1
|
Liu X, Yang X, Tao L, Li X, Chen G, Liu Q. Nano/Micro-Enabled Modification and Innovation of Conventional Adjuvants for Next-Generation Vaccines. J Funct Biomater 2025; 16:185. [PMID: 40422849 DOI: 10.3390/jfb16050185] [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: 04/06/2025] [Revised: 05/09/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
The global spread of infectious diseases has raised public awareness of vaccines, highlighting their essential role in protecting public health. Among the components of modern vaccines, adjuvants have received increasing attention for boosting immune responses and enhancing efficacy. Recent advancements in adjuvant research, particularly nanodelivery systems, have paved the way for developing more effective and safer adjuvants. This review outlines the properties, progress, and mechanisms of FDA-approved conventional adjuvants, focusing on their contributions to and challenges in vaccine success. Despite these advancements, conventional adjuvants still face suboptimal immunomodulatory effects, potential side effects, and limitations in targeting specific immune pathways. Nanodelivery systems have emerged as a transformative approach in adjuvant design, offering unique advantages such as enhancing vaccine stability, enabling controlled antigen release, and inducing specific immune responses. By addressing these limitations, nanocarriers improve the safety and efficacy of conventional adjuvants and drive the development of next-generation adjuvants for complex diseases. This review also explores strategies for incorporating nanodelivery systems into adjuvant development, emphasizing its role in optimizing vaccine formulations. By summarizing current challenges and recent advances, this review aims to provide valuable insights guiding future efforts in designing innovative adjuvants that meet the evolving needs of global immunization programs.
Collapse
Affiliation(s)
- Xingchi Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Xu Yang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Lu Tao
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Xuanchen Li
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Guoqiang Chen
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Qi Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| |
Collapse
|
2
|
Gomes SMR, Ribeiro-Alves M, Ribeiro RSDA, Brito ACDS, Lisboa VDC, de Azevedo SG, Nogueira JDS, Castilho LDR, Pôrto LCS, da Silva SAG, Dutra PML, Manfro WFP, Rodrigues LS. Evaluation of humoral and cellular immune responses in healthcare workers with varying levels of SARS-CoV-2 exposure: effects of CoronaVac vaccination followed by heterologous booster. Front Immunol 2025; 16:1576430. [PMID: 40406109 PMCID: PMC12095306 DOI: 10.3389/fimmu.2025.1576430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/16/2025] [Indexed: 05/26/2025] Open
Abstract
Background The COVID-19 pandemic demanded diverse vaccination strategies, and there is significant interest in their effectiveness in generating a robust immune response. In Brazil, the use of CoronaVac was crucial in reducing mortality; however, heterologous booster doses were necessary to enhance memory immune response. This study aimed to evaluate the humoral and cellular immunity in healthcare workers who were vaccinated with a complete regimen of CoronaVac and subsequently received heterologous booster doses over nearly one year. Methods A longitudinal study recruited healthcare professionals with varying levels of exposure to SARS-CoV-2 from the Health Complex of the Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil. Blood samples were collected at five time points, including baseline and after vaccination with CoronaVac and heterologous booster doses (ChAdOx1 nCov-19 or BNT162b2). The Th1/Th2/Th17 cytokine production was measured by Flow Cytometry, using whole blood samples stimulated or not with the SARS-CoV-2 Spike protein. In parallel, serum levels of IgG antibodies against Spike (anti-S) and Nucleocapsid (anti-N) proteins were assessed using an immunoassay. Adjustments were made for confounding factors, including age, sex, level of SARS-CoV-2 exposure, and COVID-19 infection status. Results Our results demonstrate that CoronaVac induced high anti-S IgG levels at all evaluated time points (P<0.01). Cytokine analysis revealed a sustained production of antigen-specific Th1 cytokines, including IL-2 (P<0.01) and IFN-γ (P<0.05) regardless of level of SARS-CoV-2 exposure or previous COVID-19 infection at any point during the study. Additionally, we identified six moderate to strong positive correlations (P<0.0001): IL-10 and IFN-γ (ρ=0.77), IL-6 and TNF (ρ=0.77), IL-2 and IFN-γ (ρ=0.71), IL-6 and IL-10 (ρ=0.66), anti-N IgG and anti-S IgG (ρ=0.62), and IL-2 and anti-S IgG (ρ=0.62). Conclusion The CoronaVac elicited an antigen-specific cellular immune response, characterized by enhancing the production of key cytokines such as IFN-γ and IL-2, with high levels of anti-S IgG. Furthermore, the administration of heterologous boosters significantly enhanced these immune responses, demonstrating induced-specific immunological response. These findings underscore the importance of primary vaccination and boosters in inducing immune protection against COVID-19, potentially informing future vaccination policies and approaches.
Collapse
Affiliation(s)
- Shayane Martins Rodrigues Gomes
- Discipline of Parasitology, Department of Microbiology, Immunology and Parasitology, Medical Science Faculty, Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Marcelo Ribeiro-Alves
- Laboratory of Clinical Research on ISTs/AIDS, National Institute of Infectology Evandro Chagas, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | | | - Andréia Carolinne de Souza Brito
- Discipline of Parasitology, Department of Microbiology, Immunology and Parasitology, Medical Science Faculty, Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Vinicius da Cunha Lisboa
- Laboratory of Immunopathology (LIP), Discipline of General Pathology, Medical Science Faculty, UERJ, Rio de Janeiro, Brazil
| | - Samara Galdino de Azevedo
- Laboratory of Immunopathology (LIP), Discipline of General Pathology, Medical Science Faculty, UERJ, Rio de Janeiro, Brazil
| | - Jeane de Souza Nogueira
- Laboratory of Histocompatibility and Cryopreservation, Tissue Repair and Histocompatibility Technologic Core, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Leda dos Reis Castilho
- Cell Culture Engineering Lab, Chemical Engineering Program, Coordination of Graduate Engineering Programs, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Luís Cristóvão Sobrino Pôrto
- Laboratory of Histocompatibility and Cryopreservation, Tissue Repair and Histocompatibility Technologic Core, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Silvia Amaral Gonçalves da Silva
- Discipline of Parasitology, Department of Microbiology, Immunology and Parasitology, Medical Science Faculty, Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Patrícia Maria Lourenço Dutra
- Discipline of Parasitology, Department of Microbiology, Immunology and Parasitology, Medical Science Faculty, Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Wânia Ferraz Pereira Manfro
- Discipline of Microbiology and Immunology, Department of Microbiology, Immunology and Parasitology, Medical Science Faculty, UERJ, Rio de Janeiro, Brazil
| | - Luciana Silva Rodrigues
- Laboratory of Immunopathology (LIP), Discipline of General Pathology, Medical Science Faculty, UERJ, Rio de Janeiro, Brazil
| |
Collapse
|
3
|
Pal PB, Iyer SS. Transient pain and long-term gain: adjuvant dose directs immune memory. J Clin Invest 2025; 135:e190524. [PMID: 40231461 PMCID: PMC11996847 DOI: 10.1172/jci190524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025] Open
Abstract
Vaccine hesitancy is often fueled by fears of side effects; however, most reactions result from innate immune activation and cytokine production, which are required for lasting immunity. For effective vaccines against HIV, innate activation is essential for differentiation of CD4+ T cells into T follicular helper cells (TFH), which guide rare B cells to mature into long-lived plasma cells that produce durable neutralizing antibodies (nAbs). In this issue of the JCI, Parham Ramezani-Rad et al. show that higher doses of saponin QS-21-MPLA nanoparticle (SMNP) adjuvant, combined with BG505 MD39 envelope (Env) protein, enhanced cytokine responses, drove stronger Env-specific TFH responses in blood, and increased Env-specific bone marrow plasma cells compared with lower doses. While tier 2 nAbs were sustained at memory in only a subset of animals, predominantly at the highest adjuvant dose, these findings highlight transient reactogenicity as an essential mechanism - not a flaw - for building durable immune memory.
Collapse
Affiliation(s)
- Pabitra B. Pal
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Smita S. Iyer
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- California National Primate Research Center, UCD, Davis, California, USA
| |
Collapse
|
4
|
Warrick KA, Vallez CN, Meibers HE, Pasare C. Bidirectional Communication Between the Innate and Adaptive Immune Systems. Annu Rev Immunol 2025; 43:489-514. [PMID: 40279312 PMCID: PMC12120936 DOI: 10.1146/annurev-immunol-083122-040624] [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] [Indexed: 04/27/2025]
Abstract
Effective bidirectional communication between the innate and adaptive immune systems is crucial for tissue homeostasis and protective immunity against infections. The innate immune system is responsible for the early sensing of and initial response to threats, including microbial ligands, toxins, and tissue damage. Pathogen-related information, detected primarily by the innate immune system via dendritic cells, is relayed to adaptive immune cells, leading to the priming and differentiation of naive T cells into effector and memory lineages. Memory T cells that persist long after pathogen clearance are integral for durable protective immunity. In addition to rapidly responding to reinfections, memory T cells also directly instruct the interacting myeloid cells to induce innate inflammation, which resembles microbial inflammation. As such, memory T cells act as newly emerging activators of the innate immune system and function independently of direct microbial recognition. While T cell-mediated activation of the innate immune system likely evolved as a protective mechanism to combat reinfections by virulent pathogens, the detrimental outcomes of this mechanism manifest in the forms of autoimmunity and other T cell-driven pathologies. Here, we review the complexities and layers of regulation at the interface between the innate and adaptive immune systems to highlight the implications of adaptive instruction of innate immunity in health and disease.
Collapse
Affiliation(s)
- Kathrynne A Warrick
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Charles N Vallez
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Hannah E Meibers
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Chandrashekhar Pasare
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| |
Collapse
|
5
|
Ludwig MP, Wilson JR, Galbraith MD, Bhandari N, Dunn LN, Black JC, Sullivan KD. NF-κB signaling directs a program of transient amplifications at innate immune response genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.641929. [PMID: 40161744 PMCID: PMC11952383 DOI: 10.1101/2025.03.11.641929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The cellular response to pathogens involves an intricate response directed by key innate immune signaling pathways which is characterized by cell-to-cell heterogeneity. How this heterogeneity is established and regulated remains unclear. We describe a program of transient site-specific gains (TSSG) producing extrachromosomal DNA (ecDNA) of immune-related genes in response to innate immune signaling. Activation of NF-κB drives TSSG of the interferon receptor gene cluster through inducible recruitment of the transcription factor RelA and the pre-replication complex member MCM2 to an epigenetically regulated TSSG control element. Targeted recruitment of RelA or p300 are sufficient to induce TSSG formation. RelA and MCM2 specify a program of TSSG for at least six and as many as 179 regions enriched in innate immune response genes. Identification of this program reveals regulated production of ecDNA as a mechanism of heterogeneity in the host response.
Collapse
Affiliation(s)
- Michael P. Ludwig
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- These Authors Contributed Equally
| | - Jason R. Wilson
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- These Authors Contributed Equally
| | - Matthew D. Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
| | - Nirajan Bhandari
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lauren N. Dunn
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Joshua C. Black
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kelly D. Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Lead Contact
| |
Collapse
|
6
|
Kim EH, Wahl K, Guelfi E, Lee D. Engineering the physical characteristics of biomaterials for innate immune-mediated cancer immunotherapy. J Control Release 2025; 378:814-830. [PMID: 39719214 DOI: 10.1016/j.jconrel.2024.12.046] [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: 07/04/2024] [Revised: 12/07/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024]
Abstract
It has recently been recognized that the physical characteristics of biomaterials - such as size, structure, shape, charge, mechanical strength, hydrophobicity, and multivalency - regulate immunological functions in innate immune cells. In immuno-oncology applications, biomaterials are engineered with distinct physical properties to achieve desired innate immune responses. In this review, we discuss how physical characteristics influence effector functions and innate immune signaling pathways in distinct innate immune cell subtypes. We highlight how physical properties of biomaterials impact phagocytosis regulation, biodistribution, and innate immune cell targeting. We outline the recent advances in physical engineering of biomaterials that directly or indirectly induce desired innate immune responses for cancer immunotherapy. Lastly, we discuss the challenges in current biomaterial approaches that need to be addressed to improve clinical applicability.
Collapse
Affiliation(s)
- Eun-Hye Kim
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA; Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
| | - Katelyn Wahl
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA; Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
| | - Erica Guelfi
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA; Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
| | - DaeYong Lee
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA; Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
7
|
Wang J, Tao X, Liu Z, Yan Y, Cheng P, Liu B, Du H, Niu B. Noncoding RNAs in sepsis-associated acute liver injury: Roles, mechanisms, and therapeutic applications. Pharmacol Res 2025; 212:107596. [PMID: 39800175 DOI: 10.1016/j.phrs.2025.107596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/11/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Sepsis is a life-threatening syndrome characterized by organ dysfunction caused by a dysregulated host response to infection. Sepsis-associated acute liver injury (SA-ALI) is a frequent and serious complication of sepsis that considerably impacts both short-term and long-term survival outcomes. In intensive care units (ICUs), the mortality rate of patients with SA-ALI remains high, mostly due to the absence of effective early diagnostic markers and suitable therapeutic strategies. Recent studies have demonstrated the importance of non-coding RNAs (ncRNAs) in the development and progression of SA-ALI. This review focuses on the critical roles of ncRNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), in regulating "cytokine storms", oxidative stress, mitochondrial dysfunction, and programmed cell death in SA-ALI, and summarizes the current state and limitations of existing studies on lncRNAs and circRNAs in SA-ALI. By integrating advancements in high-throughput sequencing technologies, this review provides novel insights into the dual potential of ncRNAs as diagnostic biomarkers and therapeutic targets, offers new ideas for SA-ALI diagnosis and treatment research and highlights potential challenges in clinical translation.
Collapse
Affiliation(s)
- Jialian Wang
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China
| | - Xingyu Tao
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China
| | - Zhengyang Liu
- Department of Nephrology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing 400016, China
| | - Yuan Yan
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China
| | - Peifeng Cheng
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China
| | - Bin Liu
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China
| | - Huimin Du
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Bailin Niu
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China.
| |
Collapse
|
8
|
Ronchese F, Webb GR, Ochiai S, Lamiable O, Brewerton M. How type-2 dendritic cells induce Th2 differentiation: Instruction, repression, or fostering T cell-T cell communication? Allergy 2025; 80:395-407. [PMID: 39324367 PMCID: PMC11804308 DOI: 10.1111/all.16337] [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: 04/30/2024] [Revised: 09/03/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
Allergic disease is caused by the activation of allergen-specific CD4+ type-2 T follicular helper cells (Tfh2) and T helper 2 (Th2) effector cells that secrete the cytokines IL-4, IL-5, IL-9, and IL-13 upon allergen encounter, thereby inducing IgE production by B cells and tissue inflammation. While it is accepted that the priming and differentiation of naïve CD4+ T cells into Th2 requires allergen presentation by type 2 dendritic cells (DC2s), the underlying signals remain unidentified. In this review we focus on the interaction between allergen-presenting DC2s and naïve CD4+ T cells in lymph node (LN), and the potential mechanisms by which DC2s might instruct Th2 differentiation. We outline recent advances in characterizing DC2 development and heterogeneity. We review mechanisms of allergen sensing and current proposed mechanisms of Th2 differentiation, with specific consideration of the role of DC2s and how they might contribute to each mechanism. Finally, we assess recent publications reporting a detailed analysis of DC-T cell interactions in LNs and how they support Th2 differentiation. Together, these studies are starting to shape our understanding of this key initial step of the allergic immune response.
Collapse
Affiliation(s)
| | - Greta R. Webb
- Malaghan Institute of Medical ResearchWellingtonNew Zealand
| | - Sotaro Ochiai
- Malaghan Institute of Medical ResearchWellingtonNew Zealand
| | | | - Maia Brewerton
- Malaghan Institute of Medical ResearchWellingtonNew Zealand
- Department of Clinical Immunology and AllergyAuckland City HospitalAucklandNew Zealand
| |
Collapse
|
9
|
Zhang C, Li F, Yu X, Tian H, Li Y, Liu X, Liu W, Yu B, Qiao ZA, Yu X. Periodic mesoporous organosilica-loaded mincle agonists enhance the immunogenicity of COVID-19 subunit vaccines by dual activation of B cells and dendritic cells. Acta Biomater 2025; 193:362-376. [PMID: 39730102 DOI: 10.1016/j.actbio.2024.12.056] [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: 09/26/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Effective vaccination is crucial for intervening in the COVID-19 pandemic. However, with the continuous mutation of the SARS-CoV-2, existing vaccines including subunit vaccines cannot effectively prevent virus infections. Hence, there is an urgent need to enhance the immunogenicity of existing vaccines to induce a more potent and durable immune response. We previously found that periodic mesoporous organosilica (PMO) could act as a potential nanoadjuvant for subunit vaccines, eliciting potent antigen-specific germinal center (GC) responses by activating naïve B cells. In this study, we describe the design of PMO decorated with TDB, a potent Macrophage-induced C-type lectin (Mincle) agonist, to improve the adjuvanticity of PMO for COVID-19 vaccines. We found that the TDB@PMO adjuvant can effectively deliver antigens to lymph nodes and promote antigen uptake by immune cells. More importantly, the TDB@PMO adjuvant vaccine could activate the innate immune of both naïve B cells and dendritic cells via the Mincle signaling pathway, and further enhance the GC responses and resulting in potent SARS-CoV-2 specific humoral and cellular immune responses. Overall, we have developed an effective and safe nanoadjuvant platform, laying the foundation for the design and development of subunit vaccines against pathogens such as SARS-CoV-2. STATEMENT OF SIGNIFICANCE: Adjuvants play a crucial role in enhancing the effectiveness of vaccines by boosting the immune response. The emergence of highly mutated viruses, such as coronaviruses, has presented new requirements for adjuvant design. This work designed a nanoadjuvant platform, TDB@PMO, to enhance the immune response of the COVID-19 subunit vaccine. The result demonstrated that TDB@PMO nanoadjuvant can simultaneously boost the activation effects of B cells and DC cells through the Mincle signaling pathway. Furthermore, immunization with TDB@PMO-RBD nanoadjuvanted vaccine in mice significantly enhanced germinal center responses and antibody production, while also eliciting a robust antigen-specific T cell immune response in spleen. This design provided a reference for the development of next-generation virus subunit vaccines.
Collapse
Affiliation(s)
- Chunhe Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Fangshen Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xin Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Haochen Tian
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yiyang Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xinyao Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wenmo Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Zhen-An Qiao
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Jilin University, Changchun, Jilin 130012, China.
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
10
|
Vlasova O, Antonova I, Magomedova K, Osipova A, Shtompel P, Borunova A, Zabotina T, Belitsky G, Budunova I, Jordan A, Kirsanov K, Yakubovskaya M. Anticancer Plant Secondary Metabolites Evicting Linker Histone H1.2 from Chromatin Activate Type I Interferon Signaling. Int J Mol Sci 2025; 26:375. [PMID: 39796235 PMCID: PMC11722331 DOI: 10.3390/ijms26010375] [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: 10/18/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 01/13/2025] Open
Abstract
Previously we discovered that among 15 DNA-binding plant secondary metabolites (PSMs) possessing anticancer activity, 11 compounds cause depletion of the chromatin-bound linker histones H1.2 and/or H1.4. Chromatin remodeling or multiH1 knocking-down is known to promote the upregulation of repetitive elements, ultimately triggering an interferon (IFN) response. Herein, using HeLa cells and applying fluorescent reporter assay with flow cytometry, immunofluorescence staining and quantitative RT-PCR, we studied effects of PSMs both evicting linker histones from chromatin and not influencing their location in nucleus. We found that (1) 8 PSMs, evicting linker histone H1.2 from chromatin, activated significantly the type I IFN signaling pathway and out of these compounds resveratrol, berberine, genistein, delphinidin, naringenin and curcumin also caused LINE1 expression. Fisetin and quercetin, which also induced linker histone H1.2 eviction from chromatin, significantly activated only type I IFN signaling, but not LINE1 expression; (2) curcumin, sanguinarine and kaempferol, causing significant depletion of the chromatin-bound linker histone H1.4 but not significantly influencing H1.2 presence in chromatin, activate type I IFN signaling less intensively without any changes in LINE1 expression; (3) four PSMs, which did not cause linker histone eviction, displayed neither IFN signaling activation nor enhancement of LINE1 expression. Thus, we have shown for the first time that chromatin destabilization observed by depletion of chromatin-bound linker histone H1.2 caused by anticancer DNA-binding PSMs is accompanied by enhancement of type I IFN signaling, and that LINE1 expression often impacts this activation.
Collapse
Affiliation(s)
- Olga Vlasova
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoe Shosse, 115522 Moscow, Russia (K.M.)
| | - Irina Antonova
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoe Shosse, 115522 Moscow, Russia (K.M.)
| | - Khamis Magomedova
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoe Shosse, 115522 Moscow, Russia (K.M.)
| | - Alena Osipova
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoe Shosse, 115522 Moscow, Russia (K.M.)
- SBHI Moscow Clinical Scientific Center Named After Loginov MHD, 111123 Moscow, Russia
| | - Polina Shtompel
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoe Shosse, 115522 Moscow, Russia (K.M.)
| | - Anna Borunova
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoe Shosse, 115522 Moscow, Russia (K.M.)
| | - Tatiana Zabotina
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoe Shosse, 115522 Moscow, Russia (K.M.)
| | - Gennady Belitsky
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoe Shosse, 115522 Moscow, Russia (K.M.)
| | - Irina Budunova
- Department of Dermatology, Northwestern University, Chicago, IL 60611, USA
| | - Albert Jordan
- Institut de Biologia Molecular de Barcelona (IBMB-CSIC), 08028 Barcelona, Spain
| | - Kirill Kirsanov
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoe Shosse, 115522 Moscow, Russia (K.M.)
- Institute of Medicine, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklaya St. 6, 117198 Moscow, Russia
| | - Marianna Yakubovskaya
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoe Shosse, 115522 Moscow, Russia (K.M.)
| |
Collapse
|
11
|
Liang J, Yao L, Liu Z, Chen Y, Lin Y, Tian T. Nanoparticles in Subunit Vaccines: Immunological Foundations, Categories, and Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407649. [PMID: 39501996 DOI: 10.1002/smll.202407649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/12/2024] [Indexed: 01/11/2025]
Abstract
Subunit vaccines, significant in next-generation vaccine development, offer precise targeting of immune responses by focusing on specific antigens. However, this precision often comes at the cost of eliciting strong and durable immunity, posing a great challenge to vaccine design. To address this limitation, recent advancements in nanoparticles (NPs) are utilized to enhance antigen delivery efficiency and boost vaccine efficacy. This review examines how the physicochemical properties of NPs influence various stages of the immune response during vaccine delivery and analyzes how different NP types contribute to immune activation and enhance vaccine performance. It then explores the unique characteristics and immune activation mechanisms of these NPs, along with their recent advancements, and highlights their application in subunit vaccines targeting infectious diseases and cancer. Finally, it discusses the challenges in NP-based vaccine development and proposes future directions for innovation in this promising field.
Collapse
Affiliation(s)
- Jiale Liang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lan Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhiqiang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ye Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Taoran Tian
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China
| |
Collapse
|
12
|
Jia H, Zhang L. tRNA-derived small RNAs in disease immunity. Theranostics 2025; 15:245-257. [PMID: 39744232 PMCID: PMC11667222 DOI: 10.7150/thno.102650] [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: 08/22/2024] [Accepted: 11/08/2024] [Indexed: 01/11/2025] Open
Abstract
Recently, members of a unique species of non-coding RNA, known as transfer RNA-derived small RNAs (tsRNAs) have been reported to serve multiple molecular functions, including in cells that mediate immunity. Because of their low molecular weights, tsRNAs were previously difficult to detect and were thus overlooked, until now. In this review, we delve into the biogenesis of tsRNAs and their diverse biological functions, ranging from transcriptional regulation to modulation of mRNA translation. We highlight the current evidence demonstrating their involvement in the immune response, as well as how tsRNAs modulate immunity to influence tumor growth and spread, autoimmune disease pathology and infection by pathogens. We surmise that tsRNAs are likely informative as diagnostic markers of cellular homeostasis and disease, and that therapeutic targeting of tsRNAs could be beneficial for a range of human diseases. Improved knowledge on the functions for tsRNAs in the mammalian immune system will enable us to leverage tsRNAs for their effective clinical use as treatments for human health challenges.
Collapse
Affiliation(s)
- Hongyuan Jia
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Linling Zhang
- Department of Respiratory and Critical Care, Chengdu Third People's Hospital, Chengdu, China
| |
Collapse
|
13
|
Ying X, Chen Q, Yang Y, Wu Z, Zeng W, Miao C, Huang Q, Ai K. Nanomedicines harnessing cGAS-STING pathway: sparking immune revitalization to transform 'cold' tumors into 'hot' tumors. Mol Cancer 2024; 23:277. [PMID: 39710707 DOI: 10.1186/s12943-024-02186-6] [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: 10/11/2024] [Accepted: 11/26/2024] [Indexed: 12/24/2024] Open
Abstract
cGAS-STING pathway stands at the forefront of innate immunity and plays a critical role in regulating adaptive immune responses, making it as a key orchestrator of anti-tumor immunity. Despite the great potential, clinical outcomes with cGAS-STING activators have been disappointing due to their unfavorable in vivo fate, signaling an urgent need for innovative solutions to bridge the gap in clinical translation. Recent advancements in nanotechnology have propelled cGAS-STING-targeting nanomedicines to the cutting-edge of cancer therapy, leveraging precise drug delivery systems and multifunctional platforms to achieve remarkable region-specific biodistribution and potent therapeutic efficacy. In this review, we provide an in-depth exploration of the molecular mechanisms that govern cGAS-STING signaling and its potential to dynamically modulate the anti-tumor immune cycle. We subsequently introduced several investigational cGAS-STING-dependent anti-tumor agents and summarized their clinical trial progress. Additionally, we provided a comprehensive review of the unique advantages of cGAS-STING-targeted nanomedicines, highlighting the transformative potential of nanotechnology in this field. Furthermore, we comprehensively reviewed and comparatively analyzed the latest breakthroughs cGAS-STING-targeting nanomedicine, focusing on strategies that induce cytosolic DNA generation via exogenous DNA delivery, chemotherapy, radiotherapy, or dynamic therapies, as well as the nanodelivery of STING agonists. Lastly, we discuss the future prospects and challenges in cGAS-STING-targeting nanomedicine development, offering new insights to bridge the gap between mechanistic research and drug development, thereby opening new pathways in cancer treatment.
Collapse
Affiliation(s)
- Xiaohong Ying
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Qiaohui Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Yongqi Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Ziyu Wu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Wan Zeng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Chenxi Miao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China.
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Xiangya Hospital, Ministry of Education, Central South University, Changsha, 410008, China.
| |
Collapse
|
14
|
Li Y, Yu J, Zeng Z, Lin W. Regulation of ubiquitination in sepsis: from PAMP versus DAMP to peripheral inflammation and cell death. Front Immunol 2024; 15:1513206. [PMID: 39720715 PMCID: PMC11666442 DOI: 10.3389/fimmu.2024.1513206] [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: 10/18/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Abstract
Sepsis (sepsis) is a systemic inflammatory response triggered by infection, and its pathologic features include overproduction of peripheral inflammatory factors (e.g., IL-1β, IL-6, TNF-α), which ultimately leads to cytokine storm and multiple organ dysfunction syndrome (MODS). Pathogen-associated molecular patterns (PAMP) and damage-associated molecular patterns (DAMP) induce strong immune responses and exacerbate inflammation by activating pattern recognition receptors (PRRs) in the host. Ubiquitination, as a key protein post-translational modification, dynamically regulates the activity of several inflammation-associated proteins (e.g., RIPK1, NLRP3) through the coordinated action of the E1, E2, and E3 enzymes, affects cell death pathways such as necroptosis and pyroptosis, and ultimately regulates the release of peripheral inflammatory factors. Deubiquitinating enzymes (DUBs), on the other hand, influence the intensity of the inflammatory response in sepsis by counter-regulating the ubiquitination process and balancing pro- and anti-inflammatory signals. This review focuses on how PAMP and DAMP activate inflammatory pathways via PRRs, and the central role of ubiquitination and deubiquitination in the development of sepsis, especially the mechanisms in regulating the secretion of peripheral inflammatory factors and cell death. By deeply dissecting the impact of the balance of ubiquitination and deubiquitination on inflammatory regulation, we further envision its potential as a therapeutic target in sepsis.
Collapse
Affiliation(s)
| | | | | | - Weixiong Lin
- Department of Anesthesiology I, Meizhou People’s Hospital,
Meizhou, Guangdong, China
| |
Collapse
|
15
|
Kroemer G, Montégut L, Kepp O, Zitvogel L. The danger theory of immunity revisited. Nat Rev Immunol 2024; 24:912-928. [PMID: 39511426 DOI: 10.1038/s41577-024-01102-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2024] [Indexed: 11/15/2024]
Abstract
The danger theory of immunity, introduced by Polly Matzinger in 1994, posits that tissue stress, damage or infection has a decisive role in determining immune responses. Since then, a growing body of evidence has supported the idea that the capacity to elicit cognate immune responses (immunogenicity) relies on the combination of antigenicity (the ability to be recognized by T cell receptors or antibodies) and adjuvanticity (additional signals arising owing to tissue damage). Here, we discuss the molecular foundations of the danger theory while focusing on immunologically relevant damage-associated molecular patterns, microorganism-associated molecular patterns, and neuroendocrine stress-associated immunomodulatory molecules, as well as on their receptors. We critically evaluate patient-relevant evidence, examining how cancer cells and pathogenic viruses suppress damage-associated molecular patterns to evade immune recognition, how intestinal dysbiosis can reduce immunostimulatory microorganism-associated molecular patterns and compromise immune responses, and which hereditary immune defects support the validity of the danger theory. Furthermore, we incorporate the danger hypothesis into a close-to-fail-safe hierarchy of immunological tolerance mechanisms that also involve the clonal deletion and inactivation of immune cells.
Collapse
Affiliation(s)
- Guido Kroemer
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Léa Montégut
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France.
- INSERM UMR 1015, ClinicObiome, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France.
- Université Paris-Saclay, Ile-de-France, Paris, France.
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS), Villejuif, France.
| |
Collapse
|
16
|
Martin MU, Tay CM, Siew TW. Continuous Treatment with IncobotulinumtoxinA Despite Presence of BoNT/A Neutralizing Antibodies: Immunological Hypothesis and a Case Report. Toxins (Basel) 2024; 16:422. [PMID: 39453199 PMCID: PMC11510976 DOI: 10.3390/toxins16100422] [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: 06/24/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
Botulinum Neurotoxin A (BoNT/A) is a bacterial protein that has proven to be a valuable pharmaceutical in therapeutic indications and aesthetic medicine. One major concern is the formation of neutralizing antibodies (nAbs) to the core BoNT/A protein. These can interfere with the therapy, resulting in partial or complete antibody (Ab)-mediated secondary non-response (SNR) or immunoresistance. If titers of nAbs reach a level high enough that all injected BoNT/A molecules are neutralized, immunoresistance occurs. Studies have shown that continuation of treatment of neurology patients who had developed Ab-mediated partial SNR against complexing protein-containing (CPC-) BoNT/A was in some cases successful if patients were switched to complexing protein-free (CPF-) incobotulinumtoxinA (INCO). This seems to contradict the layperson's basic immunological understanding that repeated injection with the same antigen BoNT/A should lead to an increase in antigen-specific antibody titers. As such, we strive to explain how immunological memory works in general, and based on this, we propose a working hypothesis for this paradoxical phenomenon observed in some, but not all, neurology patients with immunoresistance. A critical factor is the presence of potentially immune-stimulatory components in CPC-BoNT/A products that can act as immunologic adjuvants and activate not only naïve, but also memory B lymphocyte responses. Furthermore, we propose that continuous injection of a BoN/TA formulation with low immunogenicity, e.g., INCO, may be a viable option for aesthetic patients with existing nAbs. These concepts are supported by a real-world case example of a patient with immunoresistance whose nAb levels declined with corresponding resumption of clinical response despite regular INCO injections.
Collapse
Affiliation(s)
| | | | - Tuck Wah Siew
- Radium Medical Aesthetics, 3 Temasek Boulevard #03-325/326/327/328, Suntec City Mall, Singapore 038983, Singapore
| |
Collapse
|
17
|
Marques JT, Meignin C, Imler JL. An evolutionary perspective to innate antiviral immunity in animals. Cell Rep 2024; 43:114678. [PMID: 39196781 DOI: 10.1016/j.celrep.2024.114678] [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: 12/22/2023] [Revised: 06/22/2024] [Accepted: 08/08/2024] [Indexed: 08/30/2024] Open
Abstract
Viruses pose a significant threat to cellular organisms. Innate antiviral immunity encompasses both RNA- and protein-based mechanisms designed to sense and respond to infections, a fundamental aspect present in all living organisms. A potent RNA-based antiviral mechanism is RNA interference, where small RNA-programmed nucleases target viral RNAs. Protein-based mechanisms often rely on the induction of transcriptional responses triggered by the recognition of viral infections through innate immune receptors. These responses involve the upregulation of antiviral genes aimed at countering viral infections. In this review, we delve into recent advances in understanding the diversification of innate antiviral immunity in animals. An evolutionary perspective on the gains and losses of mechanisms in diverse animals coupled to mechanistic studies in model organisms such as the fruit fly Drosophila melanogaster is essential to provide deep understanding of antiviral immunity that can be translated to new strategies in the treatment of viral diseases.
Collapse
Affiliation(s)
- Joao T Marques
- Université de Strasbourg, INSERM U1257, CNRS UPR9022, 67084 Strasbourg, France; Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil.
| | - Carine Meignin
- Université de Strasbourg, CNRS UPR9022, 67084 Strasbourg, France
| | - Jean-Luc Imler
- Université de Strasbourg, CNRS UPR9022, 67084 Strasbourg, France; Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
18
|
Herrero-Fernández B, Ortega-Zapero M, Gómez-Bris R, Sáez A, Iborra S, Zorita V, Quintas A, Vázquez E, Dopazo A, Sánchez-Madrid F, Arribas SM, González-Granado JM. Role of lamin A/C on dendritic cell function in antiviral immunity. Cell Mol Life Sci 2024; 81:400. [PMID: 39264480 PMCID: PMC11393282 DOI: 10.1007/s00018-024-05423-9] [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: 05/13/2024] [Revised: 07/29/2024] [Accepted: 08/23/2024] [Indexed: 09/13/2024]
Abstract
Dendritic cells (DCs) play a crucial role in orchestrating immune responses, particularly in promoting IFNγ-producing-CD8 cytotoxic T lymphocytes (CTLs) and IFNγ-producing-CD4 T helper 1 (Th1) cells, which are essential for defending against viral infections. Additionally, the nuclear envelope protein lamin A/C has been implicated in T cell immunity. Nevertheless, the intricate interplay between innate and adaptive immunity in response to viral infections, particularly the role of lamin A/C in DC functions within this context, remains poorly understood. In this study, we demonstrate that mice lacking lamin A/C in myeloid LysM promoter-expressing cells exhibit a reduced capacity to induce Th1 and CD8 CTL responses, leading to impaired clearance of acute primary Vaccinia virus (VACV) infection. Remarkably, in vitro-generated granulocyte macrophage colony-stimulating factor bone marrow-derived DCs (GM-CSF BMDCs) show high levels of lamin A/C. Lamin A/C absence on GM-CSF BMDCs does not affect the expression of costimulatory molecules on the cell membrane but it reduces the cellular ability to form immunological synapses with naïve CD4 T cells. Lamin A/C deletion induces alterations in NFκB nuclear localization, thereby influencing NF-κB-dependent transcription. Furthermore, lamin A/C ablation modifies the gene accessibility of BMDCs, predisposing these cells to mount a less effective antiviral response upon TLR stimulation. This study highlights the critical role of DCs in interacting with CD4 T cells during antiviral responses and proposes some mechanisms through which lamin A/C may modulate DC function via gene accessibility and transcriptional regulation.
Collapse
Affiliation(s)
- Beatriz Herrero-Fernández
- LamImSys Lab, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, 28041, Spain
- Department of Physiology, Faculty of Medicine, Universidad Autonoma de Madrid, Madrid, 28029, Spain
| | - Marina Ortega-Zapero
- LamImSys Lab, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, 28041, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Raquel Gómez-Bris
- LamImSys Lab, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, 28041, Spain
- Department of Physiology, Faculty of Medicine, Universidad Autonoma de Madrid, Madrid, 28029, Spain
| | - Angela Sáez
- LamImSys Lab, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, 28041, Spain
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria (UFV), Pozuelo de Alarcón, 28223, Spain
| | - Salvador Iborra
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain
- Fundacion Inmunotek, Alcalá de Henares, 28805, Spain
| | - Virginia Zorita
- Centro Nacional de Investigaciones, Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Ana Quintas
- Centro Nacional de Investigaciones, Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Enrique Vázquez
- Centro Nacional de Investigaciones, Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Ana Dopazo
- Centro Nacional de Investigaciones, Cardiovasculares (CNIC), Madrid, 28029, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Centro Nacional de Investigaciones, Cardiovasculares (CNIC), Madrid, 28029, Spain
- Immunology Unit, Medicine Department, Hospital Universitario La Princesa, Universidad Autónoma de Madrid, Instituto Investigacion Sanitaria-Princesa IIS-IP, Madrid, Spain, Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Silvia Magdalena Arribas
- Department of Physiology, Faculty of Medicine, Universidad Autonoma de Madrid, Madrid, 28029, Spain.
| | - Jose Maria González-Granado
- LamImSys Lab, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, 28041, Spain.
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain.
- Centro de Investigacion Biomedica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| |
Collapse
|