1
|
Fu Y, Wang Y, Wang Y, Mou T, He X, Wang J, Xiong A, Qing B, Wu D, Li G. Biomarkers (NLR, PLR, SII) for Frequent COPD Exacerbations: Diagnostic and Clinical Management Implications in a Retrospective Study. Int J Chron Obstruct Pulmon Dis 2025; 20:987-998. [PMID: 40207023 PMCID: PMC11980941 DOI: 10.2147/copd.s510118] [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: 12/09/2024] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
Objective To evaluate the diagnostic and predictive value of the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and systemic immune-inflammation index (SII) for frequent exacerbations of chronic obstructive pulmonary disease (AECOPD), and to develop a risk stratification scoring system to optimize clinical management in resource-limited healthcare settings. Patients and Methods This retrospective observational study enrolled 16,849 AECOPD patients, categorized into frequent exacerbators (≥2 exacerbations/year, n=3,488) and non-frequent exacerbators (<2 exacerbations/year, n=13,361). Comparative analyses of clinical characteristics and inflammatory biomarkers (NLR, PLR, SII, CRP, PCT) were conducted. Spearman correlation, receiver operating characteristic (ROC) curve analysis, and binary logistic regression were employed to assess biomarker performance. A risk scoring system was developed using odds ratios (OR) and regression coefficients (β) of NLR and PLR. Results The frequent exacerbators group exhibited significantly higher median NLR (6.71 vs 5.10, P < 0.001), mean PLR (239 ± 204 vs 218 ± 195, P < 0.001), and median SII (1,137.48 vs 847.54, P < 0.001). NLR, PLR and SII showed strong positive correlations with CRP and PCT (P < 0.001). ROC analysis identified NLR (specificity = 84.1%) and PLR (sensitivity = 55%) as optimal diagnostic indicators. Regression analysis confirmed NLR and PLR as independent risk factors for frequent exacerbations. The risk stratification system categorized patients into low-risk (<290 points; annual exacerbation rate 17%), intermediate-risk (290-768 points; 19.1%), and high-risk (>768 points; 23.4%) groups. Conclusion NLR and PLR serve as cost-effective biomarkers for identifying high-risk frequent exacerbators patients with COPD in primary care settings. The percentile-based scoring system enables management strategies to address clinical needs in resource-constrained healthcare environments.
Collapse
Affiliation(s)
- Yufen Fu
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Longchang People’s Hospital, Neijiang, 642150, People’s Republic of China
| | - Yuxin Wang
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Pulmonary Medicine, Chengdu Third People’s Hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of Chongqing Medical University, Chengdu, 610031, People’s Republic of China
| | - Yujiao Wang
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Pulmonary Medicine, Chengdu Third People’s Hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of Chongqing Medical University, Chengdu, 610031, People’s Republic of China
| | - Ting Mou
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Pulmonary Medicine, Chengdu Third People’s Hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of Chongqing Medical University, Chengdu, 610031, People’s Republic of China
| | - Xiang He
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, People’s Republic of China
- Department of Pulmonary Medicine, Chengdu Third People’s Hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of Chongqing Medical University, Chengdu, 610031, People’s Republic of China
| | - Junyi Wang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, People’s Republic of China
- Department of Pulmonary Medicine, Chengdu Third People’s Hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of Chongqing Medical University, Chengdu, 610031, People’s Republic of China
| | - Anying Xiong
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, People’s Republic of China
- Department of Pulmonary Medicine, Chengdu Third People’s Hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of Chongqing Medical University, Chengdu, 610031, People’s Republic of China
| | - Bomiao Qing
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, People’s Republic of China
- Department of Pulmonary Medicine, Chengdu Third People’s Hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of Chongqing Medical University, Chengdu, 610031, People’s Republic of China
| | - Dehong Wu
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, People’s Republic of China
- Department of Pulmonary Medicine, Chengdu Third People’s Hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of Chongqing Medical University, Chengdu, 610031, People’s Republic of China
| | - Guoping Li
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, People’s Republic of China
- Department of Pulmonary Medicine, Chengdu Third People’s Hospital branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of Chongqing Medical University, Chengdu, 610031, People’s Republic of China
| |
Collapse
|
2
|
Li N, Pang Y, Xu J, Elango J, Wu W. Immunomodulatory Effects of Symplectoteuthis oualaniensis Protamine and Its PEG Derivative on Macrophages: Involvement of PI3K/Akt Signaling, Redox Regulation, and Cell Cycle Modulation. Antioxidants (Basel) 2025; 14:437. [PMID: 40298789 PMCID: PMC12024133 DOI: 10.3390/antiox14040437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/30/2025] Open
Abstract
Protamine is a promising marine-derived bioactive compound that is highly arginine-rich and has demonstrated unique advantages in medical and biological research. This study, for the first time, investigates the molecular mechanisms underlying the immunomodulatory effects of Salmon Protamine Sulfate (SPS), Symplectoteuthis oualaniensis Protamine (SOP), and its polyethylene glycol (PEG) derivative (SOP-PEG) on RAW264.7 macrophages. The results demonstrate that both SOP and SOP-PEG significantly enhance the proliferation of RAW264.7 cells by promoting the secretion of pro-inflammatory cytokines and nitric oxide (NO), increasing ROS production, and improving antioxidant capacity, in comparison to SPS. Elevated ROS levels play a crucial role in enhancing macrophage immune activity, while the enhanced antioxidant defense mechanisms help maintain redox homeostasis and protect against oxidative stress-induced cellular damage. A Western blot analysis reveals that SOP and SOP-PEG notably regulate the expression of key proteins associated with the PI3K/Akt signaling pathway and anti-apoptotic mechanisms. Furthermore, a flow cytometry analysis indicates a significant increase in the G2/M-phase cell population in the treatment groups, which is corroborated by Western blot data showing alterations in critical regulatory proteins. Notably, SOP-PEG exhibits the strongest effects in regulating macrophage immune activity, which can be attributed to the enhanced stability and prolonged bioactivity resulting from the PEGylation of SOP. This comprehensive study reveals how SOP and SOP-PEG enhance macrophage immune function through multiple mechanisms, including PI3K/Akt activation, redox regulation, and cell cycle modulation. It provides valuable insights and a theoretical foundation for their potential applications in immunotherapy and immune regulation.
Collapse
Affiliation(s)
- Na Li
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (Y.P.); (J.X.)
| | - Yida Pang
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (Y.P.); (J.X.)
| | - Jiren Xu
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (Y.P.); (J.X.)
| | - Jeevithan Elango
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (Y.P.); (J.X.)
- Department of Biomaterials Engineering, Faculty of Health Sciences, UCAM-Universidad Católica San Antonio de Murcia, Guadalupe, 30107 Murcia, Spain
- Center of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India
| | - Wenhui Wu
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (N.L.); (Y.P.); (J.X.)
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai 201306, China
- Putuo Branch of International Combined Research Center for Marine Biological Sciences, Zhoushan 316104, China
| |
Collapse
|
3
|
Nguyen JA, Orsetti TL, Vernon P, Greene CJ, McKenna N, Yates RM. Direct neutrophil and T cell contact with macrophages induces release of phagosomally processed PAMPs via eructophagy. J Cell Sci 2025; 138:jcs263731. [PMID: 40019316 DOI: 10.1242/jcs.263731] [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: 11/15/2024] [Accepted: 02/21/2025] [Indexed: 03/01/2025] Open
Abstract
Macrophages play a pivotal role in clearing debris and microbes from the microenvironment via phagocytosis and orchestrating local inflammation. Although pathogen- and damage-associated molecular patterns (PAMPs and DAMPs) are understood to mostly be released through the synthesis and secretion of soluble mediators, such as cytokines and eicosanoids, it has been recently proposed that macrophages can release previously phagocytosed and processed PAMPs and DAMPs into the local microenvironment via a process termed eructophagy, and that these, in turn, can activate recently recruited leukocytes. Additionally, it has been commonly observed that local macrophages physically interact with other leukocytes, such as neutrophils and T cells, recruited to sites of inflammation. This study demonstrates that eructophagy in macrophages is significantly induced during physical interaction with neutrophils and T cells, which is mediated by ICAM1 on macrophages and lymphocyte function-associated antigen 1 (LFA1) on neutrophils and T cells. Notably, ICAM1 activation alone is sufficient to trigger eructophagy in macrophages and is dependent on Lyn kinase. Through this mechanism, it is proposed that neutrophils and lymphocytes can influence their own activation by interacting with local macrophages containing PAMP-containing phagolysosomes, which subsequently triggers PAMP release into the local microenvironment through eructophagy.
Collapse
Affiliation(s)
- Jenny A Nguyen
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Tanis L Orsetti
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Philip Vernon
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Catherine J Greene
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Neil McKenna
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Robin M Yates
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Snyder Institute of Chronic Disease, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| |
Collapse
|
4
|
Singh SK, Pancholi SS. Current Updates on Pathogenesis, Systemic Therapy, and Treatment of Invasive Fungal Infections. Curr Drug Targets 2025; 26:203-220. [PMID: 39421988 DOI: 10.2174/0113894501337502241015121015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/12/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024]
Abstract
Numerous health hazards are associated with fungal infections, ranging from asymptomatic cases to potentially fatal invasive diseases that are especially dangerous for those with impaired immune systems. The main causes behind these diseases are opportunistic fungi, namely Aspergillus, Candida, and Cryptococcus. Invasive fungal infections (IFIs) require a global response that includes the development of vaccines, standardized protocols for diagnosis, potent antifungal medications, and strategies to stop drug-resistant strains. Improving high-risk group diagnosis and treatment is essential to lowering death rates. This review highlights the substantial health concerns associated with fungal infections, especially in immunocompromised individuals, and identifies Aspergillus, Candida, and Cryptococcus as the main pathogens. It highlights the necessity of international efforts, such as the development of novel diagnostic instruments, imaging methods, and antifungal drugs, to combat these invasive infections. The review also addresses the increasing need for novel treatment approaches in light of the developing resistance to widely used antifungal medications. Furthermore, the significance of secretory proteins in fungal pathogenicity and the potential of combination therapy are investigated. It is also suggested that a multimodal strategy be used to fight these illnesses, given the promise of multivalent vaccinations. Overall, this study emphasizes how critical it is to develop better diagnostic and treatment strategies in order to successfully control and lessen the impact of invasive fungal diseases on the health of the world.
Collapse
Affiliation(s)
- Sushil Kumar Singh
- Shree S.K. Patel College of Pharmaceutical Education and Research, Department of Pharmaceutical Technology, Ganpat University, Kherva, Mehsana, Gujarat 384012, India
| | - Shyam Sunder Pancholi
- School of Pharmacy and Technology Management, Department of Pharmaceutics (SPTM) SVKM's NMIMS (Deemed to be University) Mukesh Patel Technology Park, Babulde, Shirpur 425405, Dist. Dhule Maharashtra, India
| |
Collapse
|
5
|
Liu L, Li L, Wang T, Li Z, Yan B, Tan R, Zeng A, Ma W, Zhu X, Yin Z, Ma C. Recent nanoengineered therapeutic advancements in sepsis management. Front Bioeng Biotechnol 2024; 12:1495277. [PMID: 39703795 PMCID: PMC11655211 DOI: 10.3389/fbioe.2024.1495277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Sepsis (defined as sepsis 3.0) is a life-threatening organ dysfunction caused by a dysregulated host response to a variety of pathogenic microorganisms. Characterized by high morbidity and mortality, sepsis has become a global public health problem. However, there is a lack of appropriate diagnostic and therapeutic strategies for sepsis and current management rely on the limited treatment strategies. Recently, nanomedicines targeting and controlling the release of bio-active agents have shown excellent potency in sepsis management, with improved therapeutic efficacy and reduced adverse effects. In this review, we have summarized the advantages of nanomaterials. Also, the preparation and efficacy of the main categories of anti-sepsis nanomedicines applied in sepsis management are described in detail, including antibiotic-coated nanomaterials, antimicrobial peptides-coated nanomaterials, biomimetic nanomaterials, nanomaterials targeting macrophages and natural products loaded nanomaterials. These advances in nanomedicines establish the huge potential for nanomaterials-based sepsis management, especially in the improved pharmaceutical and pharmacological properties, enhanced therapeutic efficacy, controllable drug-targeting and reduced side effects. To further facilitate clinical translation of anti-sepsis nanomedicines, we propose that the issues involving safety, regulatory laws and cost-effectiveness should receive much more attention in the future.
Collapse
Affiliation(s)
- Li Liu
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Li Li
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Ting Wang
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Zheyu Li
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Bingpeng Yan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ruirong Tan
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Anqi Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Wenbo Ma
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Xin Zhu
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Zhujun Yin
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, The “Double-First Class” Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, China
| | - Chunhua Ma
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
6
|
Yang F, Shu R, Dai W, Li B, Liu C, Yang H, Johnson HM, Yu S, Bai D, Yang W, Deng Y. H 2Se-evolving bio-heterojunctions promote cutaneous regeneration in infected wounds by inhibiting excessive cellular senescence. Biomaterials 2024; 311:122659. [PMID: 38861831 DOI: 10.1016/j.biomaterials.2024.122659] [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: 01/25/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024]
Abstract
Pathogenic infection leads to excessive senescent cell accumulation and stagnation of wound healing. To address these issues, we devise and develop a hydrogen selenide (H2Se)-evolving bio-heterojunction (bio-HJ) composed of graphene oxide (GO) and FeSe2 to deracinate bacterial infection, suppress cellular senescence and remedy recalcitrant infected wounds. Excited by near-infrared (NIR) laser, the bio-HJ exerts desired photothermal and photodynamic effects, resulting in rapid disinfection. The crafted bio-HJ could also evolve gaseous H2Se to inhibit cellular senescence and dampen inflammation. Mechanism studies reveal the anti-senescence effects of H2Se-evolving bio-HJ are mediated by selenium pathway and glutathione peroxidase 1 (GPX1). More critically, in vivo experiments authenticate that the H2Se-evolving bio-HJ could inhibit cellular senescence and potentiate wound regeneration in rats. As envisioned, our work not only furnishes the novel gasotransmitter-delivering bio-HJ for chronic infected wounds, but also gets insight into the development of anti-senescence biomaterials.
Collapse
Affiliation(s)
- Fan Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rui Shu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenyu Dai
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bin Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chuang Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China
| | - Hang Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China
| | - Hannah M Johnson
- Department of Chemistry, Washington State University, Washington, USA
| | - Sheng Yu
- Department of Chemistry, Washington State University, Washington, USA
| | - Ding Bai
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Weizhong Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China.
| | - Yi Deng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China; Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
7
|
Vitaliti A, Reggio A, Palma A. Macrophages and autophagy: partners in crime. FEBS J 2024. [PMID: 39439196 DOI: 10.1111/febs.17305] [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: 09/02/2024] [Revised: 09/25/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
Macrophages and autophagy are intricately linked, both playing vital roles in maintaining homeostasis and responding to disease. Macrophages, known for their 'eating' function, rely on a sophisticated digestion system to process a variety of targets, from apoptotic cells to pathogens. The connection between macrophages and autophagy is established early in their development, influencing both differentiation and mature functions. Autophagy regulates essential immune functions, such as inflammation control, pathogen clearance, and antigen presentation, linking innate and adaptive immunity. Moreover, it modulates cytokine production, ensuring a balanced inflammatory response that prevents excessive tissue damage. Autophagy also plays a critical role in macrophage polarization, influencing their shift between pro-inflammatory and anti-inflammatory states. This review explores the role of autophagy in macrophages, emphasizing its impact across various tissues and pathological conditions, and detailing the cellular and molecular mechanisms by which autophagy shapes macrophage function.
Collapse
Affiliation(s)
- Alessandra Vitaliti
- Department of Chemical Science and Technologies, "Tor Vergata" University of Rome, Italy
| | - Alessio Reggio
- Saint Camillus International University of Health Sciences, Rome, Italy
| | - Alessandro Palma
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Italy
| |
Collapse
|
8
|
Huang CG, Hsieh MJ, Wu YC, Huang PW, Lin YJ, Tsao KC, Shih SR, Lee LA. Influence of Donor-Specific Characteristics on Cytokine Responses in H3N2 Influenza A Virus Infection: New Insights from an Ex Vivo Model. Int J Mol Sci 2024; 25:10941. [PMID: 39456722 PMCID: PMC11507259 DOI: 10.3390/ijms252010941] [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: 09/05/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Influenza A virus (IAV) is known for causing seasonal epidemics ranging from flu to more severe outcomes like pneumonia, cytokine storms, and acute respiratory distress syndrome. The innate immune response and inflammasome activation play pivotal roles in sensing, preventing, and clearing the infection, as well as in the potential exacerbation of disease progression. This study examines the complex relationships between donor-specific characteristics and cytokine responses during H3N2 IAV infection using an ex vivo model. At 24 h post infection in 31 human lung explant tissue samples, key cytokines such as interleukin (IL)-6, IL-10, tumor necrosis factor-alpha (TNF-α), and interferon-gamma (IFN-γ) were upregulated. Interestingly, a history of lung cancer did not impact the acute immune response. However, cigarette smoking and programmed death-ligand 1 (PD-L1) expression on macrophages significantly increased IL-2 levels. Conversely, age inversely affected IL-4 levels, and diabetes mellitus negatively influenced IL-6 levels. Additionally, both diabetes mellitus and programmed cell death protein 1 (PD-1) expression on CD3+/CD4+ T cells negatively impacted TNF-α levels, while body mass index was inversely associated with IFN-γ production. Toll-like receptor 2 (TLR2) expression emerged as crucial in mediating acute innate and adaptive immune responses. These findings highlight the intricate interplay between individual physiological traits and immune responses during influenza infection, underscoring the importance of tailored and personalized approaches in IAV treatment and prevention.
Collapse
Affiliation(s)
- Chung-Guei Huang
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 33305, Taiwan; (C.-G.H.); (P.-W.H.); (Y.-J.L.); (K.-C.T.); (S.-R.S.)
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan 33302, Taiwan
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ming-Ju Hsieh
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 33305, Taiwan; (M.-J.H.); (Y.-C.W.)
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yi-Cheng Wu
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 33305, Taiwan; (M.-J.H.); (Y.-C.W.)
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- School of Medicine, College of Life Science and Medicine, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Po-Wei Huang
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 33305, Taiwan; (C.-G.H.); (P.-W.H.); (Y.-J.L.); (K.-C.T.); (S.-R.S.)
| | - Ya-Jhu Lin
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 33305, Taiwan; (C.-G.H.); (P.-W.H.); (Y.-J.L.); (K.-C.T.); (S.-R.S.)
| | - Kuo-Chien Tsao
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 33305, Taiwan; (C.-G.H.); (P.-W.H.); (Y.-J.L.); (K.-C.T.); (S.-R.S.)
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan 33302, Taiwan
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan 33302, Taiwan
| | - Shin-Ru Shih
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 33305, Taiwan; (C.-G.H.); (P.-W.H.); (Y.-J.L.); (K.-C.T.); (S.-R.S.)
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan 33302, Taiwan
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan 33302, Taiwan
| | - Li-Ang Lee
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- School of Medicine, College of Life Science and Medicine, National Tsing Hua University, Hsinchu 300044, Taiwan
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Linkou Main Branch, Taoyuan 33305, Taiwan
| |
Collapse
|
9
|
Xue JD, Gao J, Tang AF, Feng C. Shaping the immune landscape: Multidimensional environmental stimuli refine macrophage polarization and foster revolutionary approaches in tissue regeneration. Heliyon 2024; 10:e37192. [PMID: 39296009 PMCID: PMC11408064 DOI: 10.1016/j.heliyon.2024.e37192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/21/2024] Open
Abstract
In immunology, the role of macrophages extends far beyond their traditional classification as mere phagocytes; they emerge as pivotal architects of the immune response, with their function being significantly influenced by multidimensional environmental stimuli. This review investigates the nuanced mechanisms by which diverse external signals ranging from chemical cues to physical stress orchestrate macrophage polarization, a process that is crucial for the modulation of immune responses. By transitioning between pro-inflammatory (M1) and anti-inflammatory (M2) states, macrophages exhibit remarkable plasticity, enabling them to adapt to and influence their surroundings effectively. The exploration of macrophage polarization provides a compelling narrative on how these cells can be manipulated to foster an immune environment conducive to tissue repair and regeneration. Highlighting cutting-edge research, this review presents innovative strategies that leverage the dynamic interplay between macrophages and their environment, proposing novel therapeutic avenues that harness the potential of macrophages in regenerative medicine. Moreover, this review critically evaluates the current challenges and future prospects of translating macrophage-centered strategies from the laboratory to clinical applications.
Collapse
Affiliation(s)
- Jing-Dong Xue
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Jing Gao
- Department of Obstetrics and Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ai-Fang Tang
- Department of Geratology, Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Chao Feng
- Department of Reproductive Medicine, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai 200030, China
| |
Collapse
|
10
|
Peng S, Fu H, Li R, Li H, Wang S, Li B, Sun J. A new direction in periodontitis treatment: biomaterial-mediated macrophage immunotherapy. J Nanobiotechnology 2024; 22:359. [PMID: 38907216 PMCID: PMC11193307 DOI: 10.1186/s12951-024-02592-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 05/28/2024] [Indexed: 06/23/2024] Open
Abstract
Periodontitis is a chronic inflammation caused by a bacterial infection and is intimately associated with an overactive immune response. Biomaterials are being utilized more frequently in periodontal therapy due to their designability and unique drug delivery system. However, local and systemic immune response reactions driven by the implantation of biomaterials could result in inflammation, tissue damage, and fibrosis, which could end up with the failure of the implantation. Therefore, immunological adjustment of biomaterials through precise design can reduce the host reaction while eliminating the periodontal tissue's long-term chronic inflammation response. It is important to note that macrophages are an active immune system component that can participate in the progression of periodontal disease through intricate polarization mechanisms. And modulating macrophage polarization by designing biomaterials has emerged as a new periodontal therapy technique. In this review, we discuss the role of macrophages in periodontitis and typical strategies for polarizing macrophages with biomaterials. Subsequently, we discuss the challenges and potential opportunities of using biomaterials to manipulate periodontal macrophages to facilitate periodontal regeneration.
Collapse
Affiliation(s)
- Shumin Peng
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 45000, China
- Academy of Medical Sciences at Zhengzhou University, Zhengzhou, 45000, China
| | - Haojie Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 45000, China
- Academy of Medical Sciences at Zhengzhou University, Zhengzhou, 45000, China
| | - Rui Li
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 45000, China
| | - Hui Li
- Beijing Shijitan Hospital, Capital Medical University, Beijing, 100069, China
| | - Shuyuan Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 45000, China
- Academy of Medical Sciences at Zhengzhou University, Zhengzhou, 45000, China
| | - Bingyan Li
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 45000, China
- Academy of Medical Sciences at Zhengzhou University, Zhengzhou, 45000, China
| | - Jingjing Sun
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 45000, China.
| |
Collapse
|
11
|
Winer BY, Settle AH, Yakimov AM, Jeronimo C, Lazarov T, Tipping M, Saoi M, Sawh A, Sepp ALL, Galiano M, Perry JSA, Wong YY, Geissmann F, Cross J, Zhou T, Kam LC, Pasolli HA, Hohl T, Cyster JG, Weiner OD, Huse M. Plasma membrane abundance dictates phagocytic capacity and functional cross-talk in myeloid cells. Sci Immunol 2024; 9:eadl2388. [PMID: 38848343 PMCID: PMC11485225 DOI: 10.1126/sciimmunol.adl2388] [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: 10/06/2023] [Accepted: 05/15/2024] [Indexed: 06/09/2024]
Abstract
Professional phagocytes like neutrophils and macrophages tightly control what they consume, how much they consume, and when they move after cargo uptake. We show that plasma membrane abundance is a key arbiter of these cellular behaviors. Neutrophils and macrophages lacking the G protein subunit Gβ4 exhibited profound plasma membrane expansion, accompanied by marked reduction in plasma membrane tension. These biophysical changes promoted the phagocytosis of bacteria, fungus, apoptotic corpses, and cancer cells. We also found that Gβ4-deficient neutrophils are defective in the normal inhibition of migration following cargo uptake. Sphingolipid synthesis played a central role in these phenotypes by driving plasma membrane accumulation in cells lacking Gβ4. In Gβ4 knockout mice, neutrophils not only exhibited enhanced phagocytosis of inhaled fungal conidia in the lung but also increased trafficking of engulfed pathogens to other organs. Together, these results reveal an unexpected, biophysical control mechanism central to myeloid functional decision-making.
Collapse
Affiliation(s)
- Benjamin Y. Winer
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, CA, USA
- Cardiovascular Research Institute, University of California San Francisco; San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco; San Francisco, CA, USA
| | - Alexander H. Settle
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | | | - Carlos Jeronimo
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Tomi Lazarov
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Murray Tipping
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Michelle Saoi
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | | | - Anna-Liisa L. Sepp
- Department of Biomedical Engineering, Columbia University; New York, NY, USA
| | - Michael Galiano
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Justin S. A. Perry
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Yung Yu Wong
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Frederic Geissmann
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Justin Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Ting Zhou
- Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center; New York, NY, USA
- The SKI Stem Cell Research Facility, The Center for Stem Cell Biology and Developmental Biology Program, Sloan Kettering Institute, 1275 York Avenue, New York, NY 10065, USA
| | - Lance C. Kam
- Department of Biomedical Engineering, Columbia University; New York, NY, USA
| | - H. Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University; New York, NY, USA
| | - Tobias Hohl
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Jason G. Cyster
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, CA, USA
- Howard Hughes Medical Institute; Chevy Chase, MD, USA
| | - Orion D. Weiner
- Cardiovascular Research Institute, University of California San Francisco; San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco; San Francisco, CA, USA
| | - Morgan Huse
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| |
Collapse
|
12
|
Cai C, Zeng W, Wang H, Ren S. Neutrophil-to-Lymphocyte Ratio (NLR), Platelet-to-Lymphocyte Ratio (PLR) and Monocyte-to-Lymphocyte Ratio (MLR) as Biomarkers in Diagnosis Evaluation of Acute Exacerbation of Chronic Obstructive Pulmonary Disease: A Retrospective, Observational Study. Int J Chron Obstruct Pulmon Dis 2024; 19:933-943. [PMID: 38646605 PMCID: PMC11027921 DOI: 10.2147/copd.s452444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/06/2024] [Indexed: 04/23/2024] Open
Abstract
PURPOSE Hierarchical management is advocated in China to effectively manage chronic obstructive pulmonary disease (COPD) patients and reduce the incidence and mortality of acute exacerbation of COPD (AE-COPD). However, primary and community hospitals often have limited access to advanced equipment and technology. Complete blood count (CBC), which is commonly used in these hospitals, offers the advantages of being cost-effective and easily accessible. This study aims to evaluate the significance of routine blood indicators in aiding of diagnosing AE-COPD. PATIENTS AND METHODS In this research, we enrolled a total of 112 patients diagnosed with AE-COPD, 92 patients with stable COPD, and a control group comprising 60 healthy individuals. Clinical characteristics, CBC parameters, and serum CRP levels were collected within two hours. To assess the associations between NLR/PLR/MLR and CRP by Spearman correlation test. The diagnostic accuracy of NLR, PLR and MLR in AE-COPD was assessed using Receiver Operating Characteristic Curve (ROC) and the area under the curve (AUC). Binary Logistic Regression analysis was conducted for the indicators of NLR, PLR and MLR. RESULTS We found that patients with AE-COPD had significantly higher levels of NLR, PLR and MLR in contrast to patients with stable COPD. Additionally, the study revealed a noteworthy correlation between CRP and NLR (rs=0.5319, P<0.001), PLR (rs=0.4424, P<0.001), and MLR (rs=0.4628, P<0.001). By utilizing specific cut-off values, the amalgamation of NLR, PLR and MLR augmented diagnostic sensitivity. Binary logistic regression analysis demonstrated that heightened NLR and MLR act as risk factors for the progression of AE-COPD. CONCLUSION The increasing levels of NLR, PLR and MLR could function as biomarkers, akin to CRP, for diagnosis and assessment of acute exacerbations among COPD patients. Further research is required to validate this concept.
Collapse
Affiliation(s)
- Chuang Cai
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan City, Guangdong Province, People’s Republic of China
| | - Wentan Zeng
- Department of Laboratory Medicine, Tanzhou People’s Hospital of Zhongshan, Zhongshan City Hospital of Integration of TCM & Western Medicine, Zhongshan City, Guangdong Province, People’s Republic of China
| | - Hongwei Wang
- Department of Pediatrics, Tanzhou People’s Hospital of Zhongshan, Zhongshan City hospital of integration of TCM & western medicine, Zhongshan City, Guangdong Province, People’s Republic of China
| | - Shuqi Ren
- Department of Laboratory Medicine, Tanzhou People’s Hospital of Zhongshan, Zhongshan City Hospital of Integration of TCM & Western Medicine, Zhongshan City, Guangdong Province, People’s Republic of China
| |
Collapse
|
13
|
Ji ZZ, Chan MKK, Chan ASW, Leung KT, Jiang X, To KF, Wu Y, Tang PMK. Tumour-associated macrophages: versatile players in the tumour microenvironment. Front Cell Dev Biol 2023; 11:1261749. [PMID: 37965573 PMCID: PMC10641386 DOI: 10.3389/fcell.2023.1261749] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
Tumour-Associated Macrophages (TAMs) are one of the pivotal components of the tumour microenvironment. Their roles in the cancer immunity are complicated, both pro-tumour and anti-cancer activities are reported, including not only angiogenesis, extracellular matrix remodeling, immunosuppression, drug resistance but also phagocytosis and tumour regression. Interestingly, TAMs are highly dynamic and versatile in solid tumours. They show anti-cancer or pro-tumour activities, and interplay between the tumour microenvironment and cancer stem cells and under specific conditions. In addition to the classic M1/M2 phenotypes, a number of novel dedifferentiation phenomena of TAMs are discovered due to the advanced single-cell technology, e.g., macrophage-myofibroblast transition (MMT) and macrophage-neuron transition (MNT). More importantly, emerging information demonstrated the potential of TAMs on cancer immunotherapy, suggesting by the therapeutic efficiency of the checkpoint inhibitors and chimeric antigen receptor engineered cells based on macrophages. Here, we summarized the latest discoveries of TAMs from basic and translational research and discussed their clinical relevance and therapeutic potential for solid cancers.
Collapse
Affiliation(s)
- Zoey Zeyuan Ji
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Max Kam-Kwan Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Alex Siu-Wing Chan
- Department of Applied Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Kam-Tong Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Xiaohua Jiang
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Yi Wu
- MOE Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, China
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| |
Collapse
|
14
|
Winer BY, Settle AH, Yakimov AM, Jeronimo C, Lazarov T, Tipping M, Saoi M, Sawh A, Sepp ALL, Galiano M, Wong YY, Perry JSA, Geissmann F, Cross J, Zhou T, Kam LC, Pasoli HA, Hohl T, Cyster JG, Weiner OD, Huse M. Plasma membrane abundance dictates phagocytic capacity and functional crosstalk in myeloid cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.12.556572. [PMID: 37745515 PMCID: PMC10515848 DOI: 10.1101/2023.09.12.556572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Professional phagocytes like neutrophils and macrophages tightly control what they eat, how much they eat, and when they move after eating. We show that plasma membrane abundance is a key arbiter of these cellular behaviors. Neutrophils and macrophages lacking the G-protein subunit Gb4 exhibit profound plasma membrane expansion due to enhanced production of sphingolipids. This increased membrane allocation dramatically enhances phagocytosis of bacteria, fungus, apoptotic corpses, and cancer cells. Gb4 deficient neutrophils are also defective in the normal inhibition of migration following cargo uptake. In Gb4 knockout mice, myeloid cells exhibit enhanced phagocytosis of inhaled fungal conidia in the lung but also increased trafficking of engulfed pathogens to other organs. These results reveal an unexpected, biophysical control mechanism lying at the heart of myeloid functional decision-making.
Collapse
Affiliation(s)
- Benjamin Y Winer
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, CA, USA
- Cardiovascular Research Institute, University of California San Francisco; San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco; San Francisco, CA, USA
| | - Alexander H Settle
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | | | - Carlos Jeronimo
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Tomi Lazarov
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Murray Tipping
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Michelle Saoi
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | | | - Anna-Liisa L Sepp
- Department of Biomedical Engineering, Columbia University; New York, NY, USA
| | - Michael Galiano
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Yung Yu Wong
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Justin S A Perry
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Frederic Geissmann
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Justin Cross
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Ting Zhou
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Lance C Kam
- Department of Biomedical Engineering, Columbia University; New York, NY, USA
| | - Hilda Amalia Pasoli
- Electron Microscopy Resource Center, The Rockefeller University; New York, NY, USA
| | - Tobias Hohl
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Jason G Cyster
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, CA, USA
- Howard Hughes Medical Institute; Chevy Chase, MD, USA
| | - Orion D Weiner
- Cardiovascular Research Institute, University of California San Francisco; San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco; San Francisco, CA, USA
| | - Morgan Huse
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| |
Collapse
|
15
|
Choi W, Shin WR, Kim YH, Min J. Inducing a Proinflammatory Response with Bioengineered Yeast Vacuoles with TLR2-Binding Peptides (Vac T2BP) as a Drug Carrier for Daunorubicin Delivery. ACS APPLIED MATERIALS & INTERFACES 2023; 15:41258-41270. [PMID: 37615983 DOI: 10.1021/acsami.3c06669] [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: 08/25/2023]
Abstract
Immune adjuvants have roles in immune activation for cancer therapy, and adjuvants derived from microbes have been applied. In this study, we propose the use of bioengineered vacuoles, derived from recombinant yeast with acute myeloid leukemia (AML) specificity and having a TLR-2-binding peptide (VacT2BP) on their surface, to induce a proinflammatory response as a dual-function nanomaterial for daunorubicin (DNR) delivery. Our results demonstrate that nanosized, isolated VacT2BP induced HL-60 cell-specific DNR delivery and apoptosis. Furthermore, we observed the selective release of high-mobility group box 1 from apoptotic HL-60 cells by DNR@VacT2BP. We concluded that DNR@VacT2BP exhibited target selectivity, and the indiscriminate occurrence of damage-associated molecular patterns (DAMPs) was inhibited by the VacT2BP carrier. The therapeutic efficacy of DNR@VacT2BP was confirmed in AML xenograft mice, with about 82% tumor growth inhibition. Following drug delivery, apoptotic cells and DAMPs with residual VacT2BP (apopDNR@VacT2BP) upregulated the proinflammatory immune response of macrophages. In addition, apopDNR@VacT2BP enhanced phagocytosis activity. Macrophages stimulated by apopDNR@VacT2BP suppressed cancer proliferation by about 40%. In summary, our results suggest that dual-functional vacuoles with a target-specific peptide can be a potential strategy for selective drug delivery and construction of an immune environment to fight cancer, thereby improving prognosis.
Collapse
Affiliation(s)
- Wooil Choi
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-Gu Jeonju, Jeonbuk 54896, South Korea
| | - Woo-Ri Shin
- School of Biological Sciences, Chungbuk National University, 1, Chungdae-Ro, Seowon-Gu, Cheongju 28644, South Korea
| | - Yang-Hoon Kim
- School of Biological Sciences, Chungbuk National University, 1, Chungdae-Ro, Seowon-Gu, Cheongju 28644, South Korea
| | - Jiho Min
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-Gu Jeonju, Jeonbuk 54896, South Korea
| |
Collapse
|
16
|
Méndez-Alejandre A, Raymond BBA, Trost M, Marín-Rubio JL. Bi-functional particles for real-time phagosome acidification and proteolysis multiplex assay in macrophages. Front Immunol 2023; 14:1204223. [PMID: 37638042 PMCID: PMC10456865 DOI: 10.3389/fimmu.2023.1204223] [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: 04/11/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023] Open
Abstract
Phagosome acidification and proteolysis are essential processes in the immune response to contain and eliminate pathogens. In recent years, there has been an increased desire for a rapid and accurate method of assessing these processes in real-time. Here, we outline the development of a multiplexed assay that allows simultaneous monitoring of phagosome acidification and proteolysis in the same sample using silica beads conjugated to pHrodo and DQ BSA. We describe in detail how to prepare the bi-functional particles and show proof of concept using differentially activated macrophages. This multiplexed spectrophotometric assay allows rapid and accurate assessment of phagosome acidification and proteolysis in real-time and could provide valuable information for understanding the immune response to pathogen invasion.
Collapse
Affiliation(s)
- Alba Méndez-Alejandre
- Laboratory for Biological Mass Spectrometry, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- Biology Department, Autonomous University of Madrid, Madrid, Spain
| | | | - Matthias Trost
- Laboratory for Biological Mass Spectrometry, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - José Luis Marín-Rubio
- Laboratory for Biological Mass Spectrometry, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| |
Collapse
|
17
|
Wang J, Hu D, Chen Q, Liu T, Zhou X, Xu Y, Zhou H, Gu D, Gao C. Intracellular hydrogelation of macrophage conjugated probiotics for hitchhiking delivery and combined treatment of colitis. Mater Today Bio 2023; 20:100679. [PMID: 37273799 PMCID: PMC10232887 DOI: 10.1016/j.mtbio.2023.100679] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/15/2023] [Accepted: 05/19/2023] [Indexed: 06/06/2023] Open
Abstract
Immune cell membrane coated nanomedicine was developed to neutralize cytokines via receptor-ligand interaction, which showed potential for the treatment of inflammatory bowel disease (IBD). However, cell membrane isolation and re-assembly process involved protein loss and spatial disorder, which reduced the sequestration efficiency towards cytokines. In addition, oral administration of probiotics was accepted for IBD treatment via gut microbiota modulation, but most probiotics showed weak adhesion to intestine mucosa and were quickly expelled from gastrointestinal tract. Herein, an intracellular hydrogelation technology was proposed to construct gelated peritoneal macrophage (GPM) with intact membrane structure, resulting from the avoidance of membrane isolation and re-assembly process. GPM efficiently neutralized multiple cytokines in vitro and in vivo to ameliorate inflammatory Caco-2 cells and colitis rats by regulating oxidative stress, inflammation level and intestinal barrier repair. Moreover, the probiotics (Nissle1917, EcN) were easily attached on GPM surface through specific recognition, to construct GPM-EcN conjugate for GPM hitchhiking delivery to colitis tissue. Conjugation process of GPM and EcN showed no damage on bacterial physiological function. Due to the chemical attachment on inflammatory cells, GPM carried the attached EcN hand-in-hand to accumulate in the colitis tissue of IBD rat, and enhanced intestine retention time of EcN in comparison to free EcN, which improved bacterial diversity, and shifted the microbiota community and acid metabolites to an anti-inflammatory phenotype. This study transferred the hydrogel synthesis from in vitro to intracellular cytoplasm, and came to a new insight of conjugating strategy of GPM and probiotics for hitchhiking delivery and combined anti-IBD treatment.
Collapse
Affiliation(s)
- Jingzhe Wang
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Dini Hu
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qian Chen
- Center for Evolution and Conservation Biology, Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China
| | - Tonggong Liu
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Xiaoting Zhou
- Yulin Center for Food and Drug Control, Yulin, 719000, China
| | - Yong Xu
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Hongzhong Zhou
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Dayong Gu
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Cheng Gao
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| |
Collapse
|
18
|
Wang F, Peters R, Jia J, Mudd M, Salemi M, Allers L, Javed R, Duque TLA, Paddar MA, Trosdal ES, Phinney B, Deretic V. ATG5 provides host protection acting as a switch in the atg8ylation cascade between autophagy and secretion. Dev Cell 2023; 58:866-884.e8. [PMID: 37054706 PMCID: PMC10205698 DOI: 10.1016/j.devcel.2023.03.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/26/2023] [Accepted: 03/20/2023] [Indexed: 04/15/2023]
Abstract
ATG5 is a part of the E3 ligase directing lipidation of ATG8 proteins, a process central to membrane atg8ylation and canonical autophagy. Loss of Atg5 in myeloid cells causes early mortality in murine models of tuberculosis. This in vivo phenotype is specific to ATG5. Here, we show using human cell lines that absence of ATG5, but not of other ATGs directing canonical autophagy, promotes lysosomal exocytosis and secretion of extracellular vesicles and, in murine Atg5fl/fl LysM-Cre neutrophils, their excessive degranulation. This is due to lysosomal disrepair in ATG5 knockout cells and the sequestration by an alternative conjugation complex, ATG12-ATG3, of ESCRT protein ALIX, which acts in membrane repair and exosome secretion. These findings reveal a previously undescribed function of ATG5 in its host-protective role in murine experimental models of tuberculosis and emphasize the significance of the branching aspects of the atg8ylation conjugation cascade beyond the canonical autophagy.
Collapse
Affiliation(s)
- Fulong Wang
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Ryan Peters
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Jingyue Jia
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Michal Mudd
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Michelle Salemi
- Proteomics Core Facility, UC Davis Genome Center, University of California, Davis, Davis, CA 95616, USA
| | - Lee Allers
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Ruheena Javed
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Thabata L A Duque
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Masroor A Paddar
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Einar S Trosdal
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Brett Phinney
- Proteomics Core Facility, UC Davis Genome Center, University of California, Davis, Davis, CA 95616, USA
| | - Vojo Deretic
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA.
| |
Collapse
|
19
|
Nguyen JA, Greene CJ, Yates RM. Eructophagy: macrophages use autophagic machinery to burp out parts of their meal. Autophagy 2023; 19:1042-1044. [PMID: 36264831 PMCID: PMC9980442 DOI: 10.1080/15548627.2022.2138006] [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: 07/21/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/02/2022] Open
Abstract
The phagolysosome is an antimicrobial and degradative organelle that plays key roles in macrophage-mediated inflammatory and homeostatic functions. Whereas mature phagolysosomes are known to sequester and degrade their contents into basic nutrients, they were not previously assigned an active role in amplifying inflammation. We have described a novel macrophage process in which partially digested immunostimulatory PAMPs are released extracellularly from the mature phagolysosome via discrete events we term eructophagy. Eructophagy is induced by proinflammatory stimuli, negatively regulated by IL4 and MTOR, and is dependent on key autophagy proteins, including fusion machinery of degradative and secretory autophagy. We propose that macrophages use eructophagy to release processed PAMPs/DAMPs to amplify local inflammation.
Collapse
Affiliation(s)
- Jenny A. Nguyen
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Catherine J. Greene
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Robin M. Yates
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute of Chronic Disease, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
20
|
Selvapandiyan A, Puri N, Kumar P, Alam A, Ehtesham NZ, Griffin G, Hasnain SE. Zooming in on common immune evasion mechanisms of pathogens in phagolysosomes: potential broad-spectrum therapeutic targets against infectious diseases. FEMS Microbiol Rev 2023; 47:6780197. [PMID: 36309472 DOI: 10.1093/femsre/fuac041] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 10/06/2022] [Accepted: 10/18/2022] [Indexed: 01/19/2023] Open
Abstract
The intracellular viral, bacterial, or parasitic pathogens evade the host immune challenges to propagate and cause fatal diseases. The microbes overpower host immunity at various levels including during entry into host cells, phagosome formation, phagosome maturation, phagosome-lysosome fusion forming phagolysosomes, acidification of phagolysosomes, and at times after escape into the cytosol. Phagolysosome is the final organelle in the phagocyte with sophisticated mechanisms to degrade the pathogens. The immune evasion strategies by the pathogens include the arrest of host cell apoptosis, decrease in reactive oxygen species, the elevation of Th2 anti-inflammatory response, avoidance of autophagy and antigen cross-presentation pathways, and escape from phagolysosomal killing. Since the phagolysosome organelle in relation to infection/cure is seldom discussed in the literature, we summarize here the common host as well as pathogen targets manipulated or utilized by the pathogens established in phagosomes and phagolysosomes, to hijack the host immune system for their benefit. These common molecules or pathways can be broad-spectrum therapeutic targets for drug development for intervention against infectious diseases caused by different intracellular pathogens.
Collapse
Affiliation(s)
| | - Niti Puri
- Cellular and Molecular Immunology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Pankaj Kumar
- Department of Biochemistry, Jamia Hamdard, New Delhi, 110062, India.,Centre for Tuberculosis Research, Department of Medicine, Johns Hopkins University, Baltimore, MD, 21218, United States
| | - Anwar Alam
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India.,Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology-Delhi, New Delhi, 110016, India
| | - Nasreen Zafar Ehtesham
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India
| | - George Griffin
- Department of Cellular and Molecular Medicine, St. George's University of London, London, SW17 0RE, United Kingdom
| | - Seyed Ehtesham Hasnain
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology-Delhi, New Delhi, 110016, India.,Department of Life Science, School of Basic Sciences and Research, Sharda University, Knowledge Park III, Greater Noida, 201310, India
| |
Collapse
|
21
|
Nguyen JA, Greene CJ, Cheung S, Yates RM. Multiplexed Phagosomal Assays for the Detection and Quantification of Bidirectional Exchange Between the Phagolysosomal Lumen and Extracellular Space. Methods Mol Biol 2023; 2692:171-185. [PMID: 37365468 DOI: 10.1007/978-1-0716-3338-0_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
The phagolysosome is an antimicrobial and degradative organelle that plays a key role in macrophage-mediated inflammation and homeostasis. Before being presented to the adaptive immune system, phagocytosed proteins must first be processed into immunostimulatory antigens. Until recently, little attention has been given to how other processed PAMPs and DAMPs can stimulate an immune response if they are sequestered in the phagolysosome. Eructophagy is a newly described process in macrophages that releases partially digested immunostimulatory PAMPs and DAMPs extracellularly from the mature phagolysosome to activate vicinal leukocytes. This chapter outlines approaches to observe and quantify eructophagy by simultaneously measuring several phagosomal parameters of individual phagosomes. These methods use specifically designed experimental particles capable of conjugating to multiple reporter/reference fluors in combination with real-time automated fluorescent microscopy. Through the use of high-content image analysis software, each phagosomal parameter can be evaluated quantitatively or semiquantitatively during post-analysis.
Collapse
Affiliation(s)
- Jenny A Nguyen
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Catherine J Greene
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Samuel Cheung
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Robin M Yates
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada.
- Cumming School of Medicine, Snyder Institute of Chronic Disease, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
22
|
Montanari M, Guescini M, Gundogdu O, Luchetti F, Lanuti P, Ciacci C, Burattini S, Campana R, Ortolani C, Papa S, Canonico B. Extracellular Vesicles from Campylobacter jejuni CDT-Treated Caco-2 Cells Inhibit Proliferation of Tumour Intestinal Caco-2 Cells and Myeloid U937 Cells: Detailing the Global Cell Response for Potential Application in Anti-Tumour Strategies. Int J Mol Sci 2022; 24:ijms24010487. [PMID: 36613943 PMCID: PMC9820799 DOI: 10.3390/ijms24010487] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/15/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022] Open
Abstract
Cytolethal distending toxin (CDT) is produced by a range of Gram-negative pathogenic bacteria such as Campylobacter jejuni. CDT represents an important virulence factor that is a heterotrimeric complex composed of CdtA, CdtB, and CdtC. CdtA and CdtC constitute regulatory subunits whilst CdtB acts as the catalytic subunit exhibiting phosphatase and DNase activities, resulting in cell cycle arrest and cell death. Extracellular vesicle (EV) secretion is an evolutionarily conserved process that is present throughout all kingdoms. Mammalian EVs play important roles in regular cell-to-cell communications but can also spread pathogen- and host-derived molecules during infections to alter immune responses. Here, we demonstrate that CDT targets the endo-lysosomal compartment, partially evading lysosomal degradation and exploiting unconventional secretion (EV release), which is largely involved in bacterial infections. CDT-like effects are transferred by Caco-2 cells to uninfected heterologous U937 and homologous Caco-2 cells. The journey of EVs derived from CDT-treated Caco-2 cells is associated with both intestinal and myeloid tumour cells. EV release represents the primary route of CDT dissemination, revealing an active toxin as part of the cargo. We demonstrated that bacterial toxins could represent suitable tools in cancer therapy, highlighting both the benefits and limitations. The global cell response involves a moderate induction of apoptosis and autophagic features may play a protective role against toxin-induced cell death. EVs from CDT-treated Caco-2 cells represent reliable CDT carriers, potentially suitable in colorectal cancer treatments. Our data present a potential bacterial-related biotherapeutic supporting a multidrug anticancer protocol.
Collapse
Affiliation(s)
- Mariele Montanari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Michele Guescini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Ozan Gundogdu
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Francesca Luchetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Paola Lanuti
- Department of Medicine and Aging Science, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Caterina Ciacci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Sabrina Burattini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Raffaella Campana
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Claudio Ortolani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Stefano Papa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
- Correspondence:
| | - Barbara Canonico
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| |
Collapse
|