-6 signalling mediates the Gal-8 immune-stimulatory effect.
Collapse
Affiliation(s)
- Julieta Carabelli
- Laboratorio de Inmunología Molecular, Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| | - Cecilia A Prato
- Laboratorio de Inmunología Molecular, Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| | - Liliana M Sanmarco
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina
| | - Maria P Aoki
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina
| | - Oscar Campetella
- Laboratorio de Inmunología Molecular, Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| | - María V Tribulatti
- Laboratorio de Inmunología Molecular, Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| |
Collapse
|
29
|
Harnessing T Follicular Helper Cell Responses for HIV Vaccine Development. Viruses 2018; 10:v10060336. [PMID: 29921828 PMCID: PMC6024737 DOI: 10.3390/v10060336] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 06/15/2018] [Accepted: 06/16/2018] [Indexed: 12/12/2022] Open
Abstract
Passive administration of broadly neutralizing antibodies (bNAbs) capable of recognizing a broad range of viral strains to non-human primates has led to protection from infection with chimeric SIV/HIV virus (SHIV). This data suggests that generating protective antibody responses could be an effective strategy for an HIV vaccine. However, classic vaccine approaches have failed so far to induce such protective antibodies in HIV vaccine trials. HIV-specific bNAbs identified in natural infection show high levels of somatic hypermutations, demonstrating that they underwent extensive affinity maturation. It is likely that to gain ability to recognize diverse viral strains, vaccine-induced humoral responses will also require complex, iterative maturation. T follicular helper cells (Tfh) are a specialized CD4+ T cell subset that provides help to B cells in the germinal center for the generation of high-affinity and long-lasting humoral responses. It is therefore probable that the quality and quantity of Tfh responses upon vaccination will impact development of bNAbs. Here, we review studies that advanced our understanding of Tfh differentiation, function and regulation. We discuss correlates of Tfh responses and bNAb development in natural HIV infection. Finally, we highlight recent strategies to optimize Tfh responses upon vaccination and their impact on prophylactic HIV vaccine research.
Collapse
|
30
|
Zhang ZH, Jiang BC, Liu XH, Zhang MX, Li ZSN, Zhu GZ. Interleukin-7 Regulates T Follicular Helper Cell Function in Patients with Chronic Hepatitis C. Viral Immunol 2018; 31:417-425. [PMID: 29672235 DOI: 10.1089/vim.2018.0010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Signaling through interleukin (IL)-7 is essential and required for development, differentiation, proliferation, and homeostasis of T cells. However, the role of IL-7 in regulation of CD4+ T cells in chronic viral infections was not fully elucidated. Thus, the aim of the current study was to investigate the immunomodulatory activity of IL-7 to T follicular helper (Tfh) cells and its contribution to pathogenesis of chronic HCV, hepatitis C virus (HCV) infection. A total of 47 patients with chronic hepatitis C and 19 normal controls were enrolled. Serum IL-7 and proportion of Tfh cells was measured. The regulatory function of IL-7 to Tfh cells was also investigated in CD4+ T cells and CD4+ T/HCVcc-infected Huh7.5 cell cocultured system. Serum IL-7 concentration was significantly downregulated in patients with chronic hepatitis C, and was negatively correlated with HCV RNA level. Tfh frequency and Tfh-associated cytokines (IL-21 and IL-6) were also reduced in chronic HCV-infected patients. Moreover, recombinant IL-7 stimulation elevated proportion of Tfh cells and IL-21/IL-6 secretion in both HCV-specific and nonspecific manners. Furthermore, IL-7-treated CD4+ T cells exhibited elevated antiviral activities without killing infected hepatocytes, which presented as inhibition of HCV RNA, induction of antiviral proteins, and promotion of cytokine production (especially IL-21) in cocultured system. This process might be dependent on IL-6 secretion. The current data revealed that IL-7 regulated HCV-specific and nonspecific activated Tfh cells, which might contribute to viral clearance. IL-7 could be a potential therapeutic agent for the treatment of chronic hepatitis C.
Collapse
Affiliation(s)
- Zhi-Hong Zhang
- 1 Department of Clinical Laboratory Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine , Changchun, Jilin Province, China
| | - Ben-Chun Jiang
- 1 Department of Clinical Laboratory Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine , Changchun, Jilin Province, China
| | - Xiao-Hong Liu
- 2 The Geriatric Department, The First Bethune Hospital of Jilin University , Changchun, Jilin Province, China
| | - Meng-Xuan Zhang
- 3 Clinical Medicine College, Changchun University of Chinese Medicine , Changchun, Jilin Province, China
| | - Zhen-Sheng-Nan Li
- 4 Clinical Medicine College, Jilin University , Changchun, Jilin Province, China
| | - Guang-Ze Zhu
- 1 Department of Clinical Laboratory Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine , Changchun, Jilin Province, China
| |
Collapse
|
31
|
Devarajan P, Jones MC, Kugler-Umana O, Vong AM, Xia J, Swain SL. Pathogen Recognition by CD4 Effectors Drives Key Effector and Most Memory Cell Generation Against Respiratory Virus. Front Immunol 2018; 9:596. [PMID: 29632538 PMCID: PMC5879149 DOI: 10.3389/fimmu.2018.00596] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/09/2018] [Indexed: 01/14/2023] Open
Abstract
Although much is known about the mechanisms by which pathogen recognition drives the initiation of T cell responses, including those to respiratory viruses, the role of pathogen recognition in fate decisions of T cells once they have become effectors remains poorly defined. Here, we review our recent studies that suggest that the generation of CD4 T cell memory is determined by recognition of virus at an effector “checkpoint.” We propose this is also true of more highly differentiated tissue-restricted effector cells, including cytotoxic “ThCTL” in the site of infection and TFH in secondary lymphoid organs. We point out that ThCTL are key contributors to direct viral clearance and TFH to effective Ab response, suggesting that the most protective immunity to influenza, and by analogy to other respiratory viruses, requires prolonged exposure to antigen and to infection-associated signals. We point out that many vaccines used today do not provide such prolonged signals and suggest this contributes to their limited effectiveness. We also discuss how aging impacts effective CD4 T cell responses and how new insights about the response of aged naive CD4 T cells and B cells might hold implications for effective vaccine design for both the young and aged against respiratory viruses.
Collapse
Affiliation(s)
- Priyadharshini Devarajan
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Michael C Jones
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Olivia Kugler-Umana
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Allen M Vong
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Jingya Xia
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Susan L Swain
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
32
|
Swain SL, Kugler-Umana O, Kuang Y, Zhang W. The properties of the unique age-associated B cell subset reveal a shift in strategy of immune response with age. Cell Immunol 2017; 321:52-60. [PMID: 28712455 DOI: 10.1016/j.cellimm.2017.05.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/04/2017] [Accepted: 05/10/2017] [Indexed: 01/29/2023]
Abstract
In aged mice, conventional naive B cells decrease and a new population of age-associated B cells (ABC)3 develops. When aged unprimed mice are infected with influenza virus, there is a reduced generation of helper CD4 T cell subsets and germinal center B cells, leading to limited production of IgG Ab and less generation of conventional long-lived plasma cells, compared to young. However, we find an enhanced non-follicular (GL7-) ABC response that is helper T cell-independent, but requires high viral dose and pathogen recognition pathways. The infection-induced ABC (iABC) include IAV-specific Ab-secreting cells, some of which relocate to the bone marrow and lung, and persist for >4wk., suggesting they may provide significant protection. We also speculate there is a shift with increased age to dependence on TLR-mediated pathogen-recognition in both B and CD4 T cell responses.
Collapse
Affiliation(s)
- Susan L Swain
- Department of Pathology, University of Massachusetts Medical School, United States.
| | - Olivia Kugler-Umana
- Department of Pathology, University of Massachusetts Medical School, United States
| | - Yi Kuang
- Department of Pathology, University of Massachusetts Medical School, United States
| | - Wenliang Zhang
- Department of Pathology, University of Massachusetts Medical School, United States
| |
Collapse
|