1
|
Barik P, Mondal S. Immunomodulatory effects of metal nanoparticles: current trends and future prospects. NANOSCALE 2025; 17:10433-10461. [PMID: 40202489 DOI: 10.1039/d5nr01030f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
The advent of nanotechnology has steered into a new era of medical advancements, with metal nanoparticles (MNPs) emerging as potent agents for precise regulation of the immune system. This review provides a comprehensive overview of the immunomodulatory roles of MNPs, including gold, silver, and metal oxide nanoparticles, in regulating innate and adaptive immunity. Additionally, we discuss the immunological effects of metal ions and metal complexes, offering a comparative analysis with nanoparticulate systems. We analyse cutting-edge strategies utilising MNPs to optimise vaccine efficacy, achieve targeted delivery to immune cells, and orchestrate inflammatory responses. Additionally, we discuss the therapeutic potential of MNPs in combating autoimmune diseases, cancers, and infectious agents, which is evaluated within the framework of precision medicine. Furthermore, we critically assess challenges such as biocompatibility, potential toxicity, and regulatory hurdles. Finally, we propose future directions for integrating MNPs with advanced delivery systems and other nanomaterials to propel the frontiers of immunotherapy. This review aims to provide a foundational understanding of MNP-mediated immunomodulation, inspiring further research and development in this burgeoning field.
Collapse
Affiliation(s)
- Puspendu Barik
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah, 26666, United Arab Emirates
- Department of Physics, College of Arts and Sciences, American University of Sharjah, Sharjah, 26666, United Arab Emirates
| | - Samiran Mondal
- Department of Chemistry, Rammohan College (University of Calcutta), 102/1-Raja Rammohan Sarani, Kolkata 700009, West Bengal, India.
| |
Collapse
|
2
|
Wang Y, Wang H, Xu Y, Ling J, Zong C, Zhang Y, Guo X, Zhao G, Zhou Y, Zhao J, Lou P, Liu X, Xu T, Ma Q. Mefloquine Suppresses Metastasis in Renal Cell Carcinoma Through Targeting SPC25. Cancer Sci 2025; 116:1239-1254. [PMID: 39948743 PMCID: PMC12044673 DOI: 10.1111/cas.70001] [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: 08/16/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 05/02/2025] Open
Abstract
Renal cell carcinoma (RCC) is the third most common malignant tumor in the urinary system, often presenting with distant metastases at diagnosis. Approximately one-quarter of patients undergoing nephrectomy experience distant recurrence. Despite the recent advancements in combination-targeted and immune checkpoint inhibitor therapies, the development of new therapeutic strategies and the identification of biomarkers for metastatic risk remain crucial. The study found that high SPC25 expression is closely associated with poor clinical outcomes, and knocking down SPC25 significantly inhibits tumor cell proliferation and migration. Non-targeted metabolomics analysis also revealed that SPC25 knockdown reduces tumor cell activity, resulting in a low-invasive state. Additionally, this study utilized high-throughput molecular docking to screen small molecule drugs targeting SPC25, aiming to find drugs that inhibit RCC metastasis. The research discovered that mefloquine, at concentrations that do not significantly kill tumor cells, can markedly inhibit RCC metastasis. It was the first to report that mefloquine achieves its anti-metastatic effects by binding to SPC25 and inhibiting epithelial-mesenchymal transition. These results suggest that SPC25 has the potential to serve as an early biomarker for metastatic risk in RCC and highlight a novel strategy where mefloquine inhibits RCC metastasis through SPC25 binding, offering new avenues to improve the prognosis of RCC patients.
Collapse
Affiliation(s)
- Yongbo Wang
- Cixi Biomedical Research InstituteWenzhou Medical UniversityWenzhouChina
| | - He Wang
- The Second Clinical Medical CollegeZhejiang Chinese Medical UniversityHangzhouChina
| | - Yipeng Xu
- Department of UrologyZhejiang Cancer HospitalHangzhouChina
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of MedicineChinese Academy of SciencesHangzhouChina
| | - Jiawei Ling
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Chuhong Zong
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Yan Zhang
- Postgraduate Training Base AllianceWenzhou Medical UniversityWenzhouChina
| | | | - Guanan Zhao
- Department of UrologyLishui People's HospitalLishuiChina
| | - Yuan Zhou
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Jiahui Zhao
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Pengrong Lou
- Department of Radiotherapy and ChemotherapyThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Xigao Liu
- Department of Urology, Qilu HospitalShandong UniversityJinanChina
| | - Tengfei Xu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Qi Ma
- Cixi Biomedical Research InstituteWenzhou Medical UniversityWenzhouChina
- Comprehensive Genitourinary Cancer CenterThe First Affiliated Hospital of Ningbo UniversityNingboChina
- Yi‐Huan Genitourinary Cancer GroupThe First Affiliated Hospital of Ningbo UniversityNingboChina
| |
Collapse
|
3
|
Basnet A, Thomas DD, Landreth KM, Damron FH, Liu TW. Immune Response to Bioluminescence Imaging Reporters in Murine Tumor Models. Mol Imaging Biol 2025:10.1007/s11307-025-02010-7. [PMID: 40234300 DOI: 10.1007/s11307-025-02010-7] [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: 03/04/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/17/2025]
Abstract
PURPOSE Imaging reporters have been widely employed in cancer research to monitor real-time tumor burden and metastatic spread. These tools offer a valuable approach for non-invasive imaging of tumor dynamics over time. With the established understanding that tumor immunology plays a critical role in cancer progression, it is essential to ensure that the chosen imaging reporters used to study tumor-immune interactions do not inadvertently elicit an immune response. This study aimed to investigate the immune response to bioluminescence reporters used for in vivo tracking of tumor cells in immunocompetent murine models. PROCEDURES The in vitro and in vivo growth effects of two stably expressed bioluminescence reporter genes, a red-shifted firefly luciferase and a click beetle green luciferase, were evaluated in four different cancer cell lines. Differences in parental and reporter-expressing cancer cell immune cell composition, activation, and secreted cytokine levels were evaluated using flow cytometry, cytokine arrays and ELISAs. RESULTS The data revealed no significant differences in in vitro cell proliferation between parental and reporter cancer cell lines. In vivo subcutaneous tumor growth was not observed in tumor cells stably expressing the red-shifted firefly luciferase. Cells labeled with click beetle green luciferase demonstrated no significant differences in in vivo subcutaneous tumor growth compared to parental cells. Tumor cells expressing red-shifted firefly luciferase induced an increase in activated and cytotoxic T cells compared to parental and click beetle green luciferase, suggesting enhanced immunogenicity. Furthermore, the tumor-immune composition and cytokine production were similar between parental and click beetle green luciferase-labeled tumor cells. CONCLUSIONS These findings demonstrate that the stable expression of click beetle green luciferase in cancer cells, in contrast to red-shifted firefly luciferase, has minimal immunogenicity and does not alter tumor development in immunocompetent mice. We report detailed characterization studies of bioluminescence reporter cells, providing essential considerations for their use in investigating tumor-immune interactions in syngeneic murine tumor models.
Collapse
Affiliation(s)
- Angisha Basnet
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
| | - Dylan D Thomas
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
| | - Kaitlyn M Landreth
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
| | - F Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, 26506, USA
| | - Tracy W Liu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA.
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
4
|
Han B, Zhang Y, Feng X, Yang J, Wang B, Fang J, Wang Z, Zhu J, Niu G, Guo Y. The power of microbes: the key role of gut microbiota in the initiation and progression of colorectal cancer. Front Oncol 2025; 15:1563886. [PMID: 40297806 PMCID: PMC12034544 DOI: 10.3389/fonc.2025.1563886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Colorectal cancer (CRC) is ranked as the third most prevalent malignancy and is a leading cause of cancer-related mortality globally, significantly affecting the health and longevity of middle-aged individuals and the elderly. The primary risk factors for CRC are mainly due to unhealthy dietary habits and lifestyle choices, and they have been shown to profoundly influence the composition of the gut microbiota. Given that dietary patterns are critical determinants of gut microbial diversity, a compelling association exists between gut microbiota and the pathogenesis of CRC. Recent research has increasingly focused on the intricate interplay between gut microbiota and CRC, exploring its role in disease initiation, progression, and the modulation of host immune responses. Investigations have demonstrated that certain specific microbial communities can promote inflammation, disrupt metabolic pathways, and produce carcinogenic compounds, thereby contributing to the development of CRC. Conversely, a diverse and balanced gut microbiome may confer protective effects against cancer through mechanisms such as the production of short-chain fatty acids and the enhancement of intestinal barrier integrity. This article provides a comprehensive overview of the characteristics of the gut microbial community and its complex relationship with CRC. It highlights potential mechanisms through which gut microbiota may influence CRC pathogenesis, including chronic inflammation, toxins, metabolites, epigenetic dysregulation, and immune regulatory dysfunction. Additionally, this review summarizes innovative strategies for CRC prevention and treatment, emphasizing the therapeutic potential of probiotics and natural plant extracts. By elucidating these connections, this work aims to enhance the understanding of the gut microbiome's role in CRC.
Collapse
Affiliation(s)
- Bo Han
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Yongfeng Zhang
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Xue Feng
- Department of Cardiology, 63650 Military Hospital, Urumqi, China
| | - Jun Yang
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Baolin Wang
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Jiang Fang
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Zhigang Wang
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Jun Zhu
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Ge Niu
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Youxiang Guo
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| |
Collapse
|
5
|
Dalal N, Dhandapani H, Ingle A, Sharma D, Tayalia P. Functionalized Poly(ethylene Glycol) Diacrylate Scaffolds for In Situ Immunomodulation of Dendritic Cells Targeting Melanoma Tumor. ACS Biomater Sci Eng 2025; 11:2396-2407. [PMID: 40048381 DOI: 10.1021/acsbiomaterials.4c02036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Various immunotherapeutic strategies are being developed to fight cancer, which is one of the leading causes of mortality. Dendritic cells (DCs), being professional antigen-presenting cells, after efficient manipulation with tumor-associated antigens, can lead to effective T-cell recruitment and activation at the tumor site, resulting in cytotoxic T-cell-mediated cancer cell killing. To circumvent the inefficiencies of ex vivo DC modification and patient infusion, an alternative strategy involving in situ DC activation has been explored here. Here, the vaccine components are tumor lysates, as antigens, and polyinosinic:polycytidylic acid (poly(I:C)), a toll-like receptor-3 (TLR3) agonist, as an adjuvant. Our in vitro studies demonstrate that complexing poly(I:C) with a carrier molecule, chitosan, enhances its stability and accessibility to TLR3 in the DC endosomal membrane. Material-based localized delivery of immunomodulatory factors is known to improve their stability and reduce their off-target side effects. Here, PEGDA-PLL-based macroporous scaffolds allow easy recruitment of host cells, thereby enabling effective interaction between the vaccine components loaded on them and the infiltrating immune cells. The vaccine components present in the scaffold facilitate efficient DC activation and migration, leading to subsequent T-cell activation and antitumor response, as shown by our in vivo studies.
Collapse
Affiliation(s)
- Neha Dalal
- Department of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Hemavathi Dhandapani
- Department of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Arvind Ingle
- Tata Memorial Centre Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Deepak Sharma
- Radiation Biology and Health Science Division, Bhabha Atomic Research Center, Mumbai 400085, India
| | - Prakriti Tayalia
- Department of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| |
Collapse
|
6
|
Sharma S, Gone GB, Roychowdhury P, Kim HS, Chung SJ, Kuppusamy G, De A. Photodynamic and sonodynamic therapy synergy: mechanistic insights and cellular responses against glioblastoma multiforme. J Drug Target 2025; 33:458-472. [PMID: 39556529 DOI: 10.1080/1061186x.2024.2431676] [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: 12/16/2023] [Revised: 10/30/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Glioblastoma multiforme (GBM), the most aggressive form of brain cancer, poses substantial challenges to effective treatment due to its complex and infiltrative nature, making it difficult to manage. Photodynamic therapy (PDT) and sonodynamic therapy (SDT), have emerged as promising individual treatment options against GBM due to their least-invasive approach. However, both PDT and SDT have drawbacks that require careful consideration. A combination therapy using light and sound waves has gained attention, offering new avenues to overcome challenges from individual therapies. Sono-photodynamic therapy (SPDT) has been used against various tumours. Researchers are considering SPDT as a favourable alternative to the conventional therapies for GBM. SPDT offers complementary mechanisms of action, including the production of ROS, disruption of cellular structures, and induction of apoptosis, leading to enhanced tumour cell death. This review gives an insight about PDT/SDT and their limitations in GBM treatment and the need for combination therapy. We try to unveil the process of SPDT and explore the mechanism behind improved SPDT-meditated cell death in GBM cells by focusing on the ROS-mediated cell response occurring as a result of SPDT and discussing current modifications in the existing sensitisers for their optimal use in SPDT for GBM therapy.
Collapse
Affiliation(s)
- Swati Sharma
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Geetanjali B Gone
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Parikshit Roychowdhury
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sang Jeon Chung
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Gowthmarajan Kuppusamy
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| | - Anindita De
- Department of Pharmaceutics, School of Pharmacy, JSS University, Noida, Uttar Pradesh, India
- Department of Pharmacy, Ajou University, Suwon-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
7
|
Gaoual Y, Mahyaoui A, Yachi L, Bouatia M, Aliat Z, Rahali Y. Advancements and challenges in CAR T cell therapy for pediatric brain tumors: A review. J Oncol Pharm Pract 2025:10781552251331609. [PMID: 40156311 DOI: 10.1177/10781552251331609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
Chimeric Antigen Receptor (CAR) T cell therapy represents a groundbreaking advancement in immunotherapy, initially gaining FDA approval for treating hematological malignancies. This therapy has shown promising results in solid tumors, particularly in pediatric brain tumors, which are the leading cause of cancer-related death in children. CAR T cells are engineered to target specific antigens on tumor cells, thereby reducing off-target effects and increasing the cytotoxic impact on cancer cells. Over the years, CAR T cell technology has evolved through five generations, each enhancing the structure, functionality, and safety of these cells. Despite these advancements, the application of CAR T cells in solid tumors, especially within the central nervous system (CNS), faces significant challenges. These include the physical barrier posed by the blood-brain barrier (BBB), the immunosuppressive tumor microenvironment (TME), and the heterogeneity of tumor antigens. The review discusses several promising antigenic targets for CAR T cells in pediatric brain tumors, such as HER2, EphA2, IL-13Rα2, and Survivin, which have been explored in recent clinical trials. These trials have shown early promise in improving patient outcomes, though the risks of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) remain concerns. The future of CAR T cell therapy lies in overcoming these barriers through innovative approaches like "Armored CARs" or TRUCKs, designed to modulate the TME and improve CAR T cell efficacy in solid tumors. Additionally, combination therapies and safety switches in next-generation CAR T cells are being explored to enhance therapeutic potential while minimizing adverse effects.
Collapse
Affiliation(s)
- Yasmina Gaoual
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Specialties Hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
| | - Adam Mahyaoui
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Specialties Hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
| | - Lamyae Yachi
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Children's hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
- Team of analytical chemistry and bromatology, Faculty of Medicine and Pharmacy, Mohammed V University- Rabat, 10 170 Rabat, Morocco
| | - Mustapha Bouatia
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Children's hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
- Team of analytical chemistry and bromatology, Faculty of Medicine and Pharmacy, Mohammed V University- Rabat, 10 170 Rabat, Morocco
- Ibn Sina University Hospital Center, 10 170 Rabat, Morocco
| | - Zineb Aliat
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Specialties Hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
- Team of Formulation and Quality Control of Health Products, Faculty of Medicine and Pharmacy, Mohammed V University- Rabat, 10 170 Rabat, Morocco
| | - Younes Rahali
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Specialties Hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
- Ibn Sina University Hospital Center, 10 170 Rabat, Morocco
- Team of Formulation and Quality Control of Health Products, Faculty of Medicine and Pharmacy, Mohammed V University- Rabat, 10 170 Rabat, Morocco
| |
Collapse
|
8
|
Magalhães MI, Azevedo MJ, Castro F, Oliveira MJ, Costa ÂM, Sampaio Maia B. The link between obesity and the gut microbiota and immune system in early-life. Crit Rev Microbiol 2025; 51:264-284. [PMID: 38651972 DOI: 10.1080/1040841x.2024.2342427] [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: 12/23/2023] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 04/25/2024]
Abstract
In early-life, the gut microbiota is highly modifiable, being modulated by external factors such as maternal microbiota, mode of delivery, and feeding strategies. The composition of the child's gut microbiota will deeply impact the development and maturation of its immune system, with consequences for future health. As one of the main sources of microorganisms to the child, the mother represents a crucial factor in the establishment of early-life microbiota, impacting the infant's wellbeing. Recent studies have proposed that dysbiotic maternal gut microbiota could be transmitted to the offspring, influencing the development of its immunity, and leading to the development of diseases such as obesity. This paper aims to review recent findings in gut microbiota and immune system interaction in early-life, highlighting the benefits of a balanced gut microbiota in the regulation of the immune system.
Collapse
Affiliation(s)
- Maria Inês Magalhães
- Doctoral Program in Biomedical Sciences, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
| | - Maria João Azevedo
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
- Academic Center for Dentistry Amsterdam (ACTA), Universiteit van Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Flávia Castro
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Maria José Oliveira
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ângela M Costa
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Benedita Sampaio Maia
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
| |
Collapse
|
9
|
Pappalardo F, Russo G. Comment on "A decade of thermostatted kinetic theory models for complex active matter living systems" by Carlo Bianca. Phys Life Rev 2025; 52:61-62. [PMID: 39657432 DOI: 10.1016/j.plrev.2024.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 12/12/2024]
Affiliation(s)
- Francesco Pappalardo
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria, 6, Catania 95125, Italy.
| | - Giulia Russo
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria, 6, Catania 95125, Italy
| |
Collapse
|
10
|
Liu Y, Yan D, Chen R, Zhang Y, Wang C, Qian G. Recent insights and advances in gut microbiota's influence on host antiviral immunity. Front Microbiol 2025; 16:1536778. [PMID: 40083779 PMCID: PMC11903723 DOI: 10.3389/fmicb.2025.1536778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/17/2025] [Indexed: 03/16/2025] Open
Abstract
A diverse array of microbial organisms colonizes the human body, collectively known as symbiotic microbial communities. Among the various pathogen infections that hosts encounter, viral infections represent one of the most significant public health challenges worldwide. The gut microbiota is considered an important biological barrier against viral infections and may serve as a promising target for adjuvant antiviral therapy. However, the potential impact of symbiotic microbiota on viral infection remains relatively understudied. In this review, we discuss the specific regulatory mechanisms of gut microbiota in antiviral immunity, highlighting recent advances in how gut microbiota regulate the host immune response, produce immune-related molecules, and enhance the host's defense against viruses. Finally, we also discuss the antiviral potential of oral probiotics.
Collapse
Affiliation(s)
- Ying Liu
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Danying Yan
- Department of Infectious Diseases, The First Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Ran Chen
- Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yingying Zhang
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Chuwen Wang
- Department of Infectious Diseases, The First Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Guoqing Qian
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- Department of Infectious Diseases, The First Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
11
|
Ge H, Wang M, Wei X, Chen XL, Wang X. Copper-Based Nanozymes: Potential Therapies for Infectious Wounds. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407195. [PMID: 39757568 DOI: 10.1002/smll.202407195] [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: 08/17/2024] [Revised: 11/30/2024] [Indexed: 01/07/2025]
Abstract
Bacterial infections are a significant obstacle to the healing of acute and chronic wounds, such as diabetic ulcers and burn injuries. Traditional antibiotics are the primary treatment for bacterial infections, but they present issues such as antibiotic resistance, limited efficacy, and potential side effects. This challenge leads to the exploration of nanozymes as alternative therapeutic agents. Nanozymes are nanomaterials with enzyme-like activities. Owing to their low production costs, high stability, scalability, and multifunctionality, nanozymes have emerged as a prominent focus in antimicrobial research. Among various types of nanozymes, metal-based nanozymes offer several benefits, including broad-spectrum antimicrobial activity and robust catalytic properties. Specifically, copper-based nanozymes (CuNZs) have shown considerable potential in promoting wound healing. They exhibit strong antimicrobial effects, reduce inflammation, and enhance tissue regeneration, making them highly advantageous for use in wound care. This review describes the dual functions of CuNZs in combating infection and facilitating wound repair. Recent advancements in the design and synthesis of CuNZs, evaluating their antimicrobial efficacy, healing promotion, and biosafety both in vitro and in vivo on the basis of their core components, are critically important.
Collapse
Affiliation(s)
- Haojie Ge
- Department of Burns, The First Hospital Affiliated of Anhui Medical University, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| | - Min Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Xiaolong Wei
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Xu-Lin Chen
- Department of Burns, The First Hospital Affiliated of Anhui Medical University, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
| | - Xianwen Wang
- Department of Burns, The First Hospital Affiliated of Anhui Medical University, Anhui Medical University, Hefei, Anhui, 230032, P. R. China
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| |
Collapse
|
12
|
Spiliopoulou P, Rousakis P, Panteli C, Eleutherakis‐Papaiakovou E, Migkou M, Kanellias N, Ntanasis‐Stathopoulos I, Malandrakis P, Theodorakakou F, Fotiou D, Terpos E, Gavriatopoulou M, Tsitsilonis OE, Kastritis E, Dimopoulos MA, Terzis G. Effects of Exercise Training on the Bone Marrow Immune Microenvironment and Minimal Residual Disease in Multiple Myeloma Patients Following First-Line Treatment. Scand J Med Sci Sports 2025; 35:e70020. [PMID: 39853819 PMCID: PMC11760657 DOI: 10.1111/sms.70020] [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: 09/19/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025]
Abstract
The purpose of the study was to investigate the effects of exercise training on the bone marrow immune microenvironment and on minimal residual disease of multiple myeloma patients who completed first-line induction treatment. Eight multiple myeloma patients underwent 5 months of exercise training along with standard medical treatment. Eight age- and sex-matched patients who received medical treatment only, served as controls. Before and after the intervention, white blood cells, red blood cells, and platelets, as well as the percentages of neutrophils, lymphocytes, monocytes, eosinophils, and basophils, were measured in the peripheral blood. Abnormal plasma cells, normal plasma cells, B cells, T cells, NK/NKT cells, monocytes, neutrophils, eosinophils, basophils, mast cells, myeloid progenitors, erythroid progenitors, and erythroblasts were assessed in the bone marrow. Exercise training increased the percentage of blood monocytes (mean difference 3.5% ± 2.6%; p = 0.006), while no change was detected in the control group. In the bone marrow, the CD27+ T cell subset increased (mean difference 18.2% ± 21.9%; p = 0.043) and the ratio of CD27-/CD27+ T lymphocytes decreased (pre: 1.06 ± 0.59; post: 0.76 ± 0.47; p = 0.049) in the exercise group, but remained unaltered in the control group. In conclusion, the study provides evidence that 5 months of exercise training can induce an increase in the percentage of activated T lymphocytes, as shown by the higher expression of the costimulatory CD27 marker. It also suggests that exercise-induced changes in the bone marrow microenvironment may be beneficial in the control of clonal cell proliferation.
Collapse
Affiliation(s)
- Polyxeni Spiliopoulou
- Sports Performance Laboratory, School of Physical Education and Sport ScienceNational and Kapodistrian University of AthensAthensGreece
| | - Pantelis Rousakis
- Flow Cytometry Unit, Department of BiologyNational and Kapodistrian University of AthensAthensGreece
| | - Chrysanthi Panteli
- Flow Cytometry Unit, Department of BiologyNational and Kapodistrian University of AthensAthensGreece
| | - Evangelos Eleutherakis‐Papaiakovou
- Department of Clinical Therapeutics, School of Medicine, Alexandra General HospitalNational and Kapodistrian University of AthensAthensGreece
| | - Magdalini Migkou
- Department of Clinical Therapeutics, School of Medicine, Alexandra General HospitalNational and Kapodistrian University of AthensAthensGreece
| | - Nikolaos Kanellias
- Department of Clinical Therapeutics, School of Medicine, Alexandra General HospitalNational and Kapodistrian University of AthensAthensGreece
| | - Ioannis Ntanasis‐Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, Alexandra General HospitalNational and Kapodistrian University of AthensAthensGreece
| | - Panagiotis Malandrakis
- Department of Clinical Therapeutics, School of Medicine, Alexandra General HospitalNational and Kapodistrian University of AthensAthensGreece
| | - Foteini Theodorakakou
- Department of Clinical Therapeutics, School of Medicine, Alexandra General HospitalNational and Kapodistrian University of AthensAthensGreece
| | - Despina Fotiou
- Department of Clinical Therapeutics, School of Medicine, Alexandra General HospitalNational and Kapodistrian University of AthensAthensGreece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, Alexandra General HospitalNational and Kapodistrian University of AthensAthensGreece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, Alexandra General HospitalNational and Kapodistrian University of AthensAthensGreece
| | - Ourania E. Tsitsilonis
- Flow Cytometry Unit, Department of BiologyNational and Kapodistrian University of AthensAthensGreece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, Alexandra General HospitalNational and Kapodistrian University of AthensAthensGreece
| | - Meletios Athanasios Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, Alexandra General HospitalNational and Kapodistrian University of AthensAthensGreece
| | - Gerasimos Terzis
- Sports Performance Laboratory, School of Physical Education and Sport ScienceNational and Kapodistrian University of AthensAthensGreece
| |
Collapse
|
13
|
Kiruba B, Naidu A, Sundararajan V, Lulu S S. Mapping integral cell-type-specific interferon-induced gene regulatory networks (GRNs) involved in systemic lupus erythematosus using systems and computational analysis. Heliyon 2025; 11:e41342. [PMID: 39844998 PMCID: PMC11751531 DOI: 10.1016/j.heliyon.2024.e41342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disorder characterized by the production of autoantibodies, resulting in inflammation and organ damage. Although extensive research has been conducted on SLE pathogenesis, a comprehensive understanding of its molecular landscape in different cell types has not been achieved. This study uncovers the molecular mechanisms of the disease by thoroughly examining gene regulatory networks within neutrophils, dendritic cells, T cells, and B cells. Firstly, we identified genes and ncRNAs with differential expression in SLE patients compared to controls for different cell types. Furthermore, the derived differentially expressed genes were curated based on immune functions using functional enrichment analysis to create a protein-protein interaction network. Topological network analysis of the formed genes revealed key hub genes associated with each of the cell types. To understand the regulatory mechanism through which these hub genes function in the diseased state, their associations with transcription factors, and non-coding RNAs in different immune cell types were investigated through correlation analysis and regression models. Finally, by integrating these findings, distinct gene regulatory networks were constructed, which provide a novel perspective on the molecular, cellular, and immunological landscapes of SLE. Importantly, we reveal the crucial role of IRF3, IRF7, and STAT1 in neutrophils, dendritic cells, and T cells, where their aberrant upregulation in disease states might enhance the production of type I IFN. Furthermore, we found MYB to be a crucial regulator that might activate T cells toward autoimmune responses in SLE. Similarly, in B-cell lymphocytes, we found FOXO1 to be a key player in autophagy and chemokine regulation. These findings were also validated using single-cell RNASeq analysis using an independent dataset. Genotype variations of these genes were also explored using the GWAS central database to ensure their targetability. These findings indicate that IRF3, IRF7, STAT1, MYB, and FOXO1 are promising targets for therapeutic interventions for SLE.
Collapse
Affiliation(s)
- Blessy Kiruba
- Department of Biosciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632 014, Tamil Nadu, India
| | - Akshayata Naidu
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632 014, Tamil Nadu, India
| | - Vino Sundararajan
- Department of Biosciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632 014, Tamil Nadu, India
| | - Sajitha Lulu S
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, 632 014, Tamil Nadu, India
| |
Collapse
|
14
|
Kelly C, Sartor RB, Rawls JF. Early subclinical stages of the inflammatory bowel diseases: insights from human and animal studies. Am J Physiol Gastrointest Liver Physiol 2025; 328:G17-G31. [PMID: 39499254 PMCID: PMC11901386 DOI: 10.1152/ajpgi.00252.2024] [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: 08/16/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/07/2024]
Abstract
The inflammatory bowel diseases (IBD) occur in genetically susceptible individuals that mount inappropriate immune responses to their microbiota leading to chronic intestinal inflammation. The natural history of IBD progression begins with early subclinical stages of disease that occur before clinical diagnosis. Improved understanding of those early subclinical stages could lead to new or improved strategies for IBD diagnosis, prognostication, or prevention. Here, we review our current understanding of the early subclinical stages of IBD in humans including studies from first-degree relatives of patients with IBD and members of the general population who go on to develop IBD. We also discuss representative mouse models of IBD that can be used to investigate disease dynamics and host-microbiota relationships during these early stages. In particular, we underscore how mouse models of IBD that develop disease later in life with variable penetrance may present valuable opportunities to discern early subclinical mechanisms of disease before histological inflammation and other severe symptoms become apparent.
Collapse
Affiliation(s)
- Cecelia Kelly
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, North Carolina, United States
| | - R Balfour Sartor
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, United States
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, North Carolina, United States
| |
Collapse
|
15
|
Ibeanu GC, Rowaiye AB, Okoli JC, Eze DU. Microbiome Differences in Colorectal Cancer Patients and Healthy Individuals: Implications for Vaccine Antigen Discovery. Immunotargets Ther 2024; 13:749-774. [PMID: 39698218 PMCID: PMC11652712 DOI: 10.2147/itt.s486731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
Background Colorectal cancer (CRC) is the third most prevalent cancer worldwide, with numerous risk factors contributing to its development. Recent research has illuminated the significant role of the gut microbiota in CRC pathogenesis, identifying various microbial antigens as potential targets for vaccine development. Aim This review aimed at exploring the potential sources of microbial antigens that could be harnessed to create effective CRC vaccines and understand the role of microbiome-CRC interactions in carcinogenesis. Methods A comprehensive search of original research and review articles on the pathological links between key microbial candidates, particularly those more prevalent in CRC tissues, was conducted. This involved extensive use of the PubMed and Medline databases, as well as the Google Scholar search engine, utilizing pertinent keywords. A total of one hundred and forty-three relevant articles in English, mostly published between 2018 and 2024, were selected. Results Numerous microbes, particularly bacteria and viruses, are significantly overrepresented in CRC tissues and have been shown to promote tumorigenesis by inducing inflammation and modulating the immune system. This makes them promising candidates for antigens in the development of CRC vaccines. Conclusion The selection of microbial antigens focuses on their capacity to trigger a strong immune response and their link to tumor presence and progression. Identifying and validating these antigens through preclinical testing is essential in developing a CRC vaccine.
Collapse
Affiliation(s)
- Gordon C Ibeanu
- Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC, 27707, USA
| | - Adekunle B Rowaiye
- Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC, 27707, USA
- Department of Agricultural Biotechnology, National Biotechnology Development Agency, Abuja, Nigeria
| | - Joy C Okoli
- Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC, 27707, USA
| | - Daniel U Eze
- Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC, 27707, USA
| |
Collapse
|
16
|
Thapa R, Magar AT, Shrestha J, Panth N, Idrees S, Sadaf T, Bashyal S, Elwakil BH, Sugandhi VV, Rojekar S, Nikhate R, Gupta G, Singh SK, Dua K, Hansbro PM, Paudel KR. Influence of gut and lung dysbiosis on lung cancer progression and their modulation as promising therapeutic targets: a comprehensive review. MedComm (Beijing) 2024; 5:e70018. [PMID: 39584048 PMCID: PMC11586092 DOI: 10.1002/mco2.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/26/2024] Open
Abstract
Lung cancer (LC) continues to pose the highest mortality and exhibits a common prevalence among all types of cancer. The genetic interaction between human eukaryotes and microbial cells plays a vital role in orchestrating every physiological activity of the host. The dynamic crosstalk between gut and lung microbiomes and the gut-lung axis communication network has been widely accepted as promising factors influencing LC progression. The advent of the 16s rDNA sequencing technique has opened new horizons for elucidating the lung microbiome and its potential pathophysiological role in LC and other infectious lung diseases using a molecular approach. Numerous studies have reported the direct involvement of the host microbiome in lung tumorigenesis processes and their impact on current treatment strategies such as radiotherapy, chemotherapy, or immunotherapy. The genetic and metabolomic cross-interaction, microbiome-dependent host immune modulation, and the close association between microbiota composition and treatment outcomes strongly suggest that designing microbiome-based treatment strategies and investigating new molecules targeting the common holobiome could offer potential alternatives to develop effective therapeutic principles for LC treatment. This review aims to highlight the interaction between the host and microbiome in LC progression and the possibility of manipulating altered microbiome ecology as therapeutic targets.
Collapse
Affiliation(s)
- Rajan Thapa
- Department of Pharmacy, Universal college of medical sciencesTribhuvan UniversityBhairahawaRupendehiNepal
| | - Anjana Thapa Magar
- Department of MedicineKathmandu Medical College Teaching Hospital, SinamangalKathmanduNepal
| | - Jesus Shrestha
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNew South WalesAustralia
| | - Nisha Panth
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Sobia Idrees
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Tayyaba Sadaf
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Saroj Bashyal
- Department of Pharmacy, Manmohan Memorial Institute of Health SciencesTribhuvan University, SoalteemodeKathmanduNepal
| | - Bassma H. Elwakil
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences TechnologyPharos University in AlexandriaAlexandriaEgypt
| | - Vrashabh V. Sugandhi
- Department of pharmaceutical sciences, College of Pharmacy & Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - Satish Rojekar
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ram Nikhate
- Department of PharmaceuticsDattakala Shikshan Sanstha, Dattakala college of pharmacy (Affiliated to Savitribai Phule Pune universityPuneMaharashtraIndia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical SciencesSaveetha UniversityChennaiIndia
- Centre of Medical and Bio‐allied Health Sciences ResearchAjman UniversityAjmanUAE
| | - Sachin Kumar Singh
- School of Pharmaceutical SciencesLovely Professional UniversityPhagwaraIndia
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Philip M Hansbro
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| |
Collapse
|
17
|
Niazi SK, Magoola M. MicroRNA Nobel Prize: Timely Recognition and High Anticipation of Future Products-A Prospective Analysis. Int J Mol Sci 2024; 25:12883. [PMID: 39684593 PMCID: PMC11641023 DOI: 10.3390/ijms252312883] [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: 10/21/2024] [Revised: 11/20/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
MicroRNAs (miRNAs) maintain cellular homeostasis by blocking mRNAs by binding with them to fine-tune the expression of genes across numerous biological pathways. The 2024 Nobel Prize in Medicine and Physiology for discovering miRNAs was long overdue. We anticipate a deluge of research work involving miRNAs to repeat the history of prizes awarded for research on other RNAs. Although miRNA therapies are included for several complex diseases, the realization that miRNAs regulate genes and their roles in addressing therapies for hundreds of diseases are expected; but with advancement in drug discovery tools, we anticipate even faster entry of new drugs. To promote this, we provide details of the current science, logic, intellectual property, formulations, and regulatory process with anticipation that many more researchers will introduce novel therapies based on the discussion and advice provided in this paper.
Collapse
|
18
|
Snow TAC, Singer M, Arulkumaran N. Antibiotic-Induced Immunosuppression-A Focus on Cellular Immunity. Antibiotics (Basel) 2024; 13:1034. [PMID: 39596729 PMCID: PMC11591424 DOI: 10.3390/antibiotics13111034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Antibiotics are the fundamental treatment for bacterial infections. However, they are associated with numerous side effects. Their adverse effects on the immune system are increasingly recognised, with several mechanisms identified. In this review, we focus on their direct effects on cellular immunity. We review the effects of antibiotics on mitochondrial function and how they impair specific immune cell functions including chemotaxis, phagocytosis, cytokine production, antigen presentation, and lymphocyte proliferation. Findings are described in a multitude of in vivo and in vitro models. However, their impact on patient immunity and clinical outcomes requires further research. Awareness of the potential adverse effects of antibiotics may improve antimicrobial stewardship. The use of therapeutic drug monitoring may help to reduce dose-dependent effects, which warrants further research.
Collapse
Affiliation(s)
| | | | - Nishkantha Arulkumaran
- Bloomsbury Institute of Intensive Care Medicine, University College London, London WC1E 6DH, UK; (T.A.C.S.); (M.S.)
| |
Collapse
|
19
|
Tsang AP, Krishnan SN, Eliason JN, McGue JJ, Qin A, Frankel TL, Rao A. Assessing the Tumor Immune Landscape Across Multiple Spatial Scales to Differentiate Immunotherapy Response in Metastatic Non-Small Cell Lung Cancer. J Transl Med 2024; 104:102148. [PMID: 39389312 DOI: 10.1016/j.labinv.2024.102148] [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: 05/03/2024] [Revised: 09/16/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024] Open
Abstract
Although immune checkpoint inhibitor-based therapy has shown promising results in non-small cell lung cancer patients with high programmed death-ligand 1 expression, not all patients respond to therapy. The tumor microenvironment (TME) is complex and heterogeneous, making it challenging to understand the key agents and features that influence response to therapies. In this study, we leverage multiplex fluorescent immunohistochemistry to quantitatively assess interactions between tumor and immune cells in an effort to identify patterns occurring at multiple spatial levels of the TME. To do so, we introduce several computational methods novel to a data set of 1,269 multiplex fluorescent immunohistochemistry images from a cohort of 52 patients with metastatic non-small cell lung cancer. With the spatial G-cross function, we quantify the degree of cell interaction at an entire image level, where we see significantly increased activity of cytotoxic T cells and helper T cells with epithelial tumor cells in responders to immune checkpoint inhibitor-based (P = .022 and P < .001, respectively) and decreased activity of T-regulatory cells with epithelial tumor cells compared with nonresponders (P = .010). By leveraging spatial overlap methods, we define tumor subregions (which we call the tumor "periphery," "edge." and "center") and discover more localized immune-immune interactions influencing positive response, including those between cytotoxic T cells and helper T cells with antigen presenting cells in these subregions specifically. Finally, we trained an interpretable deep learning model that identified key cellular regions of interest that most influenced response classification (area under the curve = 0.71 ± 0.02). Assessing spatial interactions within these subregions further revealed new insights that were not significant at the whole image level, particularly the elevated association of antigen presenting cells and T-regulatory cells with one another in responder groups (P = .024). Altogether, we demonstrate that elucidating patterns of cell composition and interplay across multiple levels of spatial analyses can improve our understanding of the TME and better differentiate patient responses to immunotherapy.
Collapse
MESH Headings
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/secondary
- Carcinoma, Non-Small-Cell Lung/therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Immune Checkpoint Inhibitors/pharmacology
- Immune Checkpoint Inhibitors/therapeutic use
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
- Cell Communication/drug effects
- Cell Communication/immunology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Helper-Inducer/drug effects
- T-Lymphocytes, Helper-Inducer/immunology
- Antigen-Presenting Cells/drug effects
- Antigen-Presenting Cells/immunology
- Deep Learning
- Area Under Curve
- Spatial Analysis
- Fluorescent Antibody Technique
- Cohort Studies
- Humans
- Treatment Outcome
Collapse
Affiliation(s)
- Ashley P Tsang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan.
| | - Santhoshi N Krishnan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Joel N Eliason
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Jake J McGue
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Angel Qin
- Department of Internal Medicine, Division of Hematology-Oncology, University of Michigan, Ann Arbor, Michigan
| | | | - Arvind Rao
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan; Department of Biostatistics, University of Michigan, Ann Arbor, Michigan; Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan; Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
20
|
Chen X, Wu J, Zhou B, Zhu M, Zhang J, Zhou N, Zhu YZ, Zhang X, Duan X, Men K. Bacterial Lysate-Based Bifunctional mRNA Nanoformulation for Efficient Colon Cancer Immunogene Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56580-56598. [PMID: 39397736 DOI: 10.1021/acsami.4c07684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
mRNA-based nonviral gene therapy has played an important role in cancer therapy, however, the limited delivery efficiency and therapeutic capacity still require further exploration and enhancement. Immunogene therapy provides a strategy for cancer treatment. Bacteria are tiny single-celled living organisms, many of which can be found in and on the human body and are beneficial to humans. Lactobacillus reuteri is a bacterial member of the gut flora, and recent research has shown that it can reduce intestinal inflammation by stimulating an immunomodulatory response. L. reuteri lysate represents an ideal resource for constructing advanced mRNA delivery systems with immune stimulation potential. Here, we prepared a bifunctional mRNA delivery system DMP-Lac (DOTAP-mPEG-PCL-L. reuteri lysate), which successfully codelivered L. reuteri lysate and IL-23A mRNA, exhibited a high mRNA delivery efficiency of 75.56% ± 0.85%, and strongly promoted the maturation and activation of the immune system in vivo. Both the CT26 abdominal metastasis model and the lung metastasis model also exhibited a good therapeutic effect, and the tumor inhibition rate of DMP-Lac/IL-23A group reached 97.92%. Protein chip technology verified that DMP acted as an immune adjuvant, demonstrating that the L. reuteri lysate could regulate the related immune cells, while IL-23 mRNA caused changes in downstream factors, thus producing the corresponding tumor treatment effect. The DMP-Lac/IL-23A complex exhibited strong anticancer immunotherapeutic effects. Our results demonstrated that this bifunctional mRNA formulation served as a tumor-specific nanomedicine, providing an advanced strategy for colon cancer immunogene therapy.
Collapse
Affiliation(s)
- Xiaohua Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Jieping Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Bailing Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Manfang Zhu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Jin Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Na Zhou
- State Key Laboratory for Quality Research of Chinese Medicines and School of Pharmacy, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Yi Zhun Zhu
- State Key Laboratory for Quality Research of Chinese Medicines and School of Pharmacy, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Xin Zhang
- State Key Laboratory for Quality Research of Chinese Medicines and School of Pharmacy, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| |
Collapse
|
21
|
Avdonin PP, Blinova MS, Serkova AA, Komleva LA, Avdonin PV. Immunity and Coagulation in COVID-19. Int J Mol Sci 2024; 25:11267. [PMID: 39457048 PMCID: PMC11508857 DOI: 10.3390/ijms252011267] [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: 08/19/2024] [Revised: 09/23/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Discovered in late 2019, the SARS-CoV-2 coronavirus has caused the largest pandemic of the 21st century, claiming more than seven million lives. In most cases, the COVID-19 disease caused by the SARS-CoV-2 virus is relatively mild and affects only the upper respiratory tract; it most often manifests itself with fever, chills, cough, and sore throat, but also has less-common mild symptoms. In most cases, patients do not require hospitalization, and fully recover. However, in some cases, infection with the SARS-CoV-2 virus leads to the development of a severe form of COVID-19, which is characterized by the development of life-threatening complications affecting not only the lungs, but also other organs and systems. In particular, various forms of thrombotic complications are common among patients with a severe form of COVID-19. The mechanisms for the development of thrombotic complications in COVID-19 remain unclear. Accumulated data indicate that the pathogenesis of severe COVID-19 is based on disruptions in the functioning of various innate immune systems. The key role in the primary response to a viral infection is assigned to two systems. These are the pattern recognition receptors, primarily members of the toll-like receptor (TLR) family, and the complement system. Both systems are the first to engage in the fight against the virus and launch a whole range of mechanisms aimed at its rapid elimination. Normally, their joint activity leads to the destruction of the pathogen and recovery. However, disruptions in the functioning of these innate immune systems in COVID-19 can cause the development of an excessive inflammatory response that is dangerous for the body. In turn, excessive inflammation entails activation of and damage to the vascular endothelium, as well as the development of the hypercoagulable state observed in patients seriously ill with COVID-19. Activation of the endothelium and hypercoagulation lead to the development of thrombosis and, as a result, damage to organs and tissues. Immune-mediated thrombotic complications are termed "immunothrombosis". In this review, we discuss in detail the features of immunothrombosis associated with SARS-CoV-2 infection and its potential underlying mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Pavel V. Avdonin
- Koltzov Institute of Developmental Biology RAS, ul. Vavilova, 26, 119334 Moscow, Russia; (P.P.A.)
| |
Collapse
|
22
|
Echevarria-Lima J, Moles R. Monocyte and Macrophage Functions in Oncogenic Viral Infections. Viruses 2024; 16:1612. [PMID: 39459945 PMCID: PMC11512331 DOI: 10.3390/v16101612] [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: 09/10/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Monocytes and macrophages are part of innate immunity and constitute the first line of defense against pathogens. Bone marrow-derived monocytes circulate in the bloodstream for one to three days and then typically migrate into tissues, where they differentiate into macrophages. Circulatory monocytes represent 5% of the nucleated cells in normal adult blood. Following differentiation, macrophages are distributed into various tissues and organs to take residence and maintain body homeostasis. Emerging evidence has highlighted the critical role of monocytes/macrophages in oncogenic viral infections, mainly their crucial functions in viral persistence and disease progression. These findings open opportunities to target innate immunity in the context of oncogenic viruses and to explore their potential as immunotherapies.
Collapse
Affiliation(s)
- Juliana Echevarria-Lima
- Laboratório de Imunologia Básica e Aplicada, Department of Immunology, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil;
| | - Ramona Moles
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Center for Immunology and Microbial Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
23
|
Kamali AN, Hamedifar H, Eisenhut M, Bautista JM. Multiple myeloma and the potential of new checkpoint inhibitors for immunotherapy. Ther Adv Vaccines Immunother 2024; 12:25151355241288453. [PMID: 39399301 PMCID: PMC11467827 DOI: 10.1177/25151355241288453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024] Open
Abstract
Multiple myeloma (MM), a cancer of the bone marrow, is categorized as the second most common hematological malignancy of adults in the Western world. Despite dramatic improvements in immunotherapies in the field of cancers, MM immunotherapy has not been promising until now. Recent clinical studies of immune checkpoint inhibitor therapy, either alone or in combination with anticancer drugs, showed excessive side effects or low efficacy, particularly in advanced MM patients. In this context, lymphocyte levels of exhaustion markers play a pivotal role in the MM tumor microenvironment (TME). Hence in the present review, the mechanisms relevant to MM of five inhibitory molecules including T-cell immunoreceptor with Ig and ITIM domains (TIGIT), T-cell immunoglobulin, and mucin domain 3 (Tim-3), lymphocyte activation gene-3 (LAG-3), V-domain Ig Suppressor of T-cell activation and killer immunoglobulin-like receptors along with bispecific T-cell antibodies (BsAbs) will be discussed. Further, we summarized the underlying biology of these checkpoints in cancer and their rapidly emerging role in pathways in MM along with presenting recent clinical trials in context.
Collapse
Affiliation(s)
- Ali N. Kamali
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| | - Haleh Hamedifar
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| | - Michael Eisenhut
- Department of Pediatrics, Luton & Dunstable University Hospital, Luton, UK
| | - Jose M. Bautista
- Department of Biochemistry and Molecular Biology, Complutense University of Madrid, Faculty of Veterinary Sciences, Madrid, Spain
- Research Institute Hospital 12 de Octubre, Madrid, Spain
| |
Collapse
|
24
|
Di Gioacchino M, Santilli F, Pession A. Is There a Role for Immunostimulant Bacterial Lysates in the Management of Respiratory Tract Infection? Biomolecules 2024; 14:1249. [PMID: 39456182 PMCID: PMC11505618 DOI: 10.3390/biom14101249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
Bacterial Lysates are immunostimulants clinically prescribed for the prevention of respiratory tract infections (RTIs). It has been shown that Bacterial Lysates upregulate the immune system, acting both on innate and adaptive reactions. In fact, there are demonstrations of their efficacy in restoring the integrity and immune function of epithelial barriers, activating ILC3 and dendritic cells with an enhanced Th1 response, and producing serum IgG and serum and salivary IgA specific to the administered bacterial antigens. The activated immune system also protects against other bacteria and viruses due to a trained immunity effect. Most studies show that the number of RTIs and their severity decrease in Bacterial Lysates-pretreated patients, without relevant side effects. The Bacterial Lysates treatment, in addition to reducing the number of RTIs, also prevents the deterioration of the underlying disease (i.e., COPD) induced by repeated infections. Despite these positive data, the most recent meta-analyses evidence the weakness of the studies performed, which are of low quality and have an inadequate number of patients, some of which were non-randomized while others were without a control group or were performed contemporarily in different clinical conditions or with different ages. The high heterogeneity of the studies does not allow us to state Bacterial Lysates' effectiveness in preventing RTIs with sufficient certainty. To completely define their indications, double-blind, placebo-controlled, multicenter, randomized clinical trials should be performed for each product and for each indication. The study population should be adequate for each indication. For this purpose, an adequate run-in phase will be necessary.
Collapse
Affiliation(s)
- Mario Di Gioacchino
- Institute for Clinical Immunotherapy and Advanced Biological Treatments, 65100 Pescara, Italy
| | - Francesca Santilli
- Center for Advanced Science and Technology (CAST), G. d’Annunzio University, 66100 Chieti, Italy;
- Department of Medicine and Science of Aging, G. d’Annunzio University, 66100 Chieti, Italy
| | - Andrea Pession
- Department of Medicine and Surgery, “Alma Mater Studiorum”-University of Bologna, 40100 Bologna, Italy;
| |
Collapse
|
25
|
Zaid NSN, Muhamad AS, Jawis MN, Ooi FK, Mohamed M, Mohamud R, Hamdan NF, Jusoh N. The Effect of Exercise on Immune Response in Population with Increased Risk Factors for Cardiovascular Disease: A Systematic Review. Malays J Med Sci 2024; 31:83-108. [PMID: 39416746 PMCID: PMC11477469 DOI: 10.21315/mjms2024.31.5.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 04/04/2024] [Indexed: 10/19/2024] Open
Abstract
This systematic review aimed to provide information on existing interventional studies that evaluate the efficacy of exercise in populations with increased cardiovascular disease (CVD) risk factors through immune functional perspectives. A literature search was conducted in four databases: PubMed, Scopus, Taylor & Francis and ScienceDirect from January 2012 to February 2023. The articles were screened and evaluated for quality before data were extracted. The review protocol was registered at PROSPERO (CRD42022321704). In total, 18 studies were included for quality appraisal and synthesised evidence indicated that exercise contributes to enhancing the functioning of both innate and adaptive immune responses, potentially serving as an anti-immunosenescent response to exercise in individuals with elevated CVD risk factors. Furthermore, the review emphasised that exercise, irrespective of its type, intensity or mode, was well tolerated by individuals at increased risk for CVD and may have significant implications in generating anti-inflammatory effects.
Collapse
Affiliation(s)
- Nik Siti Nik Zaid
- Exercise and Sports Science Programme, School of Health Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Ayu Suzailiana Muhamad
- Exercise and Sports Science Programme, School of Health Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Mohd Nidzam Jawis
- Exercise and Sports Science Programme, School of Health Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Foong Kiew Ooi
- Exercise and Sports Science Programme, School of Health Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Mahaneem Mohamed
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Nor Faeiza Hamdan
- Exercise and Sports Science Programme, School of Health Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Normah Jusoh
- Faculty of Sports Science and Coaching, Universiti Pendidikan Sultan Idris, Perak, Malaysia
| |
Collapse
|
26
|
Ashique S, Mishra N, Garg A, Kumar N, Khan Z, Mohanto S, Chellappan DK, Farid A, Taghizadeh-Hesary F. A Critical Review on the Role of Probiotics in Lung Cancer Biology and Prognosis. Arch Bronconeumol 2024; 60 Suppl 2:S46-S58. [PMID: 38755052 DOI: 10.1016/j.arbres.2024.04.030] [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: 02/20/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 05/18/2024]
Abstract
Lung cancer remains the leading cause of cancer-related deaths worldwide. According to the American Cancer Society (ACS), it ranks as the second most prevalent type of cancer globally. Recent findings have highlighted bidirectional gut-lung interactions, known as the gut-lung axis, in the pathophysiology of lung cancer. Probiotics are live microorganisms that boost host immunity when consumed adequately. The immunoregulatory mechanisms of probiotics are thought to operate through the generation of various metabolites that impact both the gut and distant organs (e.g., the lungs) through blood. Several randomized controlled trials have highlighted the pivotal role of probiotics in gut health especially for the prevention and treatment of malignancies, with a specific emphasis on lung cancer. Current research indicates that probiotic supplementation positively affects patients, leading to a suppression in cancer symptoms and a shortened disease course. While clinical trials validate the therapeutic benefits of probiotics, their precise mechanism of action remains unclear. This narrative review aims to provide a comprehensive overview of the present landscape of probiotics in the management of lung cancer.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences & Research, Durgapur 713212, West Bengal, India.
| | - Neeraj Mishra
- Amity Institute of Pharmacy, Amity University Madhya Pradesh, Gwalior 474005, MP, India
| | - Ashish Garg
- Guru Ramdas Khalsa Institute of Science and Technology, Pharmacy, Jabalpur, MP 483001, India
| | - Nitish Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204, India
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Arshad Farid
- Gomal Center of Biochemistry and Biotechnology, Gomal University, Dera Ismail Khan 29050, Pakistan
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Clinical Oncology, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
27
|
Mahmoudi F, Jalayeri MHT, Montaseri A, MohamedKhosroshahi L, Baradaran B. Microbial natural compounds and secondary metabolites as Immunomodulators: A review. Int J Biol Macromol 2024; 278:134778. [PMID: 39153680 DOI: 10.1016/j.ijbiomac.2024.134778] [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: 09/04/2023] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Immunomodulatory therapies are beneficial strategies for the improvement of immune system function. Today, due to the increasing prevalence of immune disorders, cancer, and new viral diseases, there is a greater need to introduce immunomodulatory compounds with more efficiency and fewer side effects. Microbial derivatives are fertile and attractive grounds for discovering lots of novel compounds with various medical properties. The discovery of many natural compounds derived from bacterial sources, such as secondary metabolites with promising immunomodulating activities, represents the importance of this topic in drug discovery and emphasizes the necessity for a coherent source of study in this area. Considering this need, in this review, we aim to focus on the current information about the immunomodulatory effects of bacterial secondary metabolites and natural immunomodulators derived from microorganisms.
Collapse
Affiliation(s)
- Fariba Mahmoudi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Hadi Tajik Jalayeri
- Clinical Research Development Unit (CRDU), Sayad Shirazi Hospital Golestan University of Medical Sciences, Gorgan, Iran
| | - Azadeh Montaseri
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy.
| | - Leila MohamedKhosroshahi
- Department of Immunology, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
28
|
Chernyak N, Bhagwat B, Naravula S, Chen Y, Solban N, Zhang F, Kofman E, Raoufi F, Dang X, Bao J, Tomazela D, Baily M, Geierstanger B, Flygare JA, Han JH, Willingham A, Seganish WM. Discovery and Evaluation of TLR-Targeted Immune Agonists. J Med Chem 2024; 67:16222-16234. [PMID: 39235949 DOI: 10.1021/acs.jmedchem.4c00804] [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: 09/07/2024]
Abstract
Toll-like receptor (TLR) activation converts immunologically inactive tumors into immunologically active tumors by activating tumor residing antigen-presenting cells and recruitment of cytotoxic T lymphocytes. Targeted immune agonists (TIAs) are antibody drug conjugates with small-molecule TLR agonist payloads. The mechanism of action of TIAs involves tumor antigen recognition, Fcγ-receptor-dependent phagocytosis, and TLR-mediated activation to drive tumor killing by myeloid cells. Several new low DAR anti-HER2 TIAs conjugated with novel TLR7 or dual-TLR7/8 agonists with cleavable and noncleavable linkers were synthesized and profiled. In vitro studies demonstrated that these TIAs activate myeloid cells only in the presence of antigen-expressing cancer cells. Evaluation in ELISpot-based assays confirmed the low immunogenicity of these constructs. Systemic administration of the novel TIAs in tumor-bearing mice resulted in tumor reduction at low doses. These results provide a strong rationale for further development of the TIAs as a novel class of immunotherapeutics.
Collapse
Affiliation(s)
- Natalia Chernyak
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Bhagyashree Bhagwat
- Discovery Biologics, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Saraswathi Naravula
- Discovery Biologics, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Ying Chen
- Pharmacokinetics, Dynamics, Metabolism and Bioanalytics, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Nicolas Solban
- Quantitative Biosciences, Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Fan Zhang
- Discovery Biologics, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Esther Kofman
- Discovery Biologics, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Fahimeh Raoufi
- Discovery Biologics, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Xibei Dang
- Pharmacokinetics, Dynamics, Metabolism and Bioanalytics, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Jianming Bao
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Daniela Tomazela
- Discovery Biologics, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Marc Baily
- Discovery Biologics, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Bernhard Geierstanger
- Discovery Biologics, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - John A Flygare
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Jin-Hwan Han
- Discovery Oncology, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Aarron Willingham
- Discovery Biologics, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - W Michael Seganish
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| |
Collapse
|
29
|
Guryanova SV. Bacteria and Allergic Diseases. Int J Mol Sci 2024; 25:10298. [PMID: 39408628 PMCID: PMC11477026 DOI: 10.3390/ijms251910298] [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: 08/26/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Microorganisms colonize all barrier tissues and are present on the skin and all mucous membranes from birth. Bacteria have many ways of influencing the host organism, including activation of innate immunity receptors by pathogen-associated molecular patterns and synthesis of various chemical compounds, such as vitamins, short-chain fatty acids, bacteriocins, toxins. Bacteria, using extracellular vesicles, can also introduce high-molecular compounds, such as proteins and nucleic acids, into the cell, regulating the metabolic pathways of the host cells. Epithelial cells and immune cells recognize bacterial bioregulators and, depending on the microenvironment and context, determine the direction and intensity of the immune response. A large number of factors influence the maintenance of symbiotic microflora, the diversity of which protects hosts against pathogen colonization. Reduced bacterial diversity is associated with pathogen dominance and allergic diseases of the skin, gastrointestinal tract, and upper and lower respiratory tract, as seen in atopic dermatitis, allergic rhinitis, chronic rhinosinusitis, food allergies, and asthma. Understanding the multifactorial influence of microflora on maintaining health and disease determines the effectiveness of therapy and disease prevention and changes our food preferences and lifestyle to maintain health and active longevity.
Collapse
Affiliation(s)
- Svetlana V. Guryanova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia; ; Tel.: +7-(915)3150073
- Medical Institute, Peoples’ Friendship University of Russia, 117198 Moscow, Russia
| |
Collapse
|
30
|
Pejcic A, Andjelkovic Z, Marjanovic D, Minic I, Matvijenko V, Arsic Z, Jovanovic R, Subaric L. Comparative analysis of antigen-presenting cells in gingival tissues in healthy and periodontitis patients. J Clin Pathol 2024; 77:702-708. [PMID: 37433669 DOI: 10.1136/jcp-2021-207975] [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: 09/27/2021] [Accepted: 07/01/2023] [Indexed: 07/13/2023]
Abstract
AIMS Microbial flora of dental plaque trigger innate and adaptive immune responses. The function of antigen-presenting cells (APCs) is to bridge the innate and adaptive immune systems. The human immune system contains three main types of APCs: dendritic cells (DC) (Langerhans cells (LCs) and interstitial DCs, IDCs), macrophages and B lymphocytes. In this study, the distribution and density of all APCs in healthy and inflamed human gingival tissue were comparatively analysed. METHODS Research was conducted on gingival biopsy specimens obtained from 55 patients and classified in three groups: healthy gingiva (control group, n=10), moderate periodontal disease (PD) (n=21) and severe PD (n=24). For APCs' identification antibodies raised against CD1a (for LCs), S100 protein (for iDCs), CD68 (for macrophages) and CD20 (for B lymphocytes) were used. RESULTS Increased density of IDCs, macrophages and B lymphocytes in lamina propria and reduced density of LCs in the gingival epithelium were found in patients with periodontitis. Simultaneously, it was noticed an increased concentration of macrophages and B cells in the gingival epithelium in patients with PD. No statistically significant difference in the distribution and density of APC was found among patients with moderate and advanced periodontitis. CONCLUSIONS It was hypothesised that in the periodontitis the role of antigen presentation was largely taken from LCs by the DCs, macrophages and B cells. These APCs are thought to have less protective and tolerogenic potential than LCs and this is a significant reason for alveolar bone destruction in periodontitis.
Collapse
Affiliation(s)
- Ana Pejcic
- Periodontology and Oral Medicine, University of Niš, Medical Faculty, Nis, Serbia
| | - Zlatibor Andjelkovic
- Institute of Histology and Embriology, University of Pristina, Medical Faculty, Kosovska Mitrovica, Serbia
| | - Dragan Marjanovic
- Periodontology and Oral Medicine, University of Pristina, Dental Clinic, Kosovska Mitrovica, Serbia
| | - Ivan Minic
- Periodontology and Oral Medicine, University of Niš, Medical Faculty, Nis, Serbia
| | - Vladimir Matvijenko
- Restorative Dentistry, University of Pristina, Dental Clinic, Kosovska Mitrovica, Serbia
| | - Zoran Arsic
- Restorative Dentistry, University of Pristina, Dental Clinic, Kosovska Mitrovica, Serbia
| | - Radovan Jovanovic
- Dental Clinic, University of Pristina, Medical Faculty, Kosovska Mitrovica, Serbia
| | - Ljiljana Subaric
- Dental Clinic, University of Pristina, Medical Faculty, Kosovska Mitrovica, Serbia
| |
Collapse
|
31
|
Winter L, Ries J, Vogl C, Trumet L, Geppert CI, Lutz R, Kesting M, Weber M. Comparative Analysis of Inhibitory and Activating Immune Checkpoints PD-1, PD-L1, CD28, and CD86 in Non-Melanoma Skin Cancer. Cells 2024; 13:1569. [PMID: 39329753 PMCID: PMC11430031 DOI: 10.3390/cells13181569] [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: 06/05/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
The establishment of immunotherapy applying immune checkpoint inhibitors (ICI) has provided an important new option for the treatment of solid malignant diseases. However, different tumor entities show dramatically different responses to this therapy. BCC responds worse to anti-PD-1 ICIs as compared to cSCC. Differential immune checkpoint expression could explain this discrepancy and, therefore, the aim of this study was to analyze activating and inhibitory immune checkpoints in cSCC and BCC tissues. Tissue microarrays of the invasive front as well as the tumor core of BCC and cSCC samples were used to evaluate PD-1, PD-L1, CD28, and CD86 expression and their topographic distribution profiles by chromogenic immunohistochemistry. QuPath was used to determine the labeling index. The expression of PD-1, PD-L1, and CD28 was significantly higher in both the tumor core and the invasive front of cSCC samples as compared to BCC (p < 0.001). In addition, the ratios of PD-L1/CD86 (p < 0.001) and CD28/CD86 (p < 0.001) were significantly higher in cSCC. The invasive front of both tumor entities showed higher expression levels of all immune markers compared to the tumor core in both tumor entities. The significantly higher expression of PD-1, PD-L1, and CD28 in cSCC, along with the predominance of the inhibitory ligand PD-L1 as compared to the activating CD86 in cSCC, provide a potential explanation for the better objective response rates to anti-PD-1 immunotherapy as compared to BCC. Furthermore, the predominant site of interaction between the immune system and the tumor was within the invasive front in both tumor types.
Collapse
Affiliation(s)
- Linus Winter
- Department of Oral and Cranio-Maxillofacial Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.W.); (C.V.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC WERA), 91052 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| | - Jutta Ries
- Department of Oral and Cranio-Maxillofacial Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.W.); (C.V.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC WERA), 91052 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| | - Christoph Vogl
- Department of Oral and Cranio-Maxillofacial Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.W.); (C.V.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC WERA), 91052 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| | - Leah Trumet
- Department of Oral and Cranio-Maxillofacial Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.W.); (C.V.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC WERA), 91052 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
- Department of Operative Dentistry and Periodontology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Carol Immanuel Geppert
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC WERA), 91052 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Rainer Lutz
- Department of Oral and Cranio-Maxillofacial Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.W.); (C.V.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC WERA), 91052 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| | - Marco Kesting
- Department of Oral and Cranio-Maxillofacial Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.W.); (C.V.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC WERA), 91052 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| | - Manuel Weber
- Department of Oral and Cranio-Maxillofacial Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.W.); (C.V.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC WERA), 91052 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| |
Collapse
|
32
|
Galli F, Bartolini D, Ronco C. Oxidative stress, defective proteostasis and immunometabolic complications in critically ill patients. Eur J Clin Invest 2024; 54:e14229. [PMID: 38676423 DOI: 10.1111/eci.14229] [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: 02/22/2024] [Revised: 03/31/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024]
Abstract
Oxidative stress (OS) develops in critically ill patients as a metabolic consequence of the immunoinflammatory and degenerative processes of the tissues. These induce increased and/or dysregulated fluxes of reactive species enhancing their pro-oxidant activity and toxicity. At the same time, OS sustains its own inflammatory and immunometabolic pathogenesis, leading to a pervasive and vitious cycle of events that contribute to defective immunity, organ dysfunction and poor prognosis. Protein damage is a key player of these OS effects; it generates increased levels of protein oxidation products and misfolded proteins in both the cellular and extracellular environment, and contributes to forms DAMPs and other proteinaceous material to be removed by endocytosis and proteostasis processes of different cell types, as endothelial cells, tissue resident monocytes-macrophages and peripheral immune cells. An excess of OS and protein damage in critical illness can overwhelm such cellular processes ultimately interfering with systemic proteostasis, and consequently with innate immunity and cell death pathways of the tissues thus sustaining organ dysfunction mechanisms. Extracorporeal therapies based on biocompatible/bioactive membranes and new adsorption techniques may hold some potential in reducing the impact of OS on the defective proteostasis of patients with critical illness. These can help neutralizing reactive and toxic species, also removing solutes in a wide spectrum of molecular weights thus improving proteostasis and its immunometabolic corelates. Pharmacological therapy is also moving steps forward which could help to enhance the efficacy of extracorporeal treatments. This narrative review article explores the aspects behind the origin and pathogenic role of OS in intensive care and critically ill patients, with a focus on protein damage as a cause of impaired systemic proteostasis and immune dysfunction in critical illness.
Collapse
Affiliation(s)
- Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Desirée Bartolini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Claudio Ronco
- Department of Medicine, International Renal Research Institute of Vicenza, University of Padova, San Bortolo Hospital Vicenza, Vicenza, Italy
| |
Collapse
|
33
|
Suh YJ, Li AT, Pandey M, Nordmann CS, Huang YL, Wu M. Decoding physical principles of cell migration under controlled environment using microfluidics. BIOPHYSICS REVIEWS 2024; 5:031302. [PMID: 39091432 PMCID: PMC11290890 DOI: 10.1063/5.0199161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 06/26/2024] [Indexed: 08/04/2024]
Abstract
Living cells can perform incredible tasks that man-made micro/nano-sized robots have not yet been able to accomplish. One example is that white blood cells can sense and move to the site of pathogen attack within minutes. The robustness and precision of cellular functions have been perfected through billions of years of evolution. In this context, we ask the question whether cells follow a set of physical principles to sense, adapt, and migrate. Microfluidics has emerged as an enabling technology for recreating well-defined cellular environment for cell migration studies, and its ability to follow single cell dynamics allows for the results to be amenable for theoretical modeling. In this review, we focus on the development of microfluidic platforms for recreating cellular biophysical (e.g., mechanical stress) and biochemical (e.g., nutrients and cytokines) environments for cell migration studies in 3D. We summarize the basic principles that cells (including bacteria, algal, and mammalian cells) use to respond to chemical gradients learned from microfluidic systems. We also discuss about novel biological insights gained from studies of cell migration under biophysical cues and the need for further quantitative studies of cell function under well-controlled biophysical environments in the future.
Collapse
Affiliation(s)
- Young Joon Suh
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Alan T. Li
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Mrinal Pandey
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Cassidy S. Nordmann
- Department of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Yu Ling Huang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Mingming Wu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
34
|
Biswas M, Nurunnabi M, Khatun Z. Understanding Mucosal Physiology and Rationale of Formulation Design for Improved Mucosal Immunity. ACS APPLIED BIO MATERIALS 2024; 7:5037-5056. [PMID: 38787767 DOI: 10.1021/acsabm.4c00395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
The oral and nasal cavities serve as critical gateways for infectious pathogens, with microorganisms primarily gaining entry through these routes. Our first line of defense against these invaders is the mucosal membrane, a protective barrier that shields the body's internal systems from infection while also contributing to vital functions like air and nutrient intake. One of the key features of this mucosal barrier is its ability to protect the physiological system from pathogens. Additionally, mucosal tolerance plays a crucial role in maintaining homeostasis by regulating the pH and water balance within the body. Recognizing the importance of the mucosal barrier, researchers have developed various mucosal formulations to enhance the immune response. Mucosal vaccines, for example, deliver antigens directly to mucosal tissues, triggering local immune stimulation and ultimately inducing systemic immunity. Studies have shown that lipid-based formulations such as liposomes and virosomes can effectively elicit both local and systemic immune responses. Furthermore, mucoadhesive polymeric particles, with their prolonged delivery to target sites, have demonstrated an enhanced immune response. This Review delves into the critical role of material selection and delivery approaches in optimizing mucosal immunity.
Collapse
Affiliation(s)
- Mila Biswas
- Department of Electrical and Computer Engineering, University of Texas at El Paso, El Paso, Texas 79902, United States
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
- Department of Biomedical Engineering, College of Engineering, University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Zehedina Khatun
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
| |
Collapse
|
35
|
Biswas R, Swetha RG, Basu S, Roy A, Ramaiah S, Anbarasu A. Designing multi-epitope vaccine against human cytomegalovirus integrating pan-genome and reverse vaccinology pipelines. Biologicals 2024; 87:101782. [PMID: 39003966 DOI: 10.1016/j.biologicals.2024.101782] [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: 12/06/2023] [Revised: 05/13/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024] Open
Abstract
Human cytomegalovirus (HCMV) is accountable for high morbidity in neonates and immunosuppressed individuals. Due to the high genetic variability of HCMV, current prophylactic measures are insufficient. In this study, we employed a pan-genome and reverse vaccinology approach to screen the target for efficient vaccine candidates. Four proteins, envelope glycoprotein M, UL41A, US23, and US28, were shortlisted based on cellular localization, high solubility, antigenicity, and immunogenicity. A total of 29 B-cell and 44 T-cell highly immunogenic and antigenic epitopes with high global population coverage were finalized using immunoinformatics tools and algorithms. Further, the epitopes that were overlapping among the finalized B-cell and T-cell epitopes were linked with suitable linkers to form various combinations of multi-epitopic vaccine constructs. Among 16 vaccine constructs, Vc12 was selected based on physicochemical and structural properties. The docking and molecular simulations of VC12 were performed, which showed its high binding affinity (-23.35 kcal/mol) towards TLR4 due to intermolecular hydrogen bonds, salt bridges, and hydrophobic interactions, and there were only minimal fluctuations. Furthermore, Vc12 eliciting a good response was checked for its expression in Escherichia coli through in silico cloning and codon optimization, suggesting it to be a potent vaccine candidate.
Collapse
Affiliation(s)
- Rhitam Biswas
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India; Department of Biotechnology, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Rayapadi G Swetha
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India; Department of Biosciences, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Soumya Basu
- Department of Biotechnology, NIST University, Berhampur, 761008, Odisha, India
| | - Aditi Roy
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India; Department of Biotechnology, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India; Department of Biosciences, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India; Department of Biotechnology, SBST, VIT, Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
36
|
Gómez García AM, López Muñoz F, García-Rico E. The Microbiota in Cancer: A Secondary Player or a Protagonist? Curr Issues Mol Biol 2024; 46:7812-7831. [PMID: 39194680 DOI: 10.3390/cimb46080463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/11/2024] [Accepted: 07/18/2024] [Indexed: 08/29/2024] Open
Abstract
The intestinal microbiota and the human body are in a permanent interaction. There is a symbiotic relationship in which the microbiota plays a vitally important role in the performance of numerous functions, including digestion, metabolism, the development of lymphoid tissue, defensive functions, and other processes. It is a true metabolic organ essential for life and has potential involvement in various pathological states, including cancer and pathologies other than those of a digestive nature. A growing topic of great interest for its implications is the relationship between the microbiota and cancer. Dysbiosis plays a role in oncogenesis, tumor progression, and even the response to cancer treatment. The effect of the microbiota on tumor development goes beyond a local effect having a systemic effect. Another aspect of great interest regarding the intestinal microbiota is its relationship with drugs, modifying their activity. There is increasing evidence that the microbiota influences the therapeutic activity and side effects of antineoplastic drugs and also modulates the response of several tumors to antineoplastic therapy through immunological circuits. These data suggest the manipulation of the microbiota as a possible adjuvant to improve oncological treatment. Is it possible to manipulate the microbiota for therapeutic purposes?
Collapse
Affiliation(s)
- Ana María Gómez García
- Internal Medicine Unit, Hospital Universitario HM Madrid, 28015 Madrid, Spain
- Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, 28692 Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, 28015 Madrid, Spain
| | - Francisco López Muñoz
- Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, 28692 Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, 28015 Madrid, Spain
| | - Eduardo García-Rico
- Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, 28692 Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, 28015 Madrid, Spain
- Medical Oncology Unit, Hospital Universitario HM Torrelodones, 28250 Torrelodones, Spain
| |
Collapse
|
37
|
Li X, Wang H, Lai W, Liao J, Mo W, Huang K, He L, Liang X, Yu Z, Xu J, Hua X, Hou F, Ding J, Jia WWG, Zhang K, Wang Y. Prevention and treatment of HPV-related cancer through a mRNA vaccine expressing APC-targeting antigen. Immunology 2024; 172:375-391. [PMID: 38471664 DOI: 10.1111/imm.13777] [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: 08/09/2023] [Accepted: 02/24/2024] [Indexed: 03/14/2024] Open
Abstract
Persistent human papillomavirus (HPV) infection is associated with multiple malignancies. Developing therapeutic vaccines to eliminate HPV-infected and malignant cells holds significant value. In this study, we introduced a lipid nanoparticle encapsulated mRNA vaccine expressing tHA-mE7-mE6. Mutations were introduced into E6 and E7 of HPV to eliminate their tumourigenicity. A truncated influenza haemagglutinin protein (tHA), which binds to the CD209 receptor on the surface of dendritic cells (DCs), was fused with mE7-mE6 in order to allow efficient uptake of antigen by antigen presenting cells. The tHA-mE7-mE6 (mRNA) showed higher therapeutic efficacy than mE7-mE6 (mRNA) in an E6 and E7+ tumour model. The treatment resulted in complete tumour regression and prevented tumour formation. Strong CD8+ T-cell immune response was induced, contributing to preventing and curing of E6 and E7+ tumour. Antigen-specific CD8+ T were found in spleens, peripheral blood and in tumours. In addition, the tumour infiltration of DC and NK cells were increased post therapy. In conclusion, this study described a therapeutic mRNA vaccine inducing strong anti-tumour immunity in peripheral and in tumour microenvironment, holding promising potential to treat HPV-induced cancer and to prevent cancer recurrence.
Collapse
Affiliation(s)
- Xiaoxuan Li
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Huiyan Wang
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wujiang Lai
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Obstetrics and Gynecology, Shunde Hospital, The First People's Hospital of Shunde, Southern Medical University, Foshan, Guangdong, China
| | - Jinrong Liao
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wenyu Mo
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Keke Huang
- Obstetrics and Gynecology, Shunde Hospital, The First People's Hospital of Shunde, Southern Medical University, Foshan, Guangdong, China
| | - Liqing He
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaomei Liang
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhibin Yu
- Shanghai Virogin Biotech Co., Ltd, Shanghai, China
| | - Jiang Xu
- Shanghai Virogin Biotech Co., Ltd, Shanghai, China
| | - Xianwu Hua
- Shanghai Virogin Biotech Co., Ltd, Shanghai, China
| | - Fujun Hou
- Shanghai Virogin Biotech Co., Ltd, Shanghai, China
| | - Jun Ding
- Virogin Biotech Canada Ltd, Vancouver, Canada
| | | | - Kuan Zhang
- Shanghai Virogin Biotech Co., Ltd, Shanghai, China
| | - Yifeng Wang
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
38
|
Shi Y, Strasser A, Green DR, Latz E, Mantovani A, Melino G. Legacy of the discovery of the T-cell receptor: 40 years of shaping basic immunology and translational work to develop novel therapies. Cell Mol Immunol 2024; 21:790-797. [PMID: 38822079 PMCID: PMC11214623 DOI: 10.1038/s41423-024-01168-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 04/18/2024] [Indexed: 06/02/2024] Open
Affiliation(s)
- Yufang Shi
- The Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Soochow University, Suzhou, 215000, China.
| | - Andreas Strasser
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Eicke Latz
- Institute of Innate Immunity, University of Bonn, Bonn, 53127, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, 53175, Germany
| | | | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
| |
Collapse
|
39
|
Sheikhlary S, Lopez DH, Moghimi S, Sun B. Recent Findings on Therapeutic Cancer Vaccines: An Updated Review. Biomolecules 2024; 14:503. [PMID: 38672519 PMCID: PMC11048403 DOI: 10.3390/biom14040503] [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: 02/23/2024] [Revised: 04/06/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Cancer remains one of the global leading causes of death and various vaccines have been developed over the years against it, including cell-based, nucleic acid-based, and viral-based cancer vaccines. Although many vaccines have been effective in in vivo and clinical studies and some have been FDA-approved, there are major limitations to overcome: (1) developing one universal vaccine for a specific cancer is difficult, as tumors with different antigens are different for different individuals, (2) the tumor antigens may be similar to the body's own antigens, and (3) there is the possibility of cancer recurrence. Therefore, developing personalized cancer vaccines with the ability to distinguish between the tumor and the body's antigens is indispensable. This paper provides a comprehensive review of different types of cancer vaccines and highlights important factors necessary for developing efficient cancer vaccines. Moreover, the application of other technologies in cancer therapy is discussed. Finally, several insights and conclusions are presented, such as the possibility of using cold plasma and cancer stem cells in developing future cancer vaccines, to tackle the major limitations in the cancer vaccine developmental process.
Collapse
Affiliation(s)
- Sara Sheikhlary
- Department of Biomedical Engineering, College of Engineering, The University of Arizona, Tucson, AZ 85721, USA
| | - David Humberto Lopez
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| | - Sophia Moghimi
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| | - Bo Sun
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| |
Collapse
|
40
|
Wang Z, Chen X, Yan L, Wang W, Zheng P, Mohammadreza A, Liu Q. Antimicrobial peptides in bone regeneration: mechanism and potential. Expert Opin Biol Ther 2024; 24:285-304. [PMID: 38567503 DOI: 10.1080/14712598.2024.2337239] [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: 11/27/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION Antimicrobial peptides (AMPs) are small-molecule peptides with a unique antimicrobial mechanism. Other notable biological activities of AMPs, including anti-inflammatory, angiogenesis, and bone formation effects, have recently received widespread attention. These remarkable bioactivities, combined with the unique antimicrobial mechanism of action of AMPs, have led to their increasingly important role in bone regeneration. AREAS COVERED In this review, on the one hand, we aimed to summarize information about the AMPs that are currently used for bone regeneration by reviewing published literature in the PubMed database. On the other hand, we also highlight some AMPs with potential roles in bone regeneration and their possible mechanisms of action. EXPERT OPINION The translation of AMPs to the clinic still faces many problems, but their unique antimicrobial mechanisms and other conspicuous biological activities suggest great potential. An in-depth understanding of the structure and mechanism of action of AMPs will help us to subsequently combine AMPs with different carrier systems and perform structural modifications to reduce toxicity and achieve stable release, which may be a key strategy for facilitating the translation of AMPs to the clinic.
Collapse
Affiliation(s)
- ZhiCheng Wang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - XiaoMan Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Liang Yan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - WenJie Wang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - PeiJia Zheng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Atashbahar Mohammadreza
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of International Education, Southern Medical University, Guangzhou, China
| | - Qi Liu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| |
Collapse
|
41
|
Auclert LZ, Chhanda MS, Derome N. Interwoven processes in fish development: microbial community succession and immune maturation. PeerJ 2024; 12:e17051. [PMID: 38560465 PMCID: PMC10981415 DOI: 10.7717/peerj.17051] [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: 07/06/2023] [Accepted: 02/13/2024] [Indexed: 04/04/2024] Open
Abstract
Fishes are hosts for many microorganisms that provide them with beneficial effects on growth, immune system development, nutrition and protection against pathogens. In order to avoid spreading of infectious diseases in aquaculture, prevention includes vaccinations and routine disinfection of eggs and equipment, while curative treatments consist in the administration of antibiotics. Vaccination processes can stress the fish and require substantial farmer's investment. Additionally, disinfection and antibiotics are not specific, and while they may be effective in the short term, they have major drawbacks in the long term. Indeed, they eliminate beneficial bacteria which are useful for the host and promote the raising of antibiotic resistance in beneficial, commensal but also in pathogenic bacterial strains. Numerous publications highlight the importance that plays the diversified microbial community colonizing fish (i.e., microbiota) in the development, health and ultimately survival of their host. This review targets the current knowledge on the bidirectional communication between the microbiota and the fish immune system during fish development. It explores the extent of this mutualistic relationship: on one hand, the effect that microbes exert on the immune system ontogeny of fishes, and on the other hand, the impact of critical steps in immune system development on the microbial recruitment and succession throughout their life. We will first describe the immune system and its ontogeny and gene expression steps in the immune system development of fishes. Secondly, the plurality of the microbiotas (depending on host organism, organ, and development stage) will be reviewed. Then, a description of the constant interactions between microbiota and immune system throughout the fish's life stages will be discussed. Healthy microbiotas allow immune system maturation and modulation of inflammation, both of which contribute to immune homeostasis. Thus, immune equilibrium is closely linked to microbiota stability and to the stages of microbial community succession during the host development. We will provide examples from several fish species and describe more extensively the mechanisms occurring in zebrafish model because immune system ontogeny is much more finely described for this species, thanks to the many existing zebrafish mutants which allow more precise investigations. We will conclude on how the conceptual framework associated to the research on the immune system will benefit from considering the relations between microbiota and immune system maturation. More precisely, the development of active tolerance of the microbiota from the earliest stages of life enables the sustainable establishment of a complex healthy microbial community in the adult host. Establishing a balanced host-microbiota interaction avoids triggering deleterious inflammation, and maintains immunological and microbiological homeostasis.
Collapse
Affiliation(s)
- Lisa Zoé Auclert
- Département de Biologie, Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec, Canada
| | - Mousumi Sarker Chhanda
- Département de Biologie, Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec, Canada
- Department of Aquaculture, Faculty of Fisheries, Hajee Mohammad Danesh Science and Technology University, Basherhat, Bangladesh
| | - Nicolas Derome
- Département de Biologie, Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec, Canada
| |
Collapse
|
42
|
Perumalsamy H, Xiao X, Kim HY, Yoon TH. scRNA-seq analysis discovered suppression of immunomodulatory dependent inflammatory response in PMBCs exposed to silver nanoparticles. J Nanobiotechnology 2024; 22:118. [PMID: 38494495 PMCID: PMC10946150 DOI: 10.1186/s12951-024-02364-0] [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: 12/04/2023] [Accepted: 02/21/2024] [Indexed: 03/19/2024] Open
Abstract
The assessment of AgNPs toxicity in vitro and in vivo models are frequently conflicting and inaccurate. Nevertheless, single cell immunological responses in a heterogenous environment have received little attention. Therefore, in this study, we have performed in-depth analysis which clearly revealed cellular-metal ion association as well as specific immunological response. Our study didn't show significant population differences in PMBC between control and AgNPs group implying no toxicological response. To confirm it further, deep profiling identified differences in subsets and differentially expressed genes (DEGs) of monocytes, B cells and T cells. Notably, monocyte subsets showed significant upregulation of metallothionein (MT) gene expression such as MT1G, MT1X, MT1E, MT1A, and MT1F. On the other hand, downregulation of pro-inflammatory genes such as IL1β and CCL3 in both CD16 + and CD16- monocyte subsets were observed. This result indicated that AgNPs association with monocyte subsets de-promoted inflammatory responsive genes suggesting no significant toxicity observed in AgNPs treated group. Other cell types such as B cells and T cells also showed negligible differences in their subsets suggesting no toxicity response. Further, AgNPs treated group showed upregulation of cell proliferation, ribosomal synthesis, downregulation of cytokine release, and T cell differentiation inhibition. Overall, our results conclude that treatment of AgNPs to PMBC cells didn't display immunological related cytotoxicity response and thus motivate researchers to use them actively for biomedical applications.
Collapse
Affiliation(s)
- Haribalan Perumalsamy
- Center for Creative Convergence Education, Hanyang University, Seoul, 04763, Republic of Korea
- Institute of Next Generation Material Design, Hanyang University, Seoul, 04763, Republic of Korea
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, 04763, Republic of Korea
| | - Xiao Xiao
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea
| | - Hyun-Yi Kim
- NGeneS Inc, 362, Gwangdeok 1-ro, Sangnok-gu, Ansan-si, Gyeonggi-do, 15495, Republic of Korea
| | - Tae-Hyun Yoon
- Institute of Next Generation Material Design, Hanyang University, Seoul, 04763, Republic of Korea.
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea.
- Department of Medical and Digital Engineering, Hanyang University, Seoul, 04763, Republic of Korea.
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
43
|
Shvets Y, Khranovska N, Senchylo N, Ostapchenko D, Tymoshenko I, Onysenko S, Kobyliak N, Falalyeyeva T. Microbiota substances modulate dendritic cells activity: A critical view. Heliyon 2024; 10:e27125. [PMID: 38444507 PMCID: PMC10912702 DOI: 10.1016/j.heliyon.2024.e27125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/23/2024] [Accepted: 02/23/2024] [Indexed: 03/07/2024] Open
Abstract
Contemporary research in the field of microbiota shows that commensal bacteria influence physiological activity of different organs and systems of a human organism, such as brain, lungs, immune and metabolic systems. This influence is realized by various processes. One of them is trough modulation of immune mechanisms. Interactions between microbiota and the human immune system are known to be complex and ambiguous. Dendritic cells (DCs) are unique cells, which initiate the development and polarization of adaptive immune response. These cells also interconnect native and specific immune reactivity. A large set of biochemical signals from microbiota in the form of different microbiota associated molecular patterns (MAMPs) and bacterial metabolites that act locally and distantly in the human organism. As a result, commensal bacteria influence the maturity and activity of dendritic cells and affect the overall immune reactivity of the human organism. It then determines the response to pathogenic microorganisms, inflammation, associated with different pathological conditions and even affects the effectiveness of vaccination.
Collapse
Affiliation(s)
- Yuliia Shvets
- Taras Shevchenko National University of Kyiv, 64/13 Volodymyrska Str., Kyiv, Ukraine
| | - Natalia Khranovska
- National Cancer Institute of Ukraine, 33/43 Yuliia Zdanovska Str., Kyiv, Ukraine
| | - Natalia Senchylo
- Taras Shevchenko National University of Kyiv, 64/13 Volodymyrska Str., Kyiv, Ukraine
| | - Danylo Ostapchenko
- Taras Shevchenko National University of Kyiv, 64/13 Volodymyrska Str., Kyiv, Ukraine
| | - Iryna Tymoshenko
- Bogomolets National Medical University, 13 Shevchenka Blvd., Kyiv, Ukraine
| | - Svitlana Onysenko
- Taras Shevchenko National University of Kyiv, 64/13 Volodymyrska Str., Kyiv, Ukraine
| | - Nazarii Kobyliak
- Bogomolets National Medical University, 13 Shevchenka Blvd., Kyiv, Ukraine
- Medical Laboratory CSD, 22b Zhmerynska Str., Kyiv, Ukraine
| | - Tetyana Falalyeyeva
- Taras Shevchenko National University of Kyiv, 64/13 Volodymyrska Str., Kyiv, Ukraine
- Medical Laboratory CSD, 22b Zhmerynska Str., Kyiv, Ukraine
| |
Collapse
|
44
|
Shi Y, Su C, Ding T, Zhao H, Wang Y, Ren Y, Wu L, Zhang Q, Liang J, Sun S, Wang J, Li J, Zeng X. Manganese suppresses the development of oral leukoplakia by activating the immune response. Oral Dis 2024; 30:462-476. [PMID: 36260219 DOI: 10.1111/odi.14412] [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: 07/16/2022] [Revised: 10/01/2022] [Accepted: 10/14/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Manganese ion (Mn2+ ) is reported to promote the antitumor immune response by activating the cGAS-STING pathway, but it is unknown whether Mn2+ can prevent the malignant transformation of precancerous lesions. The effects of Mn2+ in treating oral leukoplakia (OLK) were explored in this work. METHODS Peripheral blood Mn analysis of the patients was performed using inductively coupled plasma atomic emission spectroscopy (ICP-AES). A coculture model of dendritic cells (DCs)/macrophages, CD8+ T cells, and dysplastic oral keratinocytes (DOKs) was employed to analyze the role and mechanism of Mn2+ in a simulated OLK immune microenvironment. Western blot, RT-PCR, flow cytometry, enzyme-linked immunosorbent assay (ELISA), and lactate dehydrogenase (LDH) assays were adopted to detect the mechanism of Mn2+ in this model. 4-nitroquinoline oxide (4NQO)-induced OLK mice were used to assess the role of Mn2+ in suppressing OLK progression, and a novel Mn2+ -loaded guanosine-tannic acid hydrogel (G-TA@Mn2+ hydrogel) was fabricated and evaluated for its advantages in OLK therapy. RESULTS The content of Mn in patients' peripheral blood was negatively related to the progression of OLK. Mn2+ promoted the maturation and antigen presentation of DCs and macrophages and enhanced the activation of CD8+ T cells in the coculture model, resulting in effective killing of DOKs. Mechanistic analysis found that Mn2+ enhanced the anti-OLK immune response by activating the cGAS-STING pathway. Moreover, Mn2+ suppressed the development of 4NQO-induced carcinogenesis in the mouse model. In addition, the G-TA@Mn2+ hydrogel had better anti-OLK effects. CONCLUSIONS Mn2+ enhanced the anti-OLK immune response by activating the cGAS-STING pathway, and the G-TA@Mn2+ hydrogel is a potential novel therapeutic approach for OLK treatment.
Collapse
Affiliation(s)
- Yujie Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chongying Su
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tingting Ding
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hang Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ying Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuan Ren
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lanyan Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qiyue Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Liang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Silu Sun
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiongke Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
45
|
Rokhsartalab Azar P, Karimi S, Haghtalab A, Taram S, Hejazi M, Sadeghpour S, Pashaei MR, Ghasemnejad-Berenji H, Taheri-Anganeh M. The role of the endometrial microbiome in embryo implantation and recurrent implantation failure. J Reprod Immunol 2024; 162:104192. [PMID: 38215650 DOI: 10.1016/j.jri.2024.104192] [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: 10/16/2023] [Revised: 12/21/2023] [Accepted: 01/01/2024] [Indexed: 01/14/2024]
Abstract
There is a suggested pathophysiology associated with endometrial microbiota in cases where repeated implantation failure of high-quality embryos is observed. However, there is a suspected association between endometrial microbiota and the pathogenesis of implantation failure. However, there is still a lack of agreement on the fundamental composition of the physiological microbiome within the uterine cavity. This is primarily due to various limitations in the studies conducted, including small sample sizes and variations in experimental designs. As a result, the impact of bacterial communities in the endometrium on human reproduction is still a subject of debate. In this discourse, we undertake a comprehensive examination of the existing body of research pertaining to the uterine microbiota and its intricate interplay with the process of embryo implantation.
Collapse
Affiliation(s)
| | - Sarmad Karimi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Arian Haghtalab
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Saman Taram
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Milad Hejazi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Sonia Sadeghpour
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran; Department of Obstetrics and Gynecology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Reza Pashaei
- Department of Internal Medicine, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Hojat Ghasemnejad-Berenji
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
46
|
Acuña-Castillo C, Escobar A, García-Gómez M, Bachelet VC, Huidobro-Toro JP, Sauma D, Barrera-Avalos C. P2X7 Receptor in Dendritic Cells and Macrophages: Implications in Antigen Presentation and T Lymphocyte Activation. Int J Mol Sci 2024; 25:2495. [PMID: 38473744 DOI: 10.3390/ijms25052495] [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: 12/07/2023] [Revised: 01/15/2024] [Accepted: 01/24/2024] [Indexed: 03/14/2024] Open
Abstract
The P2X7 receptor, a member of the P2X purinergic receptor family, is a non-selective ion channel. Over the years, it has been associated with various biological functions, from modulating to regulating inflammation. However, its emerging role in antigen presentation has captured the scientific community's attention. This function is essential for the immune system to identify and respond to external threats, such as pathogens and tumor cells, through T lymphocytes. New studies show that the P2X7 receptor is crucial for controlling how antigens are presented and how T cells are activated. These studies focus on antigen-presenting cells, like dendritic cells and macrophages. This review examines how the P2X7 receptor interferes with effective antigen presentation and activates T cells and discusses the fundamental mechanisms that can affect the immune response. Understanding these P2X7-mediated processes in great detail opens up exciting opportunities to create new immunological therapies.
Collapse
Affiliation(s)
- Claudio Acuña-Castillo
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 9160000, Chile
- Centro de Biotecnología Acuícola, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 9160000, Chile
| | - Alejandro Escobar
- Laboratorio Biología Celular y Molecular, Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 8380000, Chile
| | - Moira García-Gómez
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, Chile
| | - Vivienne C Bachelet
- Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Santiago 9160000, Chile
| | - Juan Pablo Huidobro-Toro
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 9160000, Chile
| | - Daniela Sauma
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, Chile
- Centro Ciencia & Vida, Av. Del Valle Norte 725, Huechuraba 8580000, Chile
| | - Carlos Barrera-Avalos
- Centro de Biotecnología Acuícola, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 9160000, Chile
| |
Collapse
|
47
|
Kamel M, Aleya S, Alsubih M, Aleya L. Microbiome Dynamics: A Paradigm Shift in Combatting Infectious Diseases. J Pers Med 2024; 14:217. [PMID: 38392650 PMCID: PMC10890469 DOI: 10.3390/jpm14020217] [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: 12/26/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/24/2024] Open
Abstract
Infectious diseases have long posed a significant threat to global health and require constant innovation in treatment approaches. However, recent groundbreaking research has shed light on a previously overlooked player in the pathogenesis of disease-the human microbiome. This review article addresses the intricate relationship between the microbiome and infectious diseases and unravels its role as a crucial mediator of host-pathogen interactions. We explore the remarkable potential of harnessing this dynamic ecosystem to develop innovative treatment strategies that could revolutionize the management of infectious diseases. By exploring the latest advances and emerging trends, this review aims to provide a new perspective on combating infectious diseases by targeting the microbiome.
Collapse
Affiliation(s)
- Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza 11221, Egypt
| | - Sami Aleya
- Faculty of Medecine, Université de Bourgogne Franche-Comté, Hauts-du-Chazal, 25030 Besançon, France;
| | - Majed Alsubih
- Department of Civil Engineering, King Khalid University, Guraiger, Abha 62529, Saudi Arabia;
| | - Lotfi Aleya
- Laboratoire de Chrono-Environnement, Université de Bourgogne Franche-Comté, UMR CNRS 6249, La Bouloie, 25030 Besançon, France;
| |
Collapse
|
48
|
Mamun MAA, Rakib A, Mandal M, Kumar S, Singla B, Singh UP. Polyphenols: Role in Modulating Immune Function and Obesity. Biomolecules 2024; 14:221. [PMID: 38397458 PMCID: PMC10887194 DOI: 10.3390/biom14020221] [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: 12/30/2023] [Revised: 02/02/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Polyphenols, long-used components of medicinal plants, have drawn great interest in recent years as potential therapeutic agents because of their safety, efficacy, and wide range of biological effects. Approximately 75% of the world's population still use plant-based medicinal compounds, indicating the ongoing significance of phytochemicals for human health. This study emphasizes the growing body of research investigating the anti-adipogenic and anti-obesity functions of polyphenols. The functions of polyphenols, including phenylpropanoids, flavonoids, terpenoids, alkaloids, glycosides, and phenolic acids, are distinct due to changes in chemical diversity and structural characteristics. This review methodically investigates the mechanisms by which naturally occurring polyphenols mediate obesity and metabolic function in immunomodulation. To this end, hormonal control of hunger has the potential to inhibit pro-obesity enzymes such as pancreatic lipase, the promotion of energy expenditure, and the modulation of adipocytokine production. Specifically, polyphenols affect insulin, a hormone that is essential for regulating blood sugar, and they also play a role, in part, in a complex web of factors that affect the progression of obesity. This review also explores the immunomodulatory properties of polyphenols, providing insight into their ability to improve immune function and the effects of polyphenols on gut health, improving the number of commensal bacteria, cytokine production suppression, and immune cell mediation, including natural killer cells and macrophages. Taken together, continuous studies are required to understand the prudent and precise mechanisms underlying polyphenols' therapeutic potential in obesity and immunomodulation. In the interim, this review emphasizes a holistic approach to health and promotes the consumption of a wide range of foods and drinks high in polyphenols. This review lays the groundwork for future developments, indicating that the components of polyphenols and their derivatives may provide the answer to urgent worldwide health issues. This compilation of the body of knowledge paves the way for future discoveries in the global treatment of pressing health concerns in obesity and metabolic diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Udai P. Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163, USA; (M.A.A.M.); (A.R.); (M.M.); (S.K.); (B.S.)
| |
Collapse
|
49
|
Altmäe S, Plaza-Florido A, Esteban FJ, Anguita-Ruiz A, Krjutškov K, Katayama S, Einarsdottir E, Kere J, Radom-Aizik S, Ortega FB. Effects of exercise on whole-blood transcriptome profile in children with overweight/obesity. Am J Hum Biol 2024; 36:e23983. [PMID: 37715654 PMCID: PMC11482632 DOI: 10.1002/ajhb.23983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/18/2023] Open
Abstract
BACKGROUND The current knowledge about the molecular mechanisms underlying the health benefits of exercise is still limited, especially in childhood. We set out to investigate the effects of a 20-week exercise intervention on whole-blood transcriptome profile (RNA-seq) in children with overweight/obesity. METHODS Twenty-four children (10.21 ± 1.33 years, 46% girls) with overweight/obesity, were randomized to either a 20-week exercise program (intervention group; n = 10), or to a no-exercise control group (n = 14). Whole-blood transcriptome profile was analyzed using RNA-seq by STRT technique with GlobinLock technology. RESULTS Following the 20-week exercise intervention program, 161 genes were differentially expressed between the exercise and the control groups among boys, and 121 genes among girls (p-value <0.05), while after multiple correction, no significant difference between exercise and control groups persisted in gene expression profiles (FDR >0.05). Genes enriched in GO processes and molecular pathways showed different immune response in boys (antigen processing and presentation, infections, and T cell receptor complex) and in girls (Fc epsilon RI signaling pathway) (FDR <0.05). CONCLUSION These results suggest that 20-week exercise intervention program alters the molecular pathways involved in immune processes in children with overweight/obesity.
Collapse
Affiliation(s)
- Signe Altmäe
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Abel Plaza-Florido
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada Granada, Granada, Spain
- Pediatric Exercise and Genomics Research Center, Department of Pediatrics, School of Medicine, University of California at Irvine, Irvine, California, USA
| | - Francisco J. Esteban
- Systems Biology Unit, Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Jaen, Spain
| | - Augusto Anguita-Ruiz
- Barcelona Institute for Global Health, ISGlobal Barcelona, Barcelona, Spain
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada Granada, Granada, Spain
- Center of Biomedical Research, Institute of Nutrition and Food Technology "José Mataix", University of Granada, Granada, Spain
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, Madrid, Spain
| | - Kaarel Krjutškov
- Competence Centre for Health Technologies, Tartu, Estonia
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Shintaro Katayama
- Folkhälsan Research Center, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Research Programs Unit, University of Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Elisabet Einarsdottir
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Science for Life Laboratory, Department of Gene Technology, KTH-Royal Institute of Technology, Solna, Sweden
| | - Juha Kere
- Folkhälsan Research Center, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Research Programs Unit, University of Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Shlomit Radom-Aizik
- Pediatric Exercise and Genomics Research Center, Department of Pediatrics, School of Medicine, University of California at Irvine, Irvine, California, USA
| | - Francisco B. Ortega
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada Granada, Granada, Spain
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
- CIBERobn Physiopathology of Obesity and Nutrition, Granada, Spain
| |
Collapse
|
50
|
Lim KH, Wang L, Dotse E, Wang M, Tiu CY, Wijanarko KJ, Wang X, Chow KT. TLR4 sensitizes plasmacytoid dendritic cells for antiviral response against SARS-CoV-2 coronavirus. J Leukoc Biol 2024; 115:190-200. [PMID: 37747799 DOI: 10.1093/jleuko/qiad111] [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: 05/03/2023] [Revised: 08/04/2023] [Accepted: 08/24/2023] [Indexed: 09/27/2023] Open
Abstract
Plasmacytoid dendritic cells are a rare subset of dendritic cells that exhibit antiviral functions in response to toll-like receptor 7/8 stimulations. Alternative toll-like receptors such as TLR4 have been known to be active in plasmacytoid dendritic cells for immune regulatory functions. However, it is unclear whether these toll-like receptors differentially activate plasmacytoid dendritic cells as compared with canonical toll-like receptor 7/8 stimulation. Here, we assessed alternative plasmacytoid dendritic cell activation states mediated by toll-like receptors other than endosomal toll-like receptors via the RNA sequencing approach. We found that toll-like receptor 4 stimulation induced a high degree of similarity in gene expression pattern to toll-like receptor 7/8 stimulation in plasmacytoid dendritic cells. Despite high resemblance to toll-like receptor 7/8, we discovered unique genes that were activated under toll-like receptor 4 activation only, as well as genes that were induced at a higher magnitude in comparison to toll-like receptor 7/8 activation. In comparison between toll-like receptor 4-activated plasmacytoid dendritic cells and conventional dendritic cells, we revealed that plasmacytoid dendritic cells and conventional dendritic cells expressed distinct gene sets, whereby conventional dendritic cells mostly favored antigen presentation functions for adaptive immune response regulation while plasmacytoid dendritic cells leaned toward immune response against infectious diseases. Last, we determined that toll-like receptor 4 activation sensitized plasmacytoid dendritic cells against SARS-CoV-2 (COVID-19) single-stranded RNA by enhancing antiviral-related responses and type I interferon production. These findings provided greater insights into the toll-like receptor 4 activation state in plasmacytoid dendritic cells, which can be beneficial for alternative therapeutic interventions involving plasmacytoid dendritic cells for various diseases.
Collapse
Affiliation(s)
- King Hoo Lim
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Kowloon, Hong Kong
| | - Lishi Wang
- Department of Surgery, Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T., Hong Kong
| | - Eunice Dotse
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Kowloon, Hong Kong
| | - Meijun Wang
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Kowloon, Hong Kong
| | - Cheuk Ying Tiu
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Kowloon, Hong Kong
| | - Kevin Julio Wijanarko
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Kowloon, Hong Kong
| | - Xin Wang
- Department of Surgery, Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T., Hong Kong
| | - Kwan T Chow
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Kowloon, Hong Kong
- Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Kowloon, Hong Kong
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, Guangdong, China
| |
Collapse
|