1
|
Niknejad A, Esmaealzadeh N, Peyrovinasab A, Sirouskabiri S, Gholami M, Pasha AVK, Shahri S, Büsselberg D, Abdolghaffari AH. Phytochemicals Alleviate Tumorigenesis by Regulation of M1/M2 Polarization: A Systematic Review of the Current Evidence. Phytother Res 2025. [PMID: 40393795 DOI: 10.1002/ptr.8522] [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: 07/27/2024] [Revised: 01/25/2025] [Accepted: 04/27/2025] [Indexed: 05/22/2025]
Abstract
Cancers are increasingly common and significantly impact patients' quality of life and longevity. The role of macrophages in tumorigenesis is critical, and natural compounds have long been recognized as valuable sources of bioactive agents for treating this condition. However, no systematic review has been performed on the role of phytochemicals impacting tumorigenesis by M1/M2 macrophage polarization. The aim of this study is to systematically review phytochemicals that relieve tumorigenesis by impacting M1/M2 macrophage polarization and investigate related signaling pathways. This systematic review adheres to PRISMA 2020 guidelines and statements. Scientific databases, MEDLINE, Scopus, and Web of Science, have been searched from inception to October 2023. This review includes English original articles on the role of phytochemicals, whole plant extracts, and polyherbal formulas in ameliorating tumorigenesis through M1/M2 polarization while excluding non-English articles, non-original research, and unrelated studies according to title, abstract, and full-text screening. Shreds of evidence were gathered from cellular and animal studies about the beneficial impacts of phytochemicals against tumorigenesis by impacting M1/M2 macrophage polarization. Critical assessment of in vitro and in vivo studies was performed by the CRIS and ARRIVE guidelines. Due to the high level of heterogeneity of the collected data, only a narrative synthesis was performed. Of 741 collected articles, only 35 remained. Polyphenols are the most highlighted group. Phytochemicals affect cytokines related to M1, such as CD80, CD86, CD64, and iNOS, and M2, like CXCR-1, CXCR-2, and TGF-β, in various cancer models. Together, these compounds exerted protective effects against tumorigenesis in preclinical cancer models. Furthermore, high-quality clinical experiments are recommended to cover the limitations of the current study, which are reliance on preclinical evidence, lack of clinical trials, and exclusion of non-English and grey literature.
Collapse
Affiliation(s)
- Amirhossein Niknejad
- Department of Toxicology & Pharmacology, TeMS.C., Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niusha Esmaealzadeh
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Traditional Pharmacy, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Traditional Persian Medicine and Complementary Medicine (PerCoMed) Student Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirreza Peyrovinasab
- Department of Toxicology & Pharmacology, TeMS.C., Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Shirin Sirouskabiri
- Department of Toxicology & Pharmacology, TeMS.C., Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahsa Gholami
- Department of Toxicology & Pharmacology, TeMS.C., Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Aytak Vahdat Khajeh Pasha
- Department of Toxicology & Pharmacology, TeMS.C., Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Saleh Shahri
- Department of Toxicology & Pharmacology, TeMS.C., Islamic Azad University, Tehran, Iran
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, TeMS.C., Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
2
|
Liu Y, Wang TT, Lu Y, Riaz M, Qyang Y. Cardiac macrophage: Insights from murine models to translational potential for human studies. J Mol Cell Cardiol 2025; 204:17-31. [PMID: 40354877 DOI: 10.1016/j.yjmcc.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Macrophages are a cell type that are known to play dynamic roles in acute and progressive pathology. They are highly attuned to their microenvironments throughout maturation, tailoring their functional responses according to the specific tissues in which they reside and their developmental origin. Cardiac macrophages (cMacs) have emerged as focal points of interest for their interactions with the unique electrical and mechanical stimuli of the heart, as well as for their role in maintaining cardiac homeostasis. Through an in-depth analysis of their origin, lineage, and functional significance, this review aims to shed light on cMacs' distinct contributions to both normal physiological maintenance as well as disease progression. Central to our discussion is the comparison of cMac characteristics between mouse and human models, highlighting current challenges and proposing novel experimental tools for deciphering cMac function within the intricate human cardiac microenvironments based on current murine studies. Our review offers valuable insights for identifying novel therapeutic targets and interventions tailored to the distinct roles of these immune cells in cardiovascular diseases (CVDs).
Collapse
Affiliation(s)
- Yufeng Liu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department, New Haven, CT, USA; Yale Biological and Biomedical Sciences, Graduate School of Arts and Sciences, Yale University, New Haven, CT, USA
| | - Tricia T Wang
- Yale Biological and Biomedical Sciences, Graduate School of Arts and Sciences, Yale University, New Haven, CT, USA
| | - Yinsheng Lu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department, New Haven, CT, USA; Yale Stem Cell Center, New Haven, CT, USA; Department of Mechanical Engineering and Materials Science, Graduate School of Arts and Sciences, Yale University, New Haven, CT, USA
| | - Muhammad Riaz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department, New Haven, CT, USA; Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Yibing Qyang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department, New Haven, CT, USA; Department of Pathology, Yale School of Medicine, New Haven, CT, USA; Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
3
|
Puri N, Sahane P, Phatale V, Khairnar P, Shukla S, Priyadarshinee A, Jain A, Srivastava S. Nano-chameleons: A review on cluster of differentiation-driven immune cell-engineered nanoarchitectonics for non-small cell lung cancer. Int J Biol Macromol 2025; 310:143440. [PMID: 40280523 DOI: 10.1016/j.ijbiomac.2025.143440] [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: 12/18/2024] [Revised: 03/26/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
Cancer, being one of the most outrageous diseases, contributed to 48 % of the mortality in 2022, with lung cancer leading the race with a 12.4 % incidence rate. Conventional treatment modalities like radio-, chemo-, photo-, and immunotherapy employing nanocarriers often face several setbacks, such as non-specific delivery, off-site toxicity, rapid opsonization via the host immune system, and greater tumor recurrence rates. Moreover, the heterogeneous variability in the tumor microenvironment is responsible for existing therapy failure. With the advent of biomimetic nanoparticles as a novel and intriguing platform, researchers have exploited the inherent functionalities of the Cluster of Differentiation proteins (CD) as cell surface biomarkers and imparted the nanocarriers with enhanced homologous tumor targetability, immune evasion capability, and stealth properties, paving the way for improved therapy and diagnosis. This article explores pathogenesis and the multifaceted role of immune cells in non-small cell lung cancer. Moreover, the agenda of this article is to shed light on biomimetic nanoarchitectonics with respect to their fabrication, evaluation, and applications unraveling their synergistic effect with conventional therapies. Further discussion mentions the hurdles in clinical translation with viable solutions. The regulatory bottlenecks underscore the need for a regulatory roadmap with respect to commercialization. We believe that biomimetic nanoarchitectonics will be a beacon of hope in warfare against lung cancer.
Collapse
Affiliation(s)
- Niharika Puri
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Prajakta Sahane
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Vivek Phatale
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Pooja Khairnar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Shalini Shukla
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Abhipsa Priyadarshinee
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Akshita Jain
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Telangana, India.
| |
Collapse
|
4
|
Salehi Moghaddam A, Bahrami M, Sarikhani E, Tutar R, Ertas YN, Tamimi F, Hedayatnia A, Jugie C, Savoji H, Qureshi AT, Rizwan M, Maduka CV, Ashammakhi N. Engineering the Immune Response to Biomaterials. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414724. [PMID: 40232044 PMCID: PMC12097135 DOI: 10.1002/advs.202414724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/29/2025] [Indexed: 04/16/2025]
Abstract
Biomaterials are increasingly used as implants in the body, but they often elicit tissue reactions due to the immune system recognizing them as foreign bodies. These reactions typically involve the activation of innate immunity and the initiation of an inflammatory response, which can persist as chronic inflammation, causing implant failure. To reduce these risks, various strategies have been developed to modify the material composition, surface characteristics, or mechanical properties of biomaterials. Moreover, bioactive materials have emerged as a new class of biomaterials that can induce desirable tissue responses and form a strong bond between the implant and the host tissue. In recent years, different immunomodulatory strategies have been incorporated into biomaterials as drug delivery systems. Furthermore, more advanced molecule and cell-based immunomodulators have been developed and integrated with biomaterials. These emerging strategies will enable better control of the immune response to biomaterials and improve the function and longevity of implants and, ultimately, the outcome of biomaterial-based therapies.
Collapse
Affiliation(s)
- Abolfazl Salehi Moghaddam
- Department of BioengineeringP.C. Rossin College of Engineering & Applied ScienceLehigh UniversityBethlehemPA18015USA
| | - Mehran Bahrami
- Department of Mechanical Engineering & MechanicsLehigh UniversityBethlehemPA18015USA
| | - Einollah Sarikhani
- Department of Nano and Chemical EngineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Rumeysa Tutar
- Department of ChemistryFaculty of Engineering, Istanbul University‐CerrahpaşaIstanbul, Avcılar34320Turkey
| | - Yavuz Nuri Ertas
- Department of Biomedical EngineeringErciyes UniversityKayseri38039Turkey
- ERNAM – Nanotechnology Research and Application CenterErciyes UniversityKayseri38039Turkey
| | - Faleh Tamimi
- College of Dental MedicineQatar University HealthQatar UniversityP.O. Box 2713DohaQatar
| | - Ali Hedayatnia
- Azrieli Research CenterCentre Hospitalier Universitaire Sainte‐JustineMontrealQCH3T 1C5Canada
- Institute of Biomedical Engineering, Department of Pharmacology and PhysiologyFaculty of MedicineMontrealQuebecH3T 1J4Canada
- Montreal TransMedTech InstituteiTMTMontrealQuebecH3T 1J4Canada
| | - Clotilde Jugie
- Azrieli Research CenterCentre Hospitalier Universitaire Sainte‐JustineMontrealQCH3T 1C5Canada
- Montreal TransMedTech InstituteiTMTMontrealQuebecH3T 1J4Canada
| | - Houman Savoji
- Azrieli Research CenterCentre Hospitalier Universitaire Sainte‐JustineMontrealQCH3T 1C5Canada
- Institute of Biomedical Engineering, Department of Pharmacology and PhysiologyFaculty of MedicineMontrealQuebecH3T 1J4Canada
- Montreal TransMedTech InstituteiTMTMontrealQuebecH3T 1J4Canada
| | - Asma Talib Qureshi
- Department of Biomedical EngineeringMichigan Technological UniversityHoughtonMI49931USA
| | - Muhammad Rizwan
- Department of Biomedical EngineeringMichigan Technological UniversityHoughtonMI49931USA
- Health Research InstituteMichigan Technological UniversityHoughtonMI49931USA
| | - Chima V. Maduka
- BioFrontiers InstituteUniversity of ColoradoBoulderCO80303USA
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME)Colleges of Engineering and Human MedicineMichigan State UniversityEast LansingMI48824USA
- Department of BioengineeringSamueli School of EngineeringUniversity of California Los AngelesLos AngelesCA90095USA
| |
Collapse
|
5
|
Yarlagadda S, Sheremeta CL, Cheung SW, Cuffe A, Grounds MD, Smythe ML, Noakes PG. Pharmacology and macrophage modulation of HPGDS inhibitor PK007 demonstrate reduced disease severity in DMD-affected muscles of the mdx mouse model. Skelet Muscle 2025; 15:11. [PMID: 40275384 PMCID: PMC12020277 DOI: 10.1186/s13395-025-00379-1] [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: 11/29/2024] [Accepted: 03/07/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Duchenne Muscular Dystrophy (DMD) is an X-linked disease characterised by chronic inflammation, progressive muscle damage, and muscle loss. Typically, initial symptoms affect lower limb muscles, including the gastrocnemius (GA), tibialis anterior (TA), and extensor digitorum longus (EDL). During the acute phase of DMD, particularly in boys aged 2-8 years, muscle damage resulting in necrosis (myonecrosis) involves a complex immune-inflammatory response. Prostaglandin D2 (PGD2) is recognised for enhancing pro-inflammatory chemokine and interleukin signalling and recruiting infiltrating immune cells such as pro-inflammatory macrophages, exacerbating myonecrosis. METHODS To reduce levels of PGD2, a novel hematopoietic prostaglandin D2 synthase (HPGDS) inhibitor, PK007, was characterised (i) for potency and pharmacokinetic profiles and then tested in the mdx mouse model of DMD during the acute early onset of disease progression. Juvenile mdx and wild type (WT) C57Bl/10Scsn mice were orally treated with PK007 and control vehicle solution for 10 days, from postnatal day 18 to 28. This builds upon a previous study with PK007 with (ii) additional analyses of disease progression assessed for muscle grip strength, metabolic and locomotor activity, myonecrosis in a wide range of muscles (3 from hindlimb, diaphragm, heart, and tongue), macrophage infiltration and pro-inflammatory cytokines (TNF-α, IL-1β and iNOS). RESULTS PK007 exhibited high potency (17.23 ± 12 nM), a long half-life (3.0 ± 0.3 h), and good oral bioavailability (81%). Treatment with PK007 decreased serum PGD2 levels (33.36%) in mdx mice compared to control (vehicle-treated) mdx mice. In mdx mice (compared with controls), PK007 enhanced grip strength (69.05% increase) and improved locomotor activity (69.05% increase). Histological analysis revealed a significant reduction in the total myonecrotic area in PK007-treated GA (49.75%), TA (73.87%), EDL (60.31%), diaphragm (48.02%), and tongue (37.93%) muscles of mdx mice (compared with controls). Additionally, PK007 decreased macrophage cell area by 55.56% in GA and 47.83% in EDL muscles. Further expression of pro-inflammatory cytokines and enzymes such as TNF-α, IL-1β and iNOS were significantly reduced in PK007 treated mice. These results demonstrate that PK007 significantly reduces the inflammatory response, protects muscles from necrosis and increases strength in juvenile mdx mice. CONCLUSION This study lays a strong foundation for progressing the use of HPDGS inhibitors such as PK007, which specifically inhibit PGD2 and reduce inflammation, as a viable therapeutic approach for DMD. This approach protects dystrophic muscles from necrosis and reduces the severity of this debilitating disease, improving outcomes and quality of life.
Collapse
Affiliation(s)
- Sai Yarlagadda
- School of Biomedical Sciences, Faculty of Medicine, the University of Queensland, Brisbane, QLD, 4072, Australia
- Institute for Molecular Bioscience, the University of Queensland, Brisbane, QLD, 4072, Australia
| | - Chynna-Loren Sheremeta
- School of Biomedical Sciences, Faculty of Medicine, the University of Queensland, Brisbane, QLD, 4072, Australia
- Institute for Molecular Bioscience, the University of Queensland, Brisbane, QLD, 4072, Australia
| | - Sang Won Cheung
- School of Biomedical Sciences, Faculty of Medicine, the University of Queensland, Brisbane, QLD, 4072, Australia
| | - Alison Cuffe
- School of Biomedical Sciences, Faculty of Medicine, the University of Queensland, Brisbane, QLD, 4072, Australia
| | - Miranda D Grounds
- School of Human Biology, the University of Western Australia, Perth, WA, 6009, Australia
| | - Mark L Smythe
- Institute for Molecular Bioscience, the University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Peter G Noakes
- School of Biomedical Sciences, Faculty of Medicine, the University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
6
|
Matsuoka M, Soria SA, Pires JR, Sant'Ana ACP, Freire M. Natural and induced immune responses in oral cavity and saliva. BMC Immunol 2025; 26:34. [PMID: 40251519 PMCID: PMC12007159 DOI: 10.1186/s12865-025-00713-8] [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: 11/22/2024] [Accepted: 04/07/2025] [Indexed: 04/20/2025] Open
Abstract
This review comprehensively explores the intricate immune responses within the oral cavity, emphasizing the pivotal role of saliva in maintaining both oral and systemic health. Saliva, a complex biofluid, functions as a dynamic barrier against pathogens, housing diverse cellular components including epithelial cells, neutrophils, monocytes, dendritic cells, and lymphocytes, which collectively contribute to robust innate and adaptive immune responses. It acts as a physical and immunological barrier, providing the first line of defense against pathogens. The multifaceted protective mechanisms of salivary proteins, cytokines, and immunoglobulins, particularly secretory IgA (SIgA), are elucidated. We explore the natural and induced immune responses in saliva, focusing on its cellular and molecular composition. In addition to saliva, we highlight the significance of a serum-like fluid, the gingival crevicular fluid (GCF), in periodontal health and disease, and its potential as a diagnostic tool. Additionally, the review delves into the impact of diseases such as periodontitis, oral cancer, type 2 diabetes, and lupus on salivary immune responses, highlighting the potential of saliva as a non-invasive diagnostic tool for both oral and systemic conditions. We describe how oral tissue and the biofluid responds to diseases, including considerations to periodontal tissue health and in disease periodontitis. By examining the interplay between oral and systemic health through the oral-systemic axis, this review underscores the significance of salivary immune mechanisms in overall well-being and disease pathogenesis, emphasizing the importance of salivary mechanisms across the body.
Collapse
Affiliation(s)
- Michele Matsuoka
- Department of Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA, 92037, USA
| | - Salim Abraham Soria
- Department of Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA, 92037, USA
| | - Julien Rodrigues Pires
- Department of Periodontology, Bauru School of Dentistry, University of São Paulo, Bauru, 17012-901, Brazil
| | | | - Marcelo Freire
- Department of Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA, 92037, USA.
- Division of Infectious Diseases and Global Public Health Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
7
|
Ramushu P, Mangoakoane DD, Makola RT, Matsebatlela TM. Lithium Induces Oxidative Stress, Apoptotic Cell Death, and G2/M Phase Cell Cycle Arrest in A549 Lung Cancer Cells. Molecules 2025; 30:1797. [PMID: 40333785 PMCID: PMC12029159 DOI: 10.3390/molecules30081797] [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: 03/14/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025] Open
Abstract
Lithium has been identified more than six decades ago as a preferred treatment option for manic depression. Due to its affordability, stability, minimal side effects, and immunomodulatory effects, recent studies on lithium have focused on its potential anticancer properties and possible mechanisms of action. Lung cancer ranks the highest as the main cause of death in males and has high mortality rates with low survival rates. In this study, lung adenocarcinoma (A549) cells were treated with various concentrations of lithium chloride to evaluate its inflammatory and anticancer properties. The in vitro cytotoxic effects of lithium chloride were assessed using the MTT [3-(4, 5-dimethythiazol-2-yl)-2, 5-diphenyltetrazolium bromide] assay, Muse® cell death, and cell cycle analysis. The nitric oxide and oxidative stress flow cytometry Muse® assays were used to monitor inflammation profiles of lithium-treated lung adenocarcinoma cells. The MTT viability assay showed the safe use of LiCl on the noncancerous RAW 264.7 macrophage cells below a concentration of 40 mM. Lithium reduced cell viability, induced late apoptotic cell death, and disrupted normal cell cycle progression in a dose-dependent manner, leading to cell cycle arrest in the S and G2/M phases of A549 cells. The induction of cell death by lithium in A549 cells is accompanied by increased ROS and nitric oxide production. This study shows that lithium chloride possesses some immunomodulatory cytotoxic effects on A549 lung cancer cells and can be further investigated for use in lung cancer treatment.
Collapse
Affiliation(s)
- Pearl Ramushu
- Department of Biochemistry, Microbiology and Biotechnology, Faculty of Science and Agriculture, University of Limpopo, P/bag x1106, Sovenga 0727, South Africa
| | | | | | | |
Collapse
|
8
|
Santurio DS, Barros LRC, Glauche I, Fassoni AC. Mathematical modeling unveils the timeline of CAR-T cell therapy and macrophage-mediated cytokine release syndrome. PLoS Comput Biol 2025; 21:e1012908. [PMID: 40203243 PMCID: PMC11981663 DOI: 10.1371/journal.pcbi.1012908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/24/2025] [Indexed: 04/11/2025] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy holds significant potential for cancer treatment, although disease relapse and cytokine release syndrome (CRS) remain as frequent clinical challenges. To better understand the mechanisms underlying the temporal dynamics of CAR-T cell therapy response and CRS, we developed a novel multi-layer mathematical model incorporating antigen-mediated CAR-T cell expansion, antigen-negative resistance, and macrophage-associated cytokine release. Three key mechanisms of macrophage activation are considered: release of damage-associated molecular patterns, antigen-binding mediated activation, and CD40-CD40L contact. The model accurately describes 25 patient time courses with different responses and IL-6 cytokine kinetics. We successfully link the dynamic shape of the response to interpretable model parameters and investigate the influence of CAR-T cell dose and initial tumor burden on the occurrence of cytokine release and treatment outcome. By disentangling the timeline of macrophage activation, the model identified distinct contributions of each activation mechanism, suggesting the CD40-CD40L axis as a major driver of cytokine release and a clinically feasible target to control the activation process and modulate cytokine peak height. Our multi-layer model provides a comprehensive framework for understanding the complex interactions between CAR-T cells, tumor cells, and macrophages during therapy.
Collapse
Affiliation(s)
| | | | - Ingmar Glauche
- Institute for Medical Informatics and Biometry, Technische Universität Dresden, Dresden, Germany
| | - Artur c Fassoni
- Institute for Medical Informatics and Biometry, Technische Universität Dresden, Dresden, Germany
- Instituto de Matemática e Computação, Universidade Federal de Itajubá, Itajubá, Brazil
| |
Collapse
|
9
|
Li S, Zhou X, Duan Q, Niu S, Li P, Feng Y, Zhang Y, Xu X, Gong SP, Cao H. Autophagy and Its Association with Macrophages in Clonal Hematopoiesis Leading to Atherosclerosis. Int J Mol Sci 2025; 26:3252. [PMID: 40244103 PMCID: PMC11989900 DOI: 10.3390/ijms26073252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Atherosclerosis, a chronic inflammatory disease characterized by lipid accumulation and immune cell infiltration, is linked to plaque formation and cardiovascular events. While traditionally associated with lipid metabolism and endothelial dysfunction, recent research highlights the roles of autophagy and clonal hematopoiesis (CH) in its pathogenesis. Autophagy, a cellular process crucial for degrading damaged components, regulates macrophage homeostasis and inflammation, both of which are pivotal in atherosclerosis. In macrophages, autophagy influences lipid metabolism, cytokine regulation, and oxidative stress, helping to prevent plaque instability. Defective autophagy exacerbates inflammation, impairs cholesterol efflux, and accelerates disease progression. Additionally, autophagic processes in endothelial cells and smooth muscle cells further contribute to atherosclerotic pathology. Recent studies also emphasize the interplay between autophagy and CH, wherein somatic mutations in genes like TET2, JAK2, and DNMT3A drive immune cell expansion and enhance inflammatory responses in atherosclerotic plaques. These mutations modify macrophage function, intensifying the inflammatory environment and accelerating atherosclerosis. Chaperone-mediated autophagy (CMA), a selective form of autophagy, also plays a critical role in regulating macrophage inflammation by degrading pro-inflammatory cytokines and oxidized low-density lipoprotein (ox-LDL). Impaired CMA activity leads to the accumulation of these substrates, activating the NLRP3 inflammasome and worsening inflammation. Preclinical studies suggest that pharmacologically activating CMA may mitigate atherosclerosis progression. In animal models, reduced CMA activity accelerates plaque instability and increases inflammation. This review highlights the importance of autophagic regulation in macrophages, focusing on its role in inflammation, plaque formation, and the contributions of CH. Building upon current advances, we propose a hypothesis in which autophagy, programmed cell death, and clonal hematopoiesis form a critical intrinsic axis that modulates the fundamental functions of macrophages, playing a complex role in the development of atherosclerosis. Understanding these mechanisms offers potential therapeutic strategies targeting autophagy and inflammation to reduce the burden of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Shuanhu Li
- Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine of Shaanxi Administration of Traditional Chinese Medicine, Engineering Research Center of Brain Health Industry of Chinese Medicine, Pharmacology of Chinese Medicine, Shaanxi University of Chinese Medicine, University Government Committee of Shaanxi Province, Xianyang 712046, China;
| | - Xin Zhou
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Qinchun Duan
- Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences, Shaanxi Normal University, Xi’an 710062, China; (Q.D.); or (X.X.)
| | - Shukun Niu
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Pengquan Li
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Yihan Feng
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Ye Zhang
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Xuehong Xu
- Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences, Shaanxi Normal University, Xi’an 710062, China; (Q.D.); or (X.X.)
| | - Shou-Ping Gong
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Huiling Cao
- Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine of Shaanxi Administration of Traditional Chinese Medicine, Engineering Research Center of Brain Health Industry of Chinese Medicine, Pharmacology of Chinese Medicine, Shaanxi University of Chinese Medicine, University Government Committee of Shaanxi Province, Xianyang 712046, China;
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| |
Collapse
|
10
|
Mahmoud ME, Tingley D, Faizal A, Ghaffar A, Azhar M, Salman D, Isham IM, Abdul-Careem MF. Cyclooxygenase-2/Prostaglandin E2 Pathway Facilitates Infectious Bronchitis Virus-Induced Necroptosis in Chicken Macrophages, a Caspase-Independent Cell Death. Viruses 2025; 17:503. [PMID: 40284946 PMCID: PMC12030959 DOI: 10.3390/v17040503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/15/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Infectious bronchitis virus (IBV) poses a major challenge to poultry health and productivity. This study examined how inflammatory cell death pathways influence the replication and pathogenesis of two IBV strains-respiratory Connecticut (Conn) A5968 and nephropathogenic Delmarva (DMV)/1639-in chicken macrophages. Low serum conditions enhanced viral replication, reduced cell viability, and promoted apoptosis and necroptosis, with DMV/1639 showing more pronounced effects. Modulation of the cyclooxygenase-2/prostaglandin E2 (COX-2/PGE2) pathway displayed strain-specific effects, mitigating necroptosis in DMV/1639-infected cells but exacerbating apoptosis and necroptosis in Conn A5968-infected cells. Broad caspase inhibition (z-VAD-FMK) reduced necroptosis, while selective caspase-1/4 inhibition heightened apoptotic responses. Caspase-8 inhibition selectively reduced necroptosis in DMV/1639 infections but increased apoptosis and necroptosis in Conn A5968 infections. NLRP3 inflammasome and RIPK1 inhibition decreased cell viability and increased apoptosis in both strains but had distinct effects on necroptosis. These findings reveal the strain-specific regulation of viral replication, apoptosis, and necroptosis, underscoring the intricate interplay between IBV and host inflammatory pathways. Understanding these mechanisms provides novel insights into IBV pathogenesis and highlights potential therapeutic strategies to mitigate its impact on poultry health.
Collapse
Affiliation(s)
- Motamed Elsayed Mahmoud
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; (M.E.M.); (A.F.); (A.G.); (M.A.); (D.S.); (I.M.I.)
- Department of Animal Husbandry, Faculty of Veterinary Medicine, Sohag University, Sohag 84524, Egypt
| | - Dylan Tingley
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; (M.E.M.); (A.F.); (A.G.); (M.A.); (D.S.); (I.M.I.)
| | - Akeel Faizal
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; (M.E.M.); (A.F.); (A.G.); (M.A.); (D.S.); (I.M.I.)
| | - Awais Ghaffar
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; (M.E.M.); (A.F.); (A.G.); (M.A.); (D.S.); (I.M.I.)
| | - Muhammed Azhar
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; (M.E.M.); (A.F.); (A.G.); (M.A.); (D.S.); (I.M.I.)
| | - Doaa Salman
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; (M.E.M.); (A.F.); (A.G.); (M.A.); (D.S.); (I.M.I.)
| | - Ishara M. Isham
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; (M.E.M.); (A.F.); (A.G.); (M.A.); (D.S.); (I.M.I.)
| | - Mohamed Faizal Abdul-Careem
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada; (M.E.M.); (A.F.); (A.G.); (M.A.); (D.S.); (I.M.I.)
| |
Collapse
|
11
|
Wood J, Palms D, Luu QT, Vasilev K, Bright R. Investigating Simulated Cellular Interactions on Nanostructured Surfaces with Antibacterial Properties: Insights from Force Curve Simulations. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:462. [PMID: 40137635 PMCID: PMC11944641 DOI: 10.3390/nano15060462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025]
Abstract
This study investigates the simulation of interactions between cells and antibacterial nanostructured surfaces. Understanding the physical interaction forces between cells and nanostructured surfaces is crucial for developing antibacterial materials, yet existing physical models are limited. Force simulation studies can simplify analysis by focusing on mechanical interactions while disregarding factors such as bacterial deformation and complex biochemical signals. To simulate these interactions, Atomic Force Microscopy (AFM) was employed to generate force curves, allowing precise monitoring of the interaction between a 5 µm spherical cantilever tip and titanium alloy (Ti6Al4V) surfaces. AFM uniquely enables customized approaches and retraction cycles, providing detailed insights into attractive-repulsive forces across different surface morphologies. Two nanostructured surfaces, created via hydrothermal etching using KOH and NaOH, were compared to a Ti6Al4V control surface. Results demonstrated significant changes in nanomechanical properties due to surface chemistry and morphology. The Ti6Al4V control surface exhibited a 44 ± 5 N/m stiffness, which decreased to 20 ± 3 N/m on KOH-etched nanostructured (NS) surfaces and 29 ± 4 N/m on NaOH-etched NS surfaces. Additionally, surface energy decreased by magnitude on nanostructured surfaces compared to the control. The nature of interaction forces also varied: short-range forces were predominant on KOH-etched surfaces, while NaOH-etched surfaces exhibited stronger long-range forces. These findings provide valuable insights into how nanostructure patterning influences cell-like interactions, offering potential applications in antibacterial surface design. By tailoring nanomechanical properties through specific etching techniques, biomaterial performance can be optimized for clinical applications, enhancing antibacterial efficacy and reducing microbial adhesion.
Collapse
Affiliation(s)
- Jonathan Wood
- Academic Unit of STEM, University of South Australia, Adelaide, SA 5095, Australia;
| | - Dennis Palms
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia; (D.P.); (Q.T.L.); (K.V.)
| | - Quan Trong Luu
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia; (D.P.); (Q.T.L.); (K.V.)
| | - Krasimir Vasilev
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia; (D.P.); (Q.T.L.); (K.V.)
| | - Richard Bright
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia; (D.P.); (Q.T.L.); (K.V.)
| |
Collapse
|
12
|
Hodge N, Tétreault MP. Epithelial Ikkβ deletion modulates immune responses and the IFNγ/CXCL9 axis during early esophageal carcinogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643566. [PMID: 40166246 PMCID: PMC11957055 DOI: 10.1101/2025.03.18.643566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Esophageal cancer is a major cause of cancer-related death, often preceded with chronic inflammation and injuries. The NFκB/IKKβ pathway plays a central role in inflammation, yet its role in early esophageal carcinogenesis remains unclear. This study investigated the role of epithelial IKKβ in early esophageal carcinogenesis. Mice were treated with the carcinogen 4-nitroquinoline-1-oxide (4-NQO) or a vehicle for one month to induce precancerous lesions. Esophagi were harvested and examined through histological, protein, flow cytometry, and RNA analyses. Histological analysis revealed that 4-NQO treatment led to increased inflammation, intraepithelial CD45+ immune cells, and elevated IKKβ phosphorylation levels. Mice with esophageal epithelial-specific Ikkβ deletion (4-NQO/Ikkβ EEC-KO ) showed delayed progression to a precancerous state, with reduced immune cell recruitment compared to 4-NQO/controls. Immunophenotyping showed decreased recruitment of T cells, including CD4+, CD8+ and regulatory (Tregs) T cells, and increased recruitment of macrophages in 4-NQO/Ikkβ EEC-KO mice compared to 4-NQO/controls. RNA sequencing data identified 262 differentially expressed genes in 4-NQO/Ikkβ EEC-KO mice, implicating pathways related to inflammation and wound healing. Notably, the chemokine CXCL9, a T cell chemoattractant, was significantly upregulated in 4-NQO control mice, but not in 4-NQO/Ikkβ EEC-KO mice. Further analysis identified IFNγ as an upstream regulator of Cxcl9 expression, and neutralization of IFNγ reduced Cxcl9 expression levels in 4-NQO treated mice. Additionally, in vitro studies demonstrated that IFNγ upregulates Cxcl9 in an NF-κB dependent manner in esophageal keratinocytes. These findings suggest that epithelial IKKβ regulates the immune microenvironment in early esophageal carcinogenesis through the IFNγ/CXCL9 axis and influencing T cell recruitment and inflammatory responses.
Collapse
Affiliation(s)
- Nathan Hodge
- Department of Medicine, Gastroenterology and Hepatology Division, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611-3010, USA
| | - Marie-Pier Tétreault
- Department of Medicine, Gastroenterology and Hepatology Division, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611-3010, USA
| |
Collapse
|
13
|
Malik JA, Agrewala JN. Morphine's role in macrophage polarization: Exploring M1 and M2 dynamics and disease susceptibility. J Neuroimmunol 2025; 400:578534. [PMID: 39883986 DOI: 10.1016/j.jneuroim.2025.578534] [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: 08/27/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 02/01/2025]
Abstract
Morphine is a globally prevalent substance of misuse, renowned for its immunosuppressive effects mediated through opioid receptors expressed on immune cells. Macrophages are crucial antigen-presenting cells that fulfill diverse roles, such as antigen presentation, phagocytosis, wound healing, and disease protection. They are typically classified based on their activation states: M1 (proinflammatory), M2 (anti-inflammatory), and M0 (resting). Morphine significantly modulates immune responses and neuroinflammation, further complicating the landscape of opioid dependency and disease susceptibility. The association of macrophages under the influence of morphine needs to be understood under various diseased conditions. Several studies have been focused on investigating the impact of morphine on macrophage function and its implications in infectious diseases and brain-associated diseases. To light this subject, we have discussed recent advancements in understanding the influences between morphine, macrophage function, polarization, infection, brain tumors, and drug dependency. This article explores the complex relationship between morphine, macrophages, and related pathologies. Consequently, discussing deeper insights into these dynamics could guide effective treatments for substance abuse disorders.
Collapse
Affiliation(s)
- Jonaid Ahmad Malik
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar 140001, Punjab, India
| | - Javed N Agrewala
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar 140001, Punjab, India.
| |
Collapse
|
14
|
Suliman M, Saleh RO, Chandra M, Rasool KH, Jabir M, Jawad SF, Hasan TF, Singh M, Singh M, Singh A. Macrophage-derived lncRNAs in cancer: regulators of tumor progression and therapeutic targets. Med Oncol 2025; 42:91. [PMID: 40048034 DOI: 10.1007/s12032-025-02643-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Macrophages are key tumor microenvironment (TME) regulators, exhibiting remarkable plasticity that enables them to either suppress or promote cancer progression. Emerging evidence highlights the critical role of macrophage-derived long non-coding RNAs (lncRNAs) in shaping tumor immunity, influencing macrophage polarization, immune evasion, angiogenesis, metastasis, and therapy resistance. This review comprehensively elucidates the functional roles of M1- and M2-associated lncRNAs, detailing their molecular mechanisms and impact on cancer pathogenesis. In summary, elucidating the roles of lncRNAs derived from macrophages in cancer progression offers new avenues for therapeutic strategies, significantly improving patient outcomes in the fight against the disease. Further research into the functional significance of these lncRNAs and the development of targeted therapies is essential to harness their potential fully in clinical applications. We further explore their potential as biomarkers for cancer prognosis and therapeutic targets for modulating macrophage activity to enhance anti-cancer immunity. Targeting macrophage-derived lncRNAs represents a promising avenue for precision oncology, offering novel strategies to reshape the TME and improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Raed Obaid Saleh
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al Maarif, Anbar, Iraq.
| | - Muktesh Chandra
- Marwadi University Research Center, Department of Bioinformatics, Faculty of Engineering and Technology, Marwadi University, Rajkot, Gujarat, 360003, India
| | | | - Majid Jabir
- Department of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Sabrean F Jawad
- Department of Pharmacy, Al-Mustaqbal University College, 51001, Hillah, Babylon, Iraq
| | - Thikra F Hasan
- College of Health & Medical Technology, Uruk University, Baghdad, Iraq
| | - Mithilesh Singh
- Department of Pharmaceutical Chemistry, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Manmeet Singh
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| |
Collapse
|
15
|
Lin Y, Zhang X, Sun D, Wang Q, Dou S, Zhou Q. Decoding the corneal immune microenvironment in healthy and diabetic mice during corneal wound healing. Ocul Surf 2025; 37:68-79. [PMID: 40023495 DOI: 10.1016/j.jtos.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
Diabetic keratopathy (DK) is an underdiagnosed ocular complication of diabetes mellitus. The changes of ocular immune microenvironment contribute to the pathogenesis of DK, while precise mechanisms remain inadequately understood. Here, we employed single-cell RNA sequencing (scRNA-seq) to elucidate the transcriptional alterations of immune cells from diabetic and healthy control mouse corneas during homeostasis and wound healing. Unbiased clustering analysis unveiled 3 major cell subsets and 11 subdivided cell clusters, including T cells, monocyte lineages, and neutrophil subpopulations. The further sub-clustering analysis demonstrated that T cells exhibited cytotoxicity characteristics in both homeostasis and wound healing of diabetic cornea. Moreover, dendritic cells preferred the migratory and maturation phenotype and may recruit and maintain cytotoxic T cells. Macrophages in diabetic cornea preferred the pro-inflammatory M1 phenotype. Under injury conditions, diabetic corneal neutrophils exhibited a more mature and functional possession of neutrophil extracellular traps (NETs). Furthermore, cell-cell communication revealed that the immune cells exhibited hyperactivation and pro-inflammatory responses, while the monocyte lineages exhibited the activating effect on T cells in diabetic cornea. This study represents the inaugural effort to establish a comprehensive scRNA-Seq transcriptomic profile of corneal immune cells during wound healing in healthy and diabetic mice, which offers a valuable reference for subsequent investigations into the pathological roles of immune cells in DK.
Collapse
Affiliation(s)
- Yujing Lin
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Xiaowen Zhang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Di Sun
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Qun Wang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Shengqian Dou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China.
| | - Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China.
| |
Collapse
|
16
|
Gonzales G, Malka R, Bizios R, Dion GR, Guda T. Burn inhalation injury and intubation with dexamethasone-eluting endotracheal tubes modulate local microbiome and alter airway inflammation. Front Bioeng Biotechnol 2025; 13:1524013. [PMID: 40078791 PMCID: PMC11897493 DOI: 10.3389/fbioe.2025.1524013] [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: 11/06/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Background Inhalation injuries, caused by exposure to extreme heat and chemical irritants, lead to complications with speaking, swallowing, and breathing. This study investigates the effects of thermal injury and endotracheal tube (ETT) placement on the airway microbiome and inflammatory response. A secondary aim is to assess the impact of localized dexamethasone delivery via a drug-eluting ETT to reduce laryngeal scarring. Methods Inhalation injury was developed in swine by administering heated air (150°C-160°C) under endoscopic visualization. Following injury, segments of regular or dexamethasone-loaded endotracheal tubes (ETTs) were placed in the injured airways for 3 or 7 days. Computed tomography (CT) scans were used to assess airway narrowing post-injury. Biofilm formation on the ETTs was investigated using micro-CT and microscopy. The airway microbiome was analyzed via 16S rRNA sequencing. Inflammatory markers were quantified using an immunoassay and macrophage populations in laryngeal tissue were assessed with CD86 and CD206 staining. Tracheal tissues were also histologically examined for epithelial thickness, collagen area, and mucin production. Results CT scans confirmed airway narrowing post-injury, particularly around ETT sites. Biofilm formation was more extensive on dexamethasone-coated ETTs at later timepoints. Beta diversity analysis revealed significant shifts in microbial composition related to ETT type (R2 = 0.04, p < 0.05) and duration of placement (R2 = 0.22, p < 0.05). Differential abundance analysis demonstrated significant positive log fold changes in genera such as Bergeriella, Peptostreptococcus, and Bacteriodes with thermal injury over time. Inflammatory markers IFN-γ, IL-4, and IL-1β were elevated in dexamethasone-ETT groups at 3 days, then decreased by 7 days. Macrophage markers CD86 and CD206 were significantly greater in dexamethasone groups compared to regular ETT groups at 7 days (p = 0.002 and p = 0.0213, respectively). Epithelial thickness was significantly greater with regular ETT placement compared to dexamethasone ETT placement in the burn-injured airway at 3 days (p = 0.027). Conclusion Thermal inhalation injury and ETT placement significantly impact airway inflammation, structural integrity, and microbiome composition. Dexamethasone-eluting ETTs, intended to reduce inflammation, increased biofilm formation and elevated cytokine levels, suggesting complex interactions between the drug coating and the host immune response. The airway microbiome shifted significantly with specific taxa thriving in the inflamed environment.
Collapse
Affiliation(s)
- Gabriela Gonzales
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX, United States
| | - Ronit Malka
- Department of Otolaryngology – Head and Neck Surgery, Brooke Army Medical Center JBSA Fort Sam Houston, San Antonio, TX, United States
| | - Rena Bizios
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX, United States
| | - Gregory R. Dion
- Department of Otolaryngology – Head and Neck Surgery, University of Cincinnati, Cincinnati, OH, United States
| | - Teja Guda
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX, United States
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
17
|
Yang X, Fang R, Li X, Kong W, Jin Y, Jiao R, Liu Z, Zhang M, Peng Q, Zhang Y, Song N. Engineered Nanovesicles for the Precise and Noninvasive Treatment of Deep Osteomyelitis Caused by MRSA Infection with Enhanced Immune Response. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11795-11810. [PMID: 39945439 DOI: 10.1021/acsami.4c20893] [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: 02/28/2025]
Abstract
The clinical treatment of hospital-acquired persistent osteomyelitis caused by methicillin-resistant Staphylococcus aureus (MRSA) presents two major challenges: ineffective drug delivery into deep tissues and counteracting the rapid establishment of an immunosuppressive microenvironment. Indeed, MRSA can evade immunosurveillance and undermine both innate and adaptive immune responses. Herein, the engineered nanovesicles, functioning by combining sonodynamic therapy (SDT) with immune modulation, were constructed for the precise and noninvasive removal of MRSA in deep tissue and activation of the antimicrobial immune response using a newly engineered nanovesicle. Macrophage-derived M1 phenotypic microvesicles (M1-MW) internalized vancomycin-cross-linked micelles with the acoustic sensitizer indocyanine green (ICG) (VCG micelles). The vesicles of M1-MW were grafted with PEGylated mannose, allowing for targeted accumulation at the infection site. The VCG micelles were responsive to the highly reducing environment and released ICG to generate ROS after exposure to ultrasounds. This effect was combined with the presence of vancomycin to kill MRSA. In an osteomyelitis infection model, we observed an improved survival rate and reprogramming of macrophages to a pro-inflammatory M1 phenotype. The latter promoted T-cell activation and immune defense against MRSA-camouflaged homologous cell-transferred infections. Thus, our study presents a noninvasive and efficient treatment (VCG@MMW) for deep osteomyelitis with improved bacterial clearance and reduced risk of recurrence with enhanced immune response.
Collapse
Affiliation(s)
- Xingyue Yang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Ren Fang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Xiaotian Li
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Weihao Kong
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Yubao Jin
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Ruohan Jiao
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Zhenggong Liu
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Meiqi Zhang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Qixian Peng
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Yumiao Zhang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300350, P. R. China
| | - Ningning Song
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| |
Collapse
|
18
|
Fey RM, Billo A, Clister T, Doan KL, Berry EG, Tibbitts DC, Kulkarni RP. Personalization of Cancer Treatment: Exploring the Role of Chronotherapy in Immune Checkpoint Inhibitor Efficacy. Cancers (Basel) 2025; 17:732. [PMID: 40075580 PMCID: PMC11899640 DOI: 10.3390/cancers17050732] [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: 11/04/2024] [Revised: 02/01/2025] [Accepted: 02/15/2025] [Indexed: 03/14/2025] Open
Abstract
In the era of precision medicine, mounting evidence suggests that the time of therapy administration, or chronotherapy, has a great impact on treatment outcomes. Chronotherapy involves planning treatment timing by considering circadian rhythms, which are 24 h oscillations in behavior and physiology driven by synchronized molecular clocks throughout the body. The value of chronotherapy in cancer treatment is currently under investigation, notably in the effects of treatment timing on efficacy and side effects. Immune checkpoint inhibitor (ICI) therapy is a promising cancer treatment. However, many patients still experience disease progression or need to stop the therapy early due to side effects. There is accumulating evidence that the time of day at which ICI therapy is administered can have a substantial effect on ICI efficacy. Thus, it is important to investigate the intersections of circadian rhythms, chronotherapy, and ICI efficacy. In this review, we provide a brief overview of circadian rhythms in the context of immunity and cancer. Additionally, we outline current applications of chronotherapy for cancer treatment. We synthesize the 29 studies conducted to date that examine the impact of time-of-day administration on the efficacy of ICI therapy, its associated side effects, and sex differences in both efficacy and side effects. We also discuss potential mechanisms underlying these observed results. Finally, we highlight the challenges in this area and future directions for research, including the potential for a chronotherapeutic personalized medicine approach that tailors the time of ICI administration to individual patients' circadian rhythms.
Collapse
Affiliation(s)
- Rosalyn M. Fey
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Avery Billo
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Terri Clister
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Khanh L. Doan
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Elizabeth G. Berry
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Deanne C. Tibbitts
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rajan P. Kulkarni
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Portland, OR 97239, USA
- Operative Care Division, U.S. Department of Veterans Affairs Portland Health Care System, Portland, OR 97239, USA
| |
Collapse
|
19
|
Liu Y, Yang G, Liu M, Zhang Y, Xu H, Mazhar M. Cinnamaldehyde and its combination with deferoxamine ameliorate inflammation, ferroptosis and hematoma expansion after intracerebral hemorrhage in mice. J Neuroinflammation 2025; 22:45. [PMID: 39985048 PMCID: PMC11846400 DOI: 10.1186/s12974-025-03373-y] [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: 11/06/2024] [Accepted: 02/10/2025] [Indexed: 02/23/2025] Open
Abstract
Intracerebral hemorrhage (ICH) is a most serious type of hemorrhagic stroke with a continuously rising incidence globally, without effective cure available. The underlying mechanisms driving brain injury are complex and include inflammation, oxidative stress, glutamate excitotoxicity, membrane damage, lipid peroxidation, ferroptosis and other cellular death modes. Hematoma clearance is the key to limit brain damage and foster the recovery process. The quest for effective ICH remedies is continuing and strategically evolving with the expansion of knowledge and understanding of target mechanisms and novel lead compounds. In this study, we have investigated the effects of cinnamaldehyde after ICH as an individual treatment as well as in combination with deferoxamine. The autologous blood injection model was employed using C57BL/6 mice. Following 2 h of ICH induction, animals received IP injection once per day for three days; normal saline in ICH model group, cinnamaldehyde, deferoxamine, and combined cinnamaldehyde and deferoxamine in respective groups. Measurement of neurobehavioral scoring, markers of inflammation NFкB, TNFα, IL-1, IL6, iNOS; oxidative stress and ferroptosis GSH, TBARS, glutamate, choline containing phospholipids, GPX4, SLC7A11, SLC40A1, ACSL4; and hematoma clearance hemoglobin, haptoglobin, hemopexin, zonulin, CD163, LRP1, HO1, CD36, CD206, were investigated using ELISA, PCR, and western blot. Immunofluorescence for NeuN/SLC40A1, GFAP/GPX4, NeuN/HO1, Iba1/HO1 was also performed. We have found that cinnamaldehyde possess anti-inflammatory, antioxidant, anti-ferroptotic and hematoma limiting properties that were comparable to those obtained with deferoxamine. However, combination of cinnamaldehyde and deferoxamine demonstrated remarkable effectiveness in restoration of these parameters indicating their synergistic effect in ICH model.
Collapse
Affiliation(s)
- Yulin Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
- National Traditional Chinese Medicine Service Export Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Guoqiang Yang
- Department of Acupuncture and Rehabilitation, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Mengnan Liu
- Department of Cardiovascular Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Yuwei Zhang
- Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Houping Xu
- Department of Geriatrics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Maryam Mazhar
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China.
- National Traditional Chinese Medicine Service Export Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
20
|
Brown MN, Bryant DM, Bond BT, Smith HP, Smith RA, Mihalko WM. The Effects of Simulated Cobalt-Chromium-Molybdenum Wear Particles on a Macrophage-Lymphocyte Coculture for Evaluating Cellular Corrosion. J Arthroplasty 2025:S0883-5403(25)00148-2. [PMID: 39956493 DOI: 10.1016/j.arth.2025.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND This study examined the impact of simulated wear particles on inflammatory cell-induced corrosion (ICIC). METHODS A 30-day macrophage-lymphocyte coculture experiment was conducted using American Society of Testing and Materials F1537 cobalt-chromium-molybdenum disks, with activators and cobalt-chromium-molybdenum particles added at none, low (1:10), medium (1:100), and high (1:500) cell-particle ratios. Supernatants collected on days 10 and 30 were analyzed for tissue necrosis factor alpha (TNFα) and interleukin 6 (IL-6) levels via enzyme-linked immunosorbent assay. Disks were examined for ICIC damage using scanning electron microscopy, and the oxygen percentage on their surfaces was analyzed with energy-dispersive X-ray spectrometry and X-ray photoelectron spectrometry. RESULTS Most disks showed damage consistent with ICIC. Day 10 TNFα was higher in medium and high particle groups compared to groups without particles, while IL-6 was unexpectedly lower in those groups. On day 30, the activated medium particle group showed higher IL-6 than the nonactivated group. The energy-dispersive X-ray spectrometry showed no significant differences (P = 0.77), but X-ray photoelectron spectrometry results indicated significant differences (P < 0.0001) at high particle concentrations. CONCLUSIONS Overall, the data suggested that increased TNFα reflected a heightened inflammatory response, particles might temporarily inhibit IL-6 release, and there is likely a synergistic effect between activators and particles on cellular responses.
Collapse
Affiliation(s)
- Madison N Brown
- The University of Tennessee Health Science Center-Campbell Clinic, Department of Orthopaedic Surgery and Biomedical Engineering, Memphis, Tennessee
| | - Danielle M Bryant
- The University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee
| | - Bailey T Bond
- The University of Tennessee Health Science Center, College of Graduate Health Sciences, Memphis, Tennessee
| | - Harrison P Smith
- The University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee
| | - Richard A Smith
- The University of Tennessee Health Science Center, College of Graduate Health Sciences, Memphis, Tennessee
| | - William M Mihalko
- The University of Tennessee Health Science Center-Campbell Clinic, Department of Orthopaedic Surgery and Biomedical Engineering, Memphis, Tennessee
| |
Collapse
|
21
|
Sukubo NG, Bigini P, Morelli A. Nanocarriers and macrophage interaction: from a potential hurdle to an alternative therapeutic strategy. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2025; 16:97-118. [PMID: 39902342 PMCID: PMC11789677 DOI: 10.3762/bjnano.16.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 01/02/2025] [Indexed: 02/05/2025]
Abstract
In the coming decades, the development of nanocarriers (NCs) for targeted drug delivery will mark a significant advance in the field of pharmacology. NCs can improve drug solubility, ensure precise distribution, and enable passage across biological barriers. Despite these potential advantages, the interaction with many biological matrices, particularly with existing macrophages, must be considered. In this review, we will explore the dual role of macrophages in NC delivery, highlighting their physiological functions, the challenges posed by the mononuclear phagocyte system, and innovative strategies to exploit macrophage interactions for therapeutic advantage. Recent advancements in treating liver and lung diseases, particularly focusing on macrophage polarization and RNA-based therapies, have highlighted the potential developments in macrophage-NC interaction. Furthermore, we will delve into the intriguing potential of nanomedicine in neurology and traumatology, associated with macrophage interaction, and the exciting possibilities it holds for the future.
Collapse
Affiliation(s)
- Naths Grazia Sukubo
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, Italy
| | - Paolo Bigini
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milano, Italy
| | - Annalisa Morelli
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milano, Italy
| |
Collapse
|
22
|
Svadlakova T, Kolackova M, Kulich P, Kotoucek J, Rosecka M, Krejsek J, Fiala Z, Andrýs C. Human Primary Monocytes as a Model for in vitro Immunotoxicity Testing: Evaluation of the Regulatory Properties of TiO 2 Nanoparticles. Int J Nanomedicine 2025; 20:1171-1189. [PMID: 39902067 PMCID: PMC11789775 DOI: 10.2147/ijn.s498690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/07/2025] [Indexed: 02/05/2025] Open
Abstract
Introduction A critical step preceding the potential biomedical application of nanoparticles is the evaluation of their immunomodulatory effects. Such nanoparticles are expected to enter the bloodstream where they can be recognized and processed by circulating monocytes. Despite the required biocompatibility, this interaction can affect intracellular homeostasis and modulate physiological functions, particularly inflammation. This study focuses on titanium dioxide (TiO2) as an example of relatively low cytotoxic nanoparticles with potential biomedical use and aims to evaluate their possible modulatory effects on the inflammasome-based response in human primary monocytes. Methods Monocyte viability, phenotypic changes, and cytokine production were determined after exposure to TiO2 (diameter, 25 nm; P25) alone. In the case of the modulatory effects, we focused on NLRP3 activation. The production of IL-1β and IL-10 was evaluated after (a) simultaneous activation of monocytes with bacterial stimuli muramyl dipeptide (MDP), or lipopolysaccharide (LPS), and TiO2 (co-exposure model), (b) prior activation with TiO2 alone and subsequent exposure to bacterial stimuli MDP or LPS. The differentiation of TiO2-treated monocytes into macrophages and their polarization were also assessed. Results The selected TiO2 concentration range (30-120 µg/mL) did not induce any significant cytotoxic effects. The highest dose of TiO2 promoted monocyte survival and differentiation into macrophages, with the M2 subset being the most prevalent. Nanoparticles alone did not induce substantial production of inflammatory cytokines IL-1β, IL-6, or TNF-α. The immunomodulatory effect on NLRP3 depended on the type of costimulant used. While co-exposure of monocytes to MDP and TiO2 boosted NLRP3 activity, co-exposure to LPS and TiO2 inhibited NLRP3 by enhancing IL-10 release. The inhibitory effect of TiO2 on NLRP3 based on the promotion of IL-10 was confirmed in a post-exposure model for both costimulants. Conclusion This study confirmed a non-negligible modulatory effect on primary monocytes in their inflammasome-based response and differentiation ability.
Collapse
Affiliation(s)
- Tereza Svadlakova
- Department of Clinical Immunology and Allergology, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
- Department of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Martina Kolackova
- Department of Clinical Immunology and Allergology, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Pavel Kulich
- Department of Pharmacology and Toxicology, Veterinary Research Institute, Brno, Czech Republic
| | - Jan Kotoucek
- Department of Pharmacology and Toxicology, Veterinary Research Institute, Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Michaela Rosecka
- Department of Clinical Immunology and Allergology, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Jan Krejsek
- Department of Clinical Immunology and Allergology, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Zdeněk Fiala
- Department of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Ctirad Andrýs
- Department of Clinical Immunology and Allergology, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| |
Collapse
|
23
|
Hage SF, Bi DE, Kinkade S, Vera Cruz D, Srinath A, Jhaveri A, Romanos S, Bindal A, Lightle R, Little JC, Shenkar R, Alcazar-Felix RJ, Lee J, Stadnik A, Sidebottom A, Carroll TJ, Ji Y, Koskimaki J, Polster SP, Girard R, Awad IA. Circulating molecules reflect imaging biomarkers of hemorrhage in cerebral cavernous malformations. J Cereb Blood Flow Metab 2025:271678X251314366. [PMID: 39829356 PMCID: PMC11748132 DOI: 10.1177/0271678x251314366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
Increases in mean lesional iron content by quantitative susceptibility mapping (QSM) by ≥6% and/or vascular permeability by dynamic contrast enhanced quantitative perfusion (DCEQP) by ≥40% on MRI have been associated with new symptomatic hemorrhage (SH) in cerebral cavernous malformations (CCMs). It is not known if plasma biomarkers can reflect these changes within the lesion proper. This cohort study enrolled 46 CCM patients with SH in the prior year. Plasma samples, QSM and DCEQP were simultaneously acquired at the beginning and end of 60 one-year epochs of prospective follow-up. Plasma levels of 16 proteins and 12 metabolites linked to CCM hemorrhage were assessed by enzyme-linked immunosorbent assay and liquid-chromatography mass spectrometry, respectively. A weighted model combining the percent changes in plasma levels in roundabout guidance receptor-4, cluster of differentiation 14, thrombomodulin and acetyl-L-carnitine reflected a mean increase in QSM ≥ 6% (97.2% and 100% specificity/sensitivity, p = 3.1 × 10-13). A weighted combination of percent changes in plasma levels of endoglin, pipecolic acid, arachidonic acid and hypoxanthine correlated with an increase in mean DCEQP ≥40% (99.6% specificity and 100% sensitivity, p = 4.1 × 10-17). This is a first report linking with great accuracy changes of circulating molecules to imaging changes reflecting new SH during prospective follow-up of CCMs.
Collapse
Affiliation(s)
- Stephanie F Hage
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Dehua E Bi
- Department of Public Health Sciences, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Serena Kinkade
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Diana Vera Cruz
- Center for Research Informatics, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Abhinav Srinath
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Aditya Jhaveri
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Sharbel Romanos
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Akash Bindal
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Rhonda Lightle
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Jessica C Little
- Host-Microbe Metabolomics Facility, Duchossois Family Institute, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Robert Shenkar
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Roberto J Alcazar-Felix
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Justine Lee
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Agnieszka Stadnik
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Ashley Sidebottom
- Host-Microbe Metabolomics Facility, Duchossois Family Institute, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Timothy J Carroll
- Department of Diagnostic Radiology, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Yuan Ji
- Department of Public Health Sciences, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Janne Koskimaki
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Sean P Polster
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Issam A Awad
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| |
Collapse
|
24
|
Wadhonkar K, Das S, Subramanian R, Sk MH, Singh Y, Baig MS. The effect of cancer cell-derived exosomal proteins on macrophage polarization: An in-depth review. Exp Cell Res 2025; 444:114393. [PMID: 39710293 DOI: 10.1016/j.yexcr.2024.114393] [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: 10/04/2024] [Revised: 12/13/2024] [Accepted: 12/20/2024] [Indexed: 12/24/2024]
Abstract
Cancer is characterized by unregulated cell proliferation, enabling it to invade and spread to different organs and tissues in the body. Cancer progression is intricately influenced by the complex dynamics within the tumor microenvironment (TME). The TME is a composite and dynamic network comprising cancer cells and various immune cells, including tumor-associated macrophages. Exosomes facilitate the communication between different cancer cells as well as other types of cells. This review particularly focuses on exosomal proteins derived from different cancer cells in mounting the complex crosstalk between cells of cancer and macrophages within the TME. Most cancer-derived exosomal proteins polarize macrophages towards M2 phenotype, promoting cancer aggressiveness, while a few have role switching towards the M1 phenotype, inhibiting cancer proliferation, respectively. In this review, we summarize, for the first time, the dual impact of cancer cell-derived exosomal proteins on macrophage polarization and the associated signaling pathways, offering valuable insights for developing innovative therapeutic strategies against diverse cancer types.
Collapse
Affiliation(s)
- Khandu Wadhonkar
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Soumalya Das
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | | | - Mobbassar Hassan Sk
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK; Institute for Energy and Environmental Flows, University of Cambridge, Cambridge, UK
| | - Yashi Singh
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India.
| |
Collapse
|
25
|
Qiao T, Wen XH. Exploring gut microbiota as a novel therapeutic target in Crohn's disease: Insights and emerging strategies. World J Gastroenterol 2025; 31:100827. [PMID: 39811502 PMCID: PMC11684203 DOI: 10.3748/wjg.v31.i2.100827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/30/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Extensive research has investigated the etiology of Crohn's disease (CD), encompassing genetic predisposition, lifestyle factors, and environmental triggers. Recently, the gut microbiome, recognized as the human body's second-largest gene pool, has garnered significant attention for its crucial role in the pathogenesis of CD. This paper investigates the mechanisms underlying CD, focusing on the role of 'creeping fat' in disease progression and exploring emerging therapeutic strategies, including fecal microbiota transplantation, enteral nutrition, and therapeutic diets. Creeping fat has been identified as a unique pathological feature of CD and has recently been found to be associated with dysbiosis of the gut microbiome. We characterize this dysbiotic state by identifying key microbiome-bacteria, fungi, viruses, and archaea, and their contributions to CD pathogenesis. Additionally, this paper reviews contemporary therapies, emphasizing the potential of biological therapies like fecal microbiota transplantation and dietary interventions. By elucidating the complex interactions between host-microbiome dynamics and CD pathology, this article aims to advance our understanding of the disease and guide the development of more effective therapeutic strategies for managing CD.
Collapse
Affiliation(s)
- Tong Qiao
- Department of Clinical Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Xian-Hui Wen
- College of Life Science and Technology, Jinan University, Guangzhou 510632, Guangdong Province, China
| |
Collapse
|
26
|
Freitas IL, Macedo MF, Oliveira L, Oliveira P, do Vale A, dos Santos NM. AIP56, an AB toxin secreted by Photobacterium damselae subsp. piscicida, has tropism for myeloid cells. Front Immunol 2025; 15:1527088. [PMID: 39872526 PMCID: PMC11769971 DOI: 10.3389/fimmu.2024.1527088] [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: 11/12/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction The AB-type toxin AIP56 is a key virulence factor of Photobacterium damselae subsp. piscicida (Phdp), inducing apoptosis in fish immune cells. The discovery of AIP56-like and AIP56-related toxins in diverse organisms, including human-associated Vibrio strains, highlights the evolutionary conservation of this toxin family, suggesting that AIP56 and its homologs may share conserved receptors across species. These toxins have potential for biotechnological applications, such as therapeutic protein delivery and immune modulation. Methods Herein, the cell specificity of AIP56 for immune cells was characterized. The tropism of AIP56 for cells of the sea bass, mouse and human immune system was analyzed by following toxin internalization by flow cytometry and arrival of the toxin in the cytosol by evaluating the cleavage of NF-kB p65 by western blotting. Results Only a small population of sea bass neutrophils internalized AIP56, indicating that most of the neutrophilic destruction during Phdp infection and/or AIP56 intoxication does not result from the direct action of the toxin. Moreover, the cellular tropism of AIP56 for myeloid cells was observed in the three species, including its preference for macrophages. Further, mouse and human M0 and M2-like macrophages internalized more toxin than M1-like macrophages. Despite the limited interaction of lymphoid cells with AIP56, mouse B1-cells were able to internalize the toxin, possibly due to its myeloid features. Conclusion AIP56 has tropism for sea bass, mouse and human myeloid cells, with greater affinity for macrophages. This points to an evolutionary conservation of its receptor(s) and mechanism of action across species, raising the possibility that AIP56-like and -related toxins may also play a role in pathogenesis. These findings are relevant for both pathogenicity and biomedical contexts.
Collapse
Affiliation(s)
- Inês Lua Freitas
- Fish Immunology and Vaccinology Group, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Fish Immunology and Vaccinology Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- McBiology Doctoral Program, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Maria Fátima Macedo
- Cell Activation and Gene Expression, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Cell Activation and Gene Expression, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Liliana Oliveira
- Cell Activation and Gene Expression, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Cell Activation and Gene Expression, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Pedro Oliveira
- EPIUnit, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Ana do Vale
- Fish Immunology and Vaccinology Group, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Fish Immunology and Vaccinology Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Nuno M.S. dos Santos
- Fish Immunology and Vaccinology Group, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Fish Immunology and Vaccinology Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| |
Collapse
|
27
|
Li J, Ma W, Tang Z, Li Y, Zheng R, Xie Y, Li G. Macrophage‑driven pathogenesis in acute lung injury/acute respiratory disease syndrome: Harnessing natural products for therapeutic interventions (Review). Mol Med Rep 2025; 31:16. [PMID: 39513609 PMCID: PMC11551695 DOI: 10.3892/mmr.2024.13381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/27/2024] [Indexed: 11/15/2024] Open
Abstract
Acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) is a common respiratory disease characterized by hypoxemia and respiratory distress. It is associated with high morbidity and mortality. Due to the complex pathogenesis of ALI, the clinical management of patients with ALI/ARDS is challenging, resulting in numerous post‑treatment sequelae and compromising the quality of life of patients. Macrophages, as a class of innate immune cells, play an important role in ALI/ARDS. In recent years, the functions and phenotypes of macrophages have been better understood due to the development of flow cytometry, immunofluorescence, single‑cell sequencing and spatial genomics. However, no macrophage‑targeted drugs for the treatment of ALI/ARDS currently exist in clinical practice. Natural products are important for drug development, and it has been shown that numerous natural compounds from herbal medicine can alleviate ALI/ARDS caused by various factors by modulating macrophage abnormalities. In the present review, the natural products from herbal medicine that can modulate macrophage abnormalities in ALI/ARDS to treat ALI/ARDS are introduced, and their mechanisms of action, discovered in the previous five years (2019‑2024), are presented. This will provide novel ideas and directions for further research, to develop new drugs for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Jincun Li
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Wenyu Ma
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Zilei Tang
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yingming Li
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Ruiyu Zheng
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yuhuan Xie
- Yunnan Innovation Team of Application Research on Traditional Chinese Medicine Theory of Disease Prevention, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
- Yunnan Provincial University Key Laboratory of Aromatic Chinese Herb Research, Basic Medical School, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Gang Li
- Yunnan Provincial University Key Laboratory of Aromatic Chinese Herb Research, Basic Medical School, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
- Basic Medical School, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
28
|
Ayhan H, Sevin S, Karaaslan S, Ayaz F. Immunomodulatory effects of medicinal leech saliva extract on in vitro activated macrophages. Immunol Res 2024; 73:9. [PMID: 39671012 DOI: 10.1007/s12026-024-09575-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/21/2024] [Indexed: 12/14/2024]
Abstract
Leech therapy has been utilized in modern and traditional medicine. Leech saliva contains versatile peptides and molecules that can exert anti-microbial, anti-inflammatory, anti-coagulant, and analgesic activities on the patients. The active components and molecular mechanism of action of these components should be deciphered properly in order to generate biotechnological drug candidates by recombinant production of the leech saliva peptides. In our study, we conducted LC-MS/MS and proteomics analysis on the lyophilized leech saliva extract to determine the components of it. Moreover, this extract was tested on the in vitro-activated macrophages. The extract decreased the production of the pro-inflammatory cytokines by the activated mammalian macrophages compared to the positive control groups. These results suggest that the lyophilized leech saliva can be utilized as an anti-inflammatory biotechnological drug candidate against inflammatory and autoimmune disorders. In vitro studies will be conducted to further decipher the most active ingredients in the leech saliva. These active components will be tested on the animal models of the inflammatory and autoimmune disorders to show their drug potential.
Collapse
Affiliation(s)
- Hüseyin Ayhan
- Health Services Vocational School, Ankara Yıldırım Beyazıt University, Ankara, 06760, Turkey
| | - Sedat Sevin
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ankara University, Ankara, 06110, Turkey.
| | - Seyhan Karaaslan
- Oral and Dental Health Clinic, Ankara Bilkent City Hospital, Ankara, 06800, Turkey
| | - Furkan Ayaz
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Biruni University, Istanbul, 34010, Turkey.
| |
Collapse
|
29
|
Bolduan V, Palzer KA, Ries F, Busch N, Pautz A, Bros M. KSRP Deficiency Attenuates the Course of Pulmonary Aspergillosis and Is Associated with the Elevated Pathogen-Killing Activity of Innate Myeloid Immune Cells. Cells 2024; 13:2040. [PMID: 39768132 PMCID: PMC11674352 DOI: 10.3390/cells13242040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
The mRNA-binding protein KSRP (KH-type splicing regulatory protein) is known to modulate immune cell functions post-transcriptionally, e.g., by reducing the mRNA stability of cytokines. It is known that KSRP binds the AU-rich motifs (ARE) that are often located in the 3'-untranslated part of mRNA species, encoding dynamically regulated proteins as, for example, cytokines. Innate myeloid immune cells, such as polymorphonuclear neutrophils (PMNs) and macrophages (MACs), eliminate pathogens by multiple mechanisms, including phagocytosis and the secretion of chemo- and cytokines. Here, we investigated the role of KSRP in the phenotype and functions of both innate immune cell types in the mouse model of invasive pulmonary aspergillosis (IPA). Here, KSRP-/- mice showed lower levels of Aspergillus fumigatus conidia (AFC) and an increase in the frequencies of PMNs and MACs in the lungs. Our results showed that PMNs and MACs from KSRP-/- mice exhibited an enhanced phagocytic uptake of AFC, accompanied by increased ROS production in PMNs upon stimulation. A comparison of RNA sequencing data revealed that 64 genes related to inflammatory and immune responses were shared between PMNs and MACs. The majority of genes upregulated in PMNs were involved in metabolic processes, cell cycles, and DNA repair. Similarly, KSRP-deficient PMNs displayed reduced levels of apoptosis. In conclusion, our results indicate that KSRP serves as a critical negative regulator of PMN and MAC anti-pathogen activity.
Collapse
Affiliation(s)
- Vanessa Bolduan
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Kim-Alicia Palzer
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Frederic Ries
- Department of Hematology and Medical Oncology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Nora Busch
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Andrea Pautz
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| |
Collapse
|
30
|
Barilo J, Bouzeineddine NZ, Philippi A, Basta S. Polarized macrophage functions are affected differentially after CSF-1R inhibition with PLX5622. Eur J Pharmacol 2024; 984:177059. [PMID: 39419432 DOI: 10.1016/j.ejphar.2024.177059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/30/2024] [Accepted: 10/15/2024] [Indexed: 10/19/2024]
Abstract
PLX5622 is a colony stimulating factor 1 receptor (CSF-1R) inhibitor that is known to deplete microglial cells in vivo. Recently its effects on macrophages (Mφ) were also observed in vivo. Therefore, we performed this study to assess its in vitro effects on the differentiation and functions of polarized Mφ derived from different tissues. Our findings show that addition of PLX5622 early on after ex vivo isolation hinders Mφ differentiation and survival. However, its addition post Mφ differentiation did not significantly affect the viability. Furthermore, PLX5622 affects certain functions and degree of polarization of IL-4 (M2a) Mφ but not polarization of M1-like Mφ. Our study provides novel aspects on the application of PLX5622 to study Mφ functions in vitro, where polarization is affected by CSF-1R signalling and provides distinctive evidence to its ability to affect certain populations of Mφ during in vitro differentiation and maturation.
Collapse
Affiliation(s)
- Julia Barilo
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Nasry Zane Bouzeineddine
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Alecco Philippi
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Sam Basta
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada.
| |
Collapse
|
31
|
Gaur SK, Jain J, Chaudhary Y, Kaul R. Insights into the mechanism of Morbillivirus induced immune suppression. Virology 2024; 600:110212. [PMID: 39232265 DOI: 10.1016/j.virol.2024.110212] [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: 04/02/2024] [Revised: 07/26/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
Viruses enter the host cell, and various strategies are employed to evade the host immune system. These include overcoming the various components of the immune system, including modulation of the physical and chemical barriers, non-specific innate response and specific adaptive immune response. Morbilliviruses impose immune modulation by utilizing various approaches including hindering antigen presentation to T-Helper (TH) cells, hematopoiesis and suppression of effector molecule activities. These viruses can also impede the early stages of T cell activation. Despite the availability of effective vaccines, morbilliviruses are still a significant threat to mankind. After infection, they also induce a state of immune suppression in the host. The molecular mechanisms employed by morbilliviruses to induce the state of immune suppression in the infected host are still being investigated. This review is an attempt to summarize insights into some of the strategies adopted by morbilliviruses to mediate immune modulation in the host.
Collapse
Affiliation(s)
- Sharad Kumar Gaur
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India
| | - Juhi Jain
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India
| | - Yash Chaudhary
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India
| | - Rajeev Kaul
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India.
| |
Collapse
|
32
|
Niskala A, Heijman J, Dobrev D, Jespersen T, Saljic A. Targeting the NLRP3 inflammasome signalling for the management of atrial fibrillation. Br J Pharmacol 2024; 181:4939-4957. [PMID: 38877789 DOI: 10.1111/bph.16470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/12/2024] [Accepted: 05/04/2024] [Indexed: 06/16/2024] Open
Abstract
Inflammatory signalling via the nod-like receptor (NLR) family pyrin domain-containing protein-3 (NLRP3) inflammasome has recently been implicated in the pathophysiology of atrial fibrillation (AF). However, the precise role of the NLRP3 inflammasome in various cardiac cell types is poorly understood. Targeting components or products of the inflammasome and preventing their proinflammatory consequences may constitute novel therapeutic treatment strategies for AF. In this review, we summarise the current understanding of the role of the inflammasome in AF pathogenesis. We first review the NLRP3 inflammasome pathway and inflammatory signalling in cardiomyocytes, (myo)fibroblasts and immune cells, such as neutrophils, macrophages and monocytes. Because numerous compounds targeting NLRP3 signalling are currently in preclinical development, or undergoing clinical evaluation for other indications than AF, we subsequently review known therapeutics, such as colchicine and canakinumab, targeting the NLRP3 inflammasome and evaluate their potential for treating AF.
Collapse
Affiliation(s)
- Alisha Niskala
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jordi Heijman
- Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
- Gottfried Schatz Research Center, Division of Medical Physics & Biophysics, Medical University of Graz, Graz, Austria
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Medicine and Research Center, Montréal Heart Institute and University de Montréal, Montréal, Canada
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arnela Saljic
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Almeida Júnior ASD, Freitas Viana Leal MM, Marques DSC, Silva ALD, Souza Bezerra RD, Siqueira de Souza YF, Mendonça Silveira ME, Santos FA, Alves LC, de Lima Aires A, Cruz Filho IJD, do Carmo Alves de Lima M. Therapeutic potential of hydantoin and thiohydantoin compounds against Schistosoma mansoni: An integrated in vitro, DNA, ultrastructural, and ADMET in silico approach. Mol Biochem Parasitol 2024; 260:111646. [PMID: 38950658 DOI: 10.1016/j.molbiopara.2024.111646] [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: 02/10/2024] [Revised: 06/01/2024] [Accepted: 06/18/2024] [Indexed: 07/03/2024]
Abstract
The study aimed to conduct in vitro biological assessments of hydantoin and thiohydantoin compounds against mature Schistosoma mansoni worms, evaluate their cytotoxic effects and predict their pharmacokinetic parameters using computational methods. The compounds showed low in vitro cytotoxicity and were not considered hemolytic. Antiparasitic activity against adult S. mansoni worms was tested with all compounds at concentrations ranging from 200 to 6.25 μM. Compounds SC01, SC02, and SC03 exhibited low activity. Compounds SC04, SC05, SC06 and SC07 caused 100 % mortality within 24 h of incubation at a concentration of 100 and 200 μM. Thiohydantoin SC04 exhibited the highest activity, resulting in 100 % mortality after 24 h of incubation at a concentration of 50 μM and IC50 of 28 µM. In the ultrastructural analysis (SEM), the compound SC04 (200 µM) induced integumentary changes, formation of integumentary blisters, and destruction of tubercles and spicules. Therefore, the SC04 compound shows promise as an antiparasitic against S. mansoni.
Collapse
Affiliation(s)
- Antônio Sérgio de Almeida Júnior
- Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, Recife, PE 50740-600, Brazil
| | - Mayse Manuele Freitas Viana Leal
- Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, Recife, PE 50740-600, Brazil
| | - Diego Santa Clara Marques
- Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, Recife, PE 50740-600, Brazil.
| | - Anekécia Lauro da Silva
- Department of Medicine, Federal University of Vale do Rio São Francisco (UNIVASF), Avenida da Amizade, s/n, Sal Torrado, Paulo Afonso, BA 48605-780, Brazil
| | - Rafael de Souza Bezerra
- Department of Medicine, Federal University of Vale do Rio São Francisco (UNIVASF), Avenida da Amizade, s/n, Sal Torrado, Paulo Afonso, BA 48605-780, Brazil
| | - Yandra Flaviana Siqueira de Souza
- Department of Medicine, Federal University of Vale do Rio São Francisco (UNIVASF), Avenida da Amizade, s/n, Sal Torrado, Paulo Afonso, BA 48605-780, Brazil
| | - Maria Eduardade Mendonça Silveira
- Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, Recife, PE 50740-600, Brazil
| | - Fábio Ab Santos
- Aggeu Magalhães Institute. Oswaldo Cruz Foundation (IAM-FIOCRUZ), Cidade Universitária, Recife, PE 50670-420, Brazil
| | - Luiz Carlos Alves
- Aggeu Magalhães Institute. Oswaldo Cruz Foundation (IAM-FIOCRUZ), Cidade Universitária, Recife, PE 50670-420, Brazil
| | - André de Lima Aires
- Department of Tropical Medicine, Health Sciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, Recife, PE 50740-600, Brazil
| | - Iranildo José da Cruz Filho
- Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, Recife, PE 50740-600, Brazil
| | - Maria do Carmo Alves de Lima
- Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, Recife, PE 50740-600, Brazil
| |
Collapse
|
34
|
Bandaranayake UK, Sato H, Suzuki M. Development of molecular sensors based on fluorescent proteins for polarized macrophages identification. ANAL SCI 2024; 40:2133-2145. [PMID: 39235677 DOI: 10.1007/s44211-024-00649-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 08/06/2024] [Indexed: 09/06/2024]
Abstract
Macrophages are a type of white blood cells that play key roles in innate immune responses as a part of cellular immunity for host defence and tissue homeostasis. To perform diverse functions, macrophages show high plasticity by transforming to polarized states. They are mainly identified as unpolarized, pro-inflammatory and antiinflammatory states and termed as M0, M1 and M2 macrophages respectively. Discriminating polarized states is important due to strict implication with inflammatory conditions resulting in many diseases as chronic inflammation, neurodegeneration, and cancer etc. Many polarization protein markers have been identified and applied to investigate expression profiles through PCR and other techniques with antibodies. However, they are time and cost consuming and sometimes show insufficient performances. We focused on the mannose receptor (CD206) as representative marker of M2 macrophage recognising terminal mannose. We developed dose dependent mannosylated fluorescent proteins (FPs) by conjugations with mannose derivative for around 20 modifiable sites on FPs surfaces. Maximum modifications did not spoil various features of FPs. We found further sensitive and specific discriminations among M2, M1 and M0 macrophages after treating polarized macrophages with adequately conditioned FPs compared to already established approaches using anti CD206 antibody through flow cytometric analysis. These results might be derived from direct ligand utilizations and increased avidity due to multivalent bindings with abundantly modified multimeric FPs. Our strategy is simple but addresses disadvantages of preceding methods. Moreover, this strategy is applicable to detect other cell surface receptors as FPs can be modified with ligands or recognizable aptamer like molecules.
Collapse
Affiliation(s)
- Udari Kalpana Bandaranayake
- Graduate School of Science and Engineering, Saitama University, 255 Shimo-Okubo, Sakura-ku, Saitama, 338-8570, Japan
| | - Hiroki Sato
- Department of Cerebrovascular Surgery, International Medical Center, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1298, Japan
| | - Miho Suzuki
- Graduate School of Science and Engineering, Saitama University, 255 Shimo-Okubo, Sakura-ku, Saitama, 338-8570, Japan.
| |
Collapse
|
35
|
Venkataraman A, Kordic I, Li J, Zhang N, Bharadwaj NS, Fang Z, Das S, Coskun AF. Decoding senescence of aging single cells at the nexus of biomaterials, microfluidics, and spatial omics. NPJ AGING 2024; 10:57. [PMID: 39592596 PMCID: PMC11599402 DOI: 10.1038/s41514-024-00178-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
Aging has profound effects on the body, most notably an increase in the prevalence of several diseases. An important aging hallmark is the presence of senescent cells that no longer multiply nor die off properly. Another characteristic is an altered immune system that fails to properly self-surveil. In this multi-player aging process, cellular senescence induces a change in the secretory phenotype, known as senescence-associated secretory phenotype (SASP), of many cells with the intention of recruiting immune cells to accelerate the clearance of these damaged senescent cells. However, the SASP phenotype results in inducing secondary senescence of nearby cells, resulting in those cells becoming senescent, and improper immune activation resulting in a state of chronic inflammation, called inflammaging, in many diseases. Senescence in immune cells, termed immunosenescence, results in further dysregulation of the immune system. An interdisciplinary approach is needed to physiologically assess aging changes of the immune system at the cellular and tissue level. Thus, the intersection of biomaterials, microfluidics, and spatial omics has great potential to collectively model aging and immunosenescence. Each of these approaches mimics unique aspects of the body undergoes as a part of aging. This perspective highlights the key aspects of how biomaterials provide non-cellular cues to cell aging, microfluidics recapitulate flow-induced and multi-cellular dynamics, and spatial omics analyses dissect the coordination of several biomarkers of senescence as a function of cell interactions in distinct tissue environments. An overview of how senescence and immune dysregulation play a role in organ aging, cancer, wound healing, Alzheimer's, and osteoporosis is included. To illuminate the societal impact of aging, an increasing trend in anti-senescence and anti-aging interventions, including pharmacological interventions, medical procedures, and lifestyle changes is discussed, including further context of senescence.
Collapse
Affiliation(s)
- Abhijeet Venkataraman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - Ivan Kordic
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - JiaXun Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Nicholas Zhang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA
| | - Nivik Sanjay Bharadwaj
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Zhou Fang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Machine Learning Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sandip Das
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ahmet F Coskun
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA, 30332, USA.
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
36
|
Panichi V, Costantini S, Grasso M, Arciola CR, Dolzani P. Innate Immunity and Synovitis: Key Players in Osteoarthritis Progression. Int J Mol Sci 2024; 25:12082. [PMID: 39596150 PMCID: PMC11594236 DOI: 10.3390/ijms252212082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Osteoarthritis (OA) is a chronic progressive disease of the joint. Although representing the most frequent cause of disability in the elderly, OA remains partly obscure in its pathogenic mechanisms and is still the orphan of resolutive therapies. The concept of what was once considered a "wear and tear" of articular cartilage is now that of an inflammation-related disease that affects over time the whole joint. The attention is increasingly focused on the synovium. Even from the earliest clinical stages, synovial inflammation (or synovitis) is a crucial factor involved in OA progression and a major player in pain onset. The release of inflammatory molecules in the synovium mediates disease progression and worsening of clinical features. The activation of synovial tissue-resident cells recalls innate immunity cells from the bloodstream, creating a proinflammatory milieu that fuels and maintains a damaging condition of low-grade inflammation in the joint. In such a context, cellular and molecular inflammatory behaviors in the synovium could be the primum movens of the structural and functional alterations of the whole joint. This paper focuses on and discusses the involvement of innate immunity cells in synovitis and their role in the progression of OA.
Collapse
Affiliation(s)
- Veronica Panichi
- Laboratory of Immunorheumatology and Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Silvia Costantini
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40136 Bologna, Italy; (S.C.); (M.G.)
| | - Merimma Grasso
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40136 Bologna, Italy; (S.C.); (M.G.)
| | - Carla Renata Arciola
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40136 Bologna, Italy; (S.C.); (M.G.)
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Paolo Dolzani
- Laboratory of Immunorheumatology and Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| |
Collapse
|
37
|
Liu Y, Zhang J, Zhao H, Zhong F, Li J, Zhao L. VBNC Cronobacter sakazakii survives in macrophages by resisting oxidative stress and evading recognition by macrophages. BMC Microbiol 2024; 24:458. [PMID: 39506633 PMCID: PMC11539806 DOI: 10.1186/s12866-024-03595-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024] Open
Abstract
Survival in host macrophages is an effective strategy for pathogenic bacterial transmission and pathogenesis. Our previous study found that viable but non-culturable (VBNC) Cronobacter Sakazakii (C. sakazakii) can survive in macrophages, but its survival mechanism is not clear. In this study, we investigated the possible mechanisms of VBNC C. sakazakii survival in macrophages in terms of environmental tolerance within macrophages and evasion of macrophages recognition. The results revealed that VBNC C. sakazakii survived under oxidative conditions at a higher rate than the culturable C. sakazakii. Moreover, the stringent response gene (relA and spoT) and the antioxidant-related genes (sodA, katG, and trxA) were up-regulated, indicating that VBNC C. sakazakii may regulate antioxidation through stringent response. On the other hand, compared with culturable C. sakazakii, VBNC C. sakazakii caused reduced response (Toll-like receptor 4) in macrophages, which was attributed to the suppression of biosynthesis of the lipopolysaccharides (LPS). Furthermore, we found that ellagic acid can reduce the survival rate of bacteria in macrophages by improving the immune TLR4 recognition ability of macrophages. In conclusion, VBNC C. sakazakii may survive in macrophages by regulating oxidative tolerance through stringent response and altering LPS synthesis to evade TLR4 recognition by macrophages, which suggests the pathogenic risk of VBNC C. sakazakii.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China
| | - Jingfeng Zhang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China
| | - Haoqing Zhao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China
| | - Feifeng Zhong
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China
| | - Jianyu Li
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China
| | - Lichao Zhao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, Guangdong Province, 510642, China.
| |
Collapse
|
38
|
Le MH, Humayun S, Lee HJ, Mi XJ, Justine EE, Tran THM, Park HR, Kim YJ. Structural identification and immunostimulatory effect of Bacillus velezensis GV1 polysaccharides via TLR4/NF-κB signaling pathway in RAW264.7 macrophages. Int J Biol Macromol 2024; 280:135808. [PMID: 39306178 DOI: 10.1016/j.ijbiomac.2024.135808] [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: 04/04/2024] [Revised: 09/04/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Microbial polysaccharides derived from bacterial sources possess unique properties because of their structural complexity contributing to exceptional characteristics, including potent immunostimulatory effects. In this study, we extracted crude polysaccharide from Bacillus velezensis GV1 (BPS) which was isolated from Korean ginseng vinegar, and subsequently characterized for sugar composition and functional groups using FT-IR and methylation method. Structural analysis indicated that BPS was composed of mannan and glucan in a ratio of 7.5:2.5. The immunostimulatory effect of BPS was investigated in RAW264.7 macrophages. The results revealed that BPS significantly increased NO production, as well as the secretion and expression of key cytokines, such as IL-6, TNF-α, and IL-1β. These effects were confirmed using a TLR4 antagonist (TAK-242). Moreover, BPS exhibited immunostimulatory potential by promoting the NF-κB signaling pathway. In conclusion, this study establishes a foundation for the potential application of BPS as an immunostimulatory adjuvant or alternative component in functional foods, particularly for enhancing innate immune responses.
Collapse
Affiliation(s)
- Minh Ha Le
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea.
| | - Sanjida Humayun
- School of Natural Sciences and Health, Tallinn University, Narva mantee 25, 10120 Tallinn, Estonia.
| | - Hyo-Jun Lee
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea.
| | - Xiao-Jie Mi
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China.
| | - Elsa Easter Justine
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea.
| | - Thi Hoa My Tran
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea.
| | - Hye-Ryung Park
- Department of Hotel Food-service & Culinary Arts, Suwon Women's University, 1098 Juweok-ro, Hwaseong-si, Gyeonggi-do 18333, Republic of Korea.
| | - Yeon-Ju Kim
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
39
|
Roux S, Cherradi S, Duong HT. Uncovering the mechanism of troglitazone-mediated idiosyncratic drug-induced liver injury with individual-centric models. Arch Toxicol 2024; 98:3875-3884. [PMID: 39105737 PMCID: PMC11489277 DOI: 10.1007/s00204-024-03833-z] [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: 07/04/2024] [Accepted: 07/30/2024] [Indexed: 08/07/2024]
Abstract
Idiosyncratic drug-induced liver injury is a rare and unpredictable event. Deciphering its initiating-mechanism is a hard task as its occurrence is individual dependent. Thus, studies that utilize models that are not individual-centric might drive to a general mechanistic conclusion that is not necessarily true. Here, we use the individual-centric spheroid model to analyze the initiating-mechanism of troglitazone-mediated iDILI risk. Individual-centric spheroid models were generated using a proprietary cell educating technology. These educated spheroids contain hepatocytes, hepatic stellate cells, activated monocyte-derived macrophages, and dendritic cells under physiological conditions. We show that phases 1 and 2 drug-metabolizing enzymes were induced in an individual-dependent manner. However, we did not observe any association of DEMs induction and troglitazone (TGZ)-mediated iDILI risk. We analyzed TGZ-mediated iDILI and found that a 44-year-old male showed iDILI risk that is associated with TGZ-mediated suppression of IL-12 expression by autologous macrophages and dendritic cells. We performed a rescue experiment and showed that treatment of spheroids from this 44-year-old male with TGZ and recombinant IL-12 suppressed iDILI risk. We confirmed the mechanism in another 31-year-old female with iDILI risk. We demonstrate here that individual-centric spheroid are versatile models that allow to predict iDILI risk and to analyze a direct effect of the drug on activated macrophages and dendritic cells to uncover the initiating-mechanism of iDILI occurrence. This model opens perspectives for a personalized strategy to mitigate iDILI risk.
Collapse
Affiliation(s)
- Salomé Roux
- PredictCan Biotechnologies SAS, Biopôle Euromédecine, 1682 Rue de La Valsière, 34790, Grabels, France
| | - Sara Cherradi
- PredictCan Biotechnologies SAS, Biopôle Euromédecine, 1682 Rue de La Valsière, 34790, Grabels, France
| | - Hong Tuan Duong
- PredictCan Biotechnologies SAS, Biopôle Euromédecine, 1682 Rue de La Valsière, 34790, Grabels, France.
| |
Collapse
|
40
|
Tiligada E, Stefanaki C, Ennis M, Neumann D. Opportunities and challenges in the therapeutic exploitation of histamine and histamine receptor pharmacology in inflammation-driven disorders. Pharmacol Ther 2024; 263:108722. [PMID: 39306197 DOI: 10.1016/j.pharmthera.2024.108722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/31/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
Inflammation-driven diseases encompass a wide array of pathological conditions characterised by immune system dysregulation leading to tissue damage and dysfunction. Among the myriad of mediators involved in the regulation of inflammation, histamine has emerged as a key modulatory player. Histamine elicits its actions through four rhodopsin-like G-protein-coupled receptors (GPCRs), named chronologically in order of discovery as histamine H1, H2, H3 and H4 receptors (H1-4R). The relatively low affinity H1R and H2R play pivotal roles in mediating allergic inflammation and gastric acid secretion, respectively, whereas the high affinity H3R and H4R are primarily linked to neurotransmission and immunomodulation, respectively. Importantly, however, besides the H4R, both H1R and H2R are also crucial in driving immune responses, the H2R tending to promote yet ill-defined and unexploited suppressive, protective and/or resolving processes. The modulatory action of histamine via its receptors on inflammatory cells is described in detail. The potential therapeutic value of the most recently discovered H4R in inflammatory disorders is illustrated via a selection of preclinical models. The clinical trials with antagonists of this receptor are discussed and possible reasons for their lack of success described.
Collapse
Affiliation(s)
- Ekaterini Tiligada
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| | - Charikleia Stefanaki
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece; University Research Institute of Maternal and Child Health and Precision Medicine, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Madeleine Ennis
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, UK
| | - Detlef Neumann
- Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
41
|
Fernando V, Zheng X, Sharma V, Sweef O, Choi ES, Furuta S. Reprogramming of breast tumor-associated macrophages with modulation of arginine metabolism. Life Sci Alliance 2024; 7:e202302339. [PMID: 39191486 PMCID: PMC11350068 DOI: 10.26508/lsa.202302339] [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: 08/25/2023] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024] Open
Abstract
HER2+ breast tumors have abundant immune-suppressive cells, including M2-type tumor-associated macrophages (TAMs). Although TAMs consist of the immune-stimulatory M1 type and immune-suppressive M2 type, the M1/M2-TAM ratio is reduced in immune-suppressive tumors, contributing to their immunotherapy refractoriness. M1- versus M2-TAM formation depends on differential arginine metabolism, where M1-TAMs convert arginine to nitric oxide (NO) and M2-TAMs convert arginine to polyamines (PAs). We hypothesize that such distinct arginine metabolism in M1- versus M2-TAMs is attributed to different availability of BH4 (NO synthase cofactor) and that its replenishment would reprogram M2-TAMs to M1-TAMs. Recently, we reported that sepiapterin (SEP), the endogenous BH4 precursor, elevates the expression of M1-TAM markers within HER2+ tumors. Here, we show that SEP restores BH4 levels in M2-like macrophages, which then redirects arginine metabolism to NO synthesis and converts M2 type to M1 type. The reprogrammed macrophages exhibit full-fledged capabilities of antigen presentation and induction of effector T cells to trigger immunogenic cell death of HER2+ cancer cells. This study substantiates the utility of SEP in the metabolic shift of the HER2+ breast tumor microenvironment as a novel immunotherapeutic strategy.
Collapse
Affiliation(s)
- Veani Fernando
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, Toledo, OH, USA
- Division of Rheumatology, University of Colorado, Anschutz Medical Campus Barbara Davis Center, Aurora, CO, USA
| | - Xunzhen Zheng
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, Toledo, OH, USA
| | - Vandana Sharma
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, Toledo, OH, USA
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, USA
| | - Osama Sweef
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Eun-Seok Choi
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Saori Furuta
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, Toledo, OH, USA
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH, USA
| |
Collapse
|
42
|
Zorrilla E, Della Pietra A, Russo AF. Interplay between cannabinoids and the neuroimmune system in migraine. J Headache Pain 2024; 25:178. [PMID: 39407099 PMCID: PMC11481476 DOI: 10.1186/s10194-024-01883-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
Migraine is a common and complex neurological disorder that has a high impact on quality of life. Recent advances with drugs that target the neuropeptide calcitonin gene-related peptide (CGRP) have helped, but treatment options remain insufficient. CGRP is released from trigeminal sensory fibers and contributes to peripheral sensitization, perhaps in part due to actions on immune cells in the trigeminovascular system. In this review, we will discuss the potential of cannabinoid targeting of immune cells as an innovative therapeutic target for migraine treatment. We will cover endogenous endocannabinoids, plant-derived phytocannabinoids and synthetically derived cannabinoids. The focus will be on six types of immune cells known to express multiple cannabinoid receptors: macrophages, monocytes, mast cells, dendritic cells, B cells, and T cells. These cells also contain receptors for CGRP and as such, cannabinoids might potentially modulate the efficacy of current CGRP-targeting drugs. Unfortunately, to date most studies on cannabinoids and immune cells have relied on cell cultures and only a single preclinical study has tested cannabinoid actions on immune cells in a migraine model. Encouragingly, in that study a synthetically created stable chiral analog of an endocannabinoid reduced meningeal mast cell degranulation. Likewise, clinical trials evaluating the safety and efficacy of cannabinoid-based therapies for migraine patients have been limited but are encouraging. Thus, the field is at its infancy and there are significant gaps in our understanding of the impact of cannabinoids on immune cells in migraine. Future research exploring the interactions between cannabinoids and immune cells could lead to more targeted and effective migraine treatments.
Collapse
Affiliation(s)
- Erik Zorrilla
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, 52242, USA
| | - Adriana Della Pietra
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, 52242, USA
| | - Andrew F Russo
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, 52242, USA.
- Department of Neurology, University of Iowa, Iowa City, IA, 52242, USA.
- Veterans Affairs Healthcare System, Iowa City, IA, 52246, USA.
| |
Collapse
|
43
|
Luu AM, Shepardson KM, Rynda-Apple A. A Comprehensive Protocol for the Collection, Differentiation, Cryopreservation, and Resuscitation of Primary Murine Bone Marrow Derived Macrophages (BMDM). Immunol Invest 2024; 53:1001-1012. [PMID: 39115808 PMCID: PMC11451725 DOI: 10.1080/08820139.2024.2382805] [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] [Indexed: 10/02/2024]
Abstract
BACKGROUND The field of immunology has undoubtedly benefited from the in vitro use of cell lines for immunological studies; however, due to the "immortal" nature of many cell lines, they are not always the best model. Thus, direct collection and culture of primary cells from model organisms is a solution that many researchers utilize. To the best of our knowledge, there is not a singular protocol which encompasses the entire process of bone marrow cell collection through cryopreservation and resuscitation of cells from a murine model. METHODS Bone marrow cells were collected from mice with a C57BL6 genetic background. Cells were differentiated using L929 conditioned media. Cells were assessed using a combination of microscopy, differential staining, immunocytochemistry, and trypan blue. Results: Primary murine BMDMs that underwent cryopreservation followed by resuscitation retained a high degree of viability. Furthermore, these BMDMs retained on overall ability to clear S. aureus. RESULTS Primary murine BMDMs that underwent cryopreservation followed by resuscitation retained a high degree of viability. Furthermore, these BMDMs retained on overall ability to clear S. aureus. CONCLUSION Crypopreserved and resuscitated primary murine BMDMs were viable and retained their pverall S. aureus clearance ability.
Collapse
Affiliation(s)
- Abby M Luu
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Kelly M Shepardson
- Department of Molecular Cell Biology, University of California Merced, Merced, California, USA
| | - Agnieszka Rynda-Apple
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| |
Collapse
|
44
|
Ferreira DA, Medeiros ABA, Soares MM, Lima ÉDA, de Oliveira GCSL, Leite MBDS, Machado MV, Villar JAFP, Barbosa LA, Scavone C, Moura MT, Rodrigues-Mascarenhas S. Evaluation of Anti-Inflammatory Activity of the New Cardiotonic Steroid γ-Benzylidene Digoxin 8 (BD-8) in Mice. Cells 2024; 13:1568. [PMID: 39329752 PMCID: PMC11430542 DOI: 10.3390/cells13181568] [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: 07/31/2024] [Revised: 09/10/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Cardiotonic steroids are known to bind to Na+/K+-ATPase and regulate several biological processes, including the immune response. The synthetic cardiotonic steroid γ-Benzylidene Digoxin 8 (BD-8) is emerging as a promising immunomodulatory molecule, although it has remained largely unexplored. Therefore, we tested the immunomodulatory potential of BD-8 both in vitro and in vivo. Hence, primary mouse macrophages were incubated with combinations of BD-8 and the pro-inflammatory fungal protein zymosan (ZYM). Nitric oxide (NO) production was determined by Griess reagent and cytokines production was assessed by enzyme-linked immunosorbent assay. Inducible nitric oxide synthase (iNOS), reactive oxygen species (ROS), p-nuclear factor kappa B p65 (NF-κB p65), p-extracellular signal-regulated kinase (p-ERK), and p-p38 were evaluated by flow cytometry. Macrophages exposed to BD-8 displayed reduced phagocytic activity, NO levels, and production of the proinflammatory cytokine IL-1β induced by ZYM. Furthermore, BD-8 diminished the expression of iNOS and phosphorylation of NF-κB p65, ERK, and p38. Additionally, BD-8 exhibited anti-inflammatory capacity in vivo in a carrageenan-induced mouse paw edema model. Taken together, these findings demonstrate the anti-inflammatory activity of BD-8 and further reinforce the potential of cardiotonic steroids and their derivatives as immunomodulatory molecules.
Collapse
Affiliation(s)
- Davi Azevedo Ferreira
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraiba, João Pessoa 58.051-900, PB, Brazil; (D.A.F.); (A.B.A.M.); (M.M.S.); (É.d.A.L.); (G.C.S.L.d.O.); (M.B.d.S.L.)
| | - Anna Beatriz Araujo Medeiros
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraiba, João Pessoa 58.051-900, PB, Brazil; (D.A.F.); (A.B.A.M.); (M.M.S.); (É.d.A.L.); (G.C.S.L.d.O.); (M.B.d.S.L.)
| | - Mariana Mendonça Soares
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraiba, João Pessoa 58.051-900, PB, Brazil; (D.A.F.); (A.B.A.M.); (M.M.S.); (É.d.A.L.); (G.C.S.L.d.O.); (M.B.d.S.L.)
| | - Éssia de Almeida Lima
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraiba, João Pessoa 58.051-900, PB, Brazil; (D.A.F.); (A.B.A.M.); (M.M.S.); (É.d.A.L.); (G.C.S.L.d.O.); (M.B.d.S.L.)
| | - Gabriela Carolina Santos Lima de Oliveira
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraiba, João Pessoa 58.051-900, PB, Brazil; (D.A.F.); (A.B.A.M.); (M.M.S.); (É.d.A.L.); (G.C.S.L.d.O.); (M.B.d.S.L.)
| | - Mateus Bernardo da Silva Leite
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraiba, João Pessoa 58.051-900, PB, Brazil; (D.A.F.); (A.B.A.M.); (M.M.S.); (É.d.A.L.); (G.C.S.L.d.O.); (M.B.d.S.L.)
| | - Matheus Vieira Machado
- Laboratory of Cellular Biochemistry, Campus Centro-Oeste Dona Lindú, Federal University of São João del-Rei, Divinópolis 35.501-296, MG, Brazil; (M.V.M.); (J.A.F.P.V.); (L.A.B.)
| | - José Augusto Ferreira Perez Villar
- Laboratory of Cellular Biochemistry, Campus Centro-Oeste Dona Lindú, Federal University of São João del-Rei, Divinópolis 35.501-296, MG, Brazil; (M.V.M.); (J.A.F.P.V.); (L.A.B.)
| | - Leandro Augusto Barbosa
- Laboratory of Cellular Biochemistry, Campus Centro-Oeste Dona Lindú, Federal University of São João del-Rei, Divinópolis 35.501-296, MG, Brazil; (M.V.M.); (J.A.F.P.V.); (L.A.B.)
| | - Cristoforo Scavone
- Laboratory of Neuropharmacology Research, Department of Pharmacology, Institute of Biomedical Sciences ICB-1, University of São Paulo, São Paulo 05.508-900, SP, Brazil;
| | - Marcelo Tigre Moura
- Laboratory of Cellular Reprogramming, Biotechnology Center, Federal University of Paraiba, João Pessoa 58.051-900, PB, Brazil;
| | - Sandra Rodrigues-Mascarenhas
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraiba, João Pessoa 58.051-900, PB, Brazil; (D.A.F.); (A.B.A.M.); (M.M.S.); (É.d.A.L.); (G.C.S.L.d.O.); (M.B.d.S.L.)
| |
Collapse
|
45
|
Pandur E, Pap R, Sipos K. Activated THP-1 Macrophage-Derived Factors Increase the Cytokine, Fractalkine, and EGF Secretions, the Invasion-Related MMP Production, and Antioxidant Activity of HEC-1A Endometrium Cells. Int J Mol Sci 2024; 25:9624. [PMID: 39273575 PMCID: PMC11395051 DOI: 10.3390/ijms25179624] [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: 08/01/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Endometrium receptivity is a multifactor-regulated process involving progesterone receptor-regulated signaling, cytokines and chemokines, and additional growth regulatory factors. In the female reproductive system, macrophages have distinct roles in the regulation of receptivity, embryo implantation, immune tolerance, and angiogenesis or oxidative stress. In the present study, we investigated the effects of PMA-activated THP-1 macrophages on the receptivity-related genes, cytokines and chemokines, growth regulators, and oxidative stress-related molecules of HEC-1A endometrium cells. We established a non-contact co-culture in which the culture medium of the PMA-activated macrophages exhibiting the pro-inflammatory phenotype was used for the treatment of the endometrial cells. In the endometrium cells, the expression of the growth-related factors activin and bone morphogenetic protein 2, the growth hormone EGF, and the activation of the downstream signaling molecules pERK1/2 and pAkt were analyzed by ELISA and Western blot. The secretions of cytokines and chemokines, which are involved in the establishment of endometrial receptivity, and the expression of matrix metalloproteinases implicated in invasion were also determined. Based on the results, the PMA-activated THP-1 macrophages exhibiting a pro-inflammatory phenotype may play a role in the regulation of HEC-1A endometrium cells. They alter the secretion of cytokines and chemokines, as well as the protein level of MMPs of HEC-1A cells. Moreover, activated THP-1 macrophages may elevate oxidative stress protection of HEC-1A endometrium cells. All these suggest that pro-inflammatory macrophages have a special role in the regulation of receptivity-related and implantation-related factors of HEC-1A cells.
Collapse
Affiliation(s)
- Edina Pandur
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, 7624 Pécs, Hungary; (R.P.); (K.S.)
- National Laboratory of Human Reproduction, University of Pécs, 7624 Pécs, Hungary
| | - Ramóna Pap
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, 7624 Pécs, Hungary; (R.P.); (K.S.)
- National Laboratory of Human Reproduction, University of Pécs, 7624 Pécs, Hungary
| | - Katalin Sipos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, 7624 Pécs, Hungary; (R.P.); (K.S.)
- National Laboratory of Human Reproduction, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
46
|
Anwar C, Lin JR, Tsai ML, Ho CT, Lai CS. Calebin A attenuated inflammation in RAW264.7 macrophages and adipose tissue to improve hepatic glucose metabolism and hyperglycemia in high-fat diet-fed obese mice. Eur J Pharmacol 2024; 978:176789. [PMID: 38945287 DOI: 10.1016/j.ejphar.2024.176789] [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: 04/03/2024] [Revised: 06/21/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024]
Abstract
The increased incidence of obesity, which become a global health problem, requires more functional food products with minor side and excellent effects. Calebin A (CbA) is a non-curcuminoid compound, which is reported to be an effective treatment for lipid metabolism and thermogenesis. However, its ability and mechanism of action in improving obesity-associated hyperglycemia remain unclear. This study was designed to explore the effect and mechanism of CbA in hyperglycemia via improvement of inflammation and glucose metabolism in the adipose tissue and liver in high-fat diet (HFD)-fed mice. After 10 weeks fed HFD, obese mice supplemented with CbA (25 and 100 mg/kg) for another 10 weeks showed a remarkable reducing adiposity and blood glucose. CbA modulated M1/M2 macrophage polarization, ameliorated inflammatory cytokines, and restored adiponectin as well as Glut 4 expression in the adipose tissue. In the in vitro study, CbA attenuated pro-inflammatory markers while upregulated anti-inflammatory IL-10 in LPS + IFNγ-generated M1 phenotype macrophages. In the liver, CbA attenuated steatosis, inflammatory infiltration, and protein levels of inflammatory TNF-α and IL-6. Moreover, CbA markedly upregulated Adiponectin receptor 1, AMPK, and insulin downstream Akt signaling to improve glycogen content and increase Glut2 protein. These findings indicated that CbA may be a novel therapeutic approach to treat obesity and hyperglycemia phenotype targeting on adipose inflammation and hepatic insulin signaling.
Collapse
Affiliation(s)
- Choirul Anwar
- Institute of Aquatic Science and Technology, Collage of Hydrosphere Science, National Kaohsiung University of Science and Technology, Kaohsiung, 81157, Taiwan
| | - Jing-Ru Lin
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung, 81157, Taiwan
| | - Mei-Ling Tsai
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung, 81157, Taiwan
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, 08901, USA.
| | - Ching-Shu Lai
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung, 81157, Taiwan.
| |
Collapse
|
47
|
Artemova D, Vishnyakova P, Elchaninov A, Gantsova E, Sukhikh G, Fatkhudinov T. M1 macrophages as promising agents for cell therapy of endometriosis. Heliyon 2024; 10:e36340. [PMID: 39253270 PMCID: PMC11381802 DOI: 10.1016/j.heliyon.2024.e36340] [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: 03/19/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
Endometriosis is a chronic estrogen-dependent disease characterized by the presence of endometrial glands and stroma outside their normal anatomical location. While laparoscopic removal of foci remains the gold standard therapy, it has limited efficacy and certain risks. However, cell therapy using pro-inflammatory M1 macrophages presents a promising and minimally invasive alternative for treating endometriosis. This approach showcases the potential for innovative and effective treatments for this condition. This study aims to explore the anti-endometriosis properties of M1 macrophages. A reproducible syngeneic mouse model of endometriosis was utilized, revealing that formed foci are primarily composed of macrophages with an anti-inflammatory M2 phenotype rather than M1 macrophages. To investigate further, chemically reprogrammed M1 macrophages were labeled with the membrane fluorescent tag PKH26 and administered to animals with endometriosis. Therapy resulted in a decrease in the number and size of foci, accompanied by a shift in the phenotypic composition of peritoneal macrophages. Specifically, the content of M2 macrophages decreased while that of M1 macrophages increased, resembling the composition of healthy animals. Our study conclusively demonstrates the anti-endometriosis properties of M1 macrophages, providing a strong foundation for future research in the cell therapy of endometriosis.
Collapse
Affiliation(s)
- Daria Artemova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 3 Tsurupa Street, 117418, Moscow, Russian Federation
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198, Moscow, Russian Federation
| | - Polina Vishnyakova
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198, Moscow, Russian Federation
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, 117997, Moscow, Russian Federation
| | - Andrey Elchaninov
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 3 Tsurupa Street, 117418, Moscow, Russian Federation
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198, Moscow, Russian Federation
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, 117997, Moscow, Russian Federation
| | - Elena Gantsova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 3 Tsurupa Street, 117418, Moscow, Russian Federation
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198, Moscow, Russian Federation
| | - Gennady Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, 117997, Moscow, Russian Federation
| | - Timur Fatkhudinov
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 3 Tsurupa Street, 117418, Moscow, Russian Federation
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198, Moscow, Russian Federation
| |
Collapse
|
48
|
Pu Z, Chen S, Lu Y, Wu Z, Cai Z, Mou L. Exploring the molecular mechanisms of macrophages in islet transplantation using single-cell analysis. Front Immunol 2024; 15:1407118. [PMID: 39267737 PMCID: PMC11391485 DOI: 10.3389/fimmu.2024.1407118] [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: 03/26/2024] [Accepted: 08/02/2024] [Indexed: 09/15/2024] Open
Abstract
Background Islet transplantation is a promising treatment for type 1 diabetes that aims to restore insulin production and improve glucose control, but long-term graft survival remains a challenge due to immune rejection. Methods ScRNA-seq data from syngeneic and allogeneic islet transplantation grafts were obtained from GSE198865. Seurat was used for filtering and clustering, and UMAP was used for dimension reduction. Differentially expressed genes were analyzed between syngeneic and allogeneic islet transplantation grafts. Gene set variation analysis (GSVA) was performed on the HALLMARK gene sets from MSigDB. Monocle 2 was used to reconstruct differentiation trajectories, and cytokine signature enrichment analysis was used to compare cytokine responses between syngeneic and allogeneic grafts. Results Three distinct macrophage clusters (Mø-C1, Mø-C2, and Mø-C3) were identified, revealing complex interactions and regulatory mechanisms within macrophage populations. The significant activation of macrophages in allogeneic transplants was marked by the upregulation of allograft rejection-related genes and pathways involved in inflammatory and interferon responses. GSVA revealed eight pathways significantly upregulated in the Mø-C2 cluster. Trajectory analysis revealed that Mø-C3 serves as a common progenitor, branching into Mø-C1 and Mø-C2. Cytokine signature enrichment analysis revealed significant differences in cytokine responses, highlighting the distinct immunological environments created by syngeneic and allogeneic grafts. Conclusion This study significantly advances the understanding of macrophage roles within the context of islet transplantation by revealing the interactions between immune pathways and cellular fate processes. The findings highlight potential therapeutic targets for enhancing graft survival and function, emphasizing the importance of understanding the immunological aspects of transplant acceptance and longevity.
Collapse
Affiliation(s)
- Zuhui Pu
- Imaging Department, Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
- MetaLife Lab, Shenzhen Institute of Translational Medicine, Shenzhen, Guangdong, China
| | - Shujuan Chen
- Department of Endocrinology, Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Ying Lu
- MetaLife Lab, Shenzhen Institute of Translational Medicine, Shenzhen, Guangdong, China
| | - Zijing Wu
- MetaLife Lab, Shenzhen Institute of Translational Medicine, Shenzhen, Guangdong, China
| | - Zhiming Cai
- BGI Medical Group, Shenzhen, Guangdong, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Lisha Mou
- MetaLife Lab, Shenzhen Institute of Translational Medicine, Shenzhen, Guangdong, China
- Department of Endocrinology, Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
49
|
Arshi A, Mahmoudi E, Raeisi F, Dehghan Tezerjani M, Bahramian E, Ahmed Y, Peng C. Exploring potential roles of long non-coding RNAs in cancer immunotherapy: a comprehensive review. Front Immunol 2024; 15:1446937. [PMID: 39257589 PMCID: PMC11384988 DOI: 10.3389/fimmu.2024.1446937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024] Open
Abstract
Cancer treatment has long been fraught with challenges, including drug resistance, metastasis, and recurrence, making it one of the most difficult diseases to treat effectively. Traditional therapeutic approaches often fall short due to their inability to target cancer stem cells and the complex genetic and epigenetic landscape of tumors. In recent years, cancer immunotherapy has revolutionized the field, offering new hope and viable alternatives to conventional treatments. A particularly promising area of research focuses on non-coding RNAs (ncRNAs), especially long non-coding RNAs (lncRNAs), and their role in cancer resistance and the modulation of signaling pathways. To address these challenges, we performed a comprehensive review of recent studies on lncRNAs and their impact on cancer immunotherapy. Our review highlights the crucial roles that lncRNAs play in affecting both innate and adaptive immunity, thereby influencing the outcomes of cancer treatments. Key observations from our review indicate that lncRNAs can modify the tumor immune microenvironment, enhance immune cell infiltration, and regulate cytokine production, all of which contribute to tumor growth and resistance to therapies. These insights suggest that lncRNAs could serve as potential targets for precision medicine, opening up new avenues for developing more effective cancer immunotherapies. By compiling recent research on lncRNAs across various cancers, this review aims to shed light on their mechanisms within the tumor immune microenvironment.
Collapse
Affiliation(s)
- Asghar Arshi
- Department of Biology, York University, Toronto, ON, Canada
| | - Esmaeil Mahmoudi
- Young Researchers and Elite Club, Islamic Azad University, Shahrekord, Iran
| | | | - Masoud Dehghan Tezerjani
- Department of bioinformatics, School of Advanced Medical Technologies, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Bahramian
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR, United States
| | - Yeasin Ahmed
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR, United States
| | - Chun Peng
- Department of Biology, York University, Toronto, ON, Canada
| |
Collapse
|
50
|
Lou YL, Xie DL, Huang XH, Zheng MM, Chen N, Xu JR. The role of MNK1-mTORC1 pathway in modulating macrophage responses to Vibrio vulnificus infection. Microbiol Spectr 2024; 12:e0334023. [PMID: 38980024 PMCID: PMC11302032 DOI: 10.1128/spectrum.03340-23] [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: 09/12/2023] [Accepted: 06/06/2024] [Indexed: 07/10/2024] Open
Abstract
Vibrio vulnificus (Vv) is known to cause life-threatening infections, particularly septicemia. These patients often exhibit elevated levels of pro-inflammatory cytokines. While it is established that mitogen-activated protein kinase (MAPK)-interacting kinase (MNK) contributes to the production of pro-inflammatory cytokines, the role of MNK in macrophages during Vv infection remains unclear. In this study, we investigate the impact of MNK on macrophages. We demonstrate that the inhibition of MNK in J774A.1 cells, when treated with lipopolysaccharide or Vv, resulted in decreased production of tumor necrosis factor alpha and interleukin-6, without affecting their transcription. Interestingly, treatment with MNK inhibitor CGP57380 led to enhanced phosphorylation of MNK1 but decreased phosphorylation of eIF4E. Moreover, MNK1 knockout cells exhibited an increased capacity for phagocytosis and clearance of Vv, with more acidic phagosomes than the parental cells. Notably, CGP57380 did not impact phagocytosis, bacterial clearance, or phagosome acidification in Vv-infected J774A.1 cells. Considering the reported association between MNK and mammalian target of rapamycin complex 1 (mTORC1) activation, we investigated the mTORC1 signaling in MNK1 knockout cells infected with Vv. Our results revealed that attenuation of the mTORC1 signaling in these cells and treatment with the mTORC1 inhibitor rapamycin significantly enhanced bacterial clearance in J774A.1 cells following Vv infection. In summary, our findings suggest that MNK promotes the Vv-induced cytokine production in J774A.1 cells without affecting their transcription levels. MNK1 appears to impair the phagocytosis, bacterial clearance, and phagosome acidification in Vv-infected J774A.1 cells through the MNK1-mTORC1 signaling pathway rather than the MNK1-eIF4E signaling pathway. Our findings highlight the importance of the MNK1-mTORC1 pathway in modulating macrophage responses to Vv infection. IMPORTANCE Mitogen-activated protein kinase (MAPK)-interacting kinase (MNK) plays a role in promoting the production of tumor necrosis factor alpha and interleukin-6 in macrophages during Vibrio vulnificus (Vv) infection. Inhibition or knockout of MNK1 in J774A.1 cells resulted in reduced cytokine production without affecting their transcription levels. MNK1 also impairs phagocytosis, bacterial clearance, and phagosome acidification in Vv-infected cells through the MNK1-mammalian target of rapamycin complex 1 (mTORC1) signaling pathway. The findings highlight the importance of the MNK1-mTORC1 pathway in modulating macrophage responses to Vv infection.
Collapse
Affiliation(s)
- Yong-Liang Lou
- Department of Immunology and Pathogenic Biology, School of Medicine, Xi'an Jiaotong University, Xi'an, Shanxi, China
- The School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key Laboratory of Sanitary Microbiology, Wenzhou, Zhejiang, China
| | - Dan-Li Xie
- The School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key Laboratory of Sanitary Microbiology, Wenzhou, Zhejiang, China
| | - Xian-Hui Huang
- The School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key Laboratory of Sanitary Microbiology, Wenzhou, Zhejiang, China
| | - Meng-Meng Zheng
- The School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key Laboratory of Sanitary Microbiology, Wenzhou, Zhejiang, China
- Scientific Research Center, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Na Chen
- The School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Laboratory Medicine, The First People’s Hospital of Linping District, Hangzhou, Zhejiang, China
| | - Ji-Ru Xu
- Department of Immunology and Pathogenic Biology, School of Medicine, Xi'an Jiaotong University, Xi'an, Shanxi, China
| |
Collapse
|