1
|
Nakamizo S, Kabashima K. Cutaneous granulomas: mechanisms, cellular interactions and therapeutic insights. Br J Dermatol 2025; 192:974-982. [PMID: 40080709 DOI: 10.1093/bjd/ljaf096] [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: 01/19/2025] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 03/15/2025]
Abstract
Granulomas are specialized biologic defence mechanisms that form in response to infections by pathogens, foreign bodies or specific stimuli such as antimicrobials or fungi. These structures function to isolate foreign materials and pathogens that cannot be eliminated by immune cells, primarily through macrophage activity. In the skin, granulomas are a hallmark of several conditions, including sarcoidosis, granuloma annulare, tuberculosis and leprosy, each exhibiting distinct pathological and immunological features. Granulomas can also arise from lipid accumulation, as observed in xanthogranuloma, or be triggered by inflammatory processes associated with unidentified antigens. Among their cellular components, Langhans-type multinucleated giant cells play a pivotal role in granuloma structure and function, contributing to pathogen containment and tissue remodelling, although their precise mechanisms of action remain an area of active investigation. In addition to these giant cells, recent studies have identified triggering receptors expressed on myeloid cells 2 (TREM2)+ macrophages as key contributors to granuloma formation and maintenance. These macrophages are involved in extracellular degradation of foreign substances and play a role in adapting to the hypoxic and nutrient-poor microenvironment of granulomas through metabolic reprogramming, including the pentose phosphate pathway. Recent advances in molecular biology, such as single-cell RNA sequencing, have provided unprecedented insights into the cellular heterogeneity and molecular pathways involved in granuloma formation. These techniques have revealed disease-specific differences in immune cell profiles and activation states, offering new perspectives on the underlying mechanisms of granulomatous diseases. Despite these advances, the precise processes driving granuloma formation and their functional significance remain largely unclear. This review addresses the central question, 'What is a granuloma?', by synthesizing recent findings, with a particular focus on cutaneous granulomas, and presenting interpretations grounded in the current body of literature. We also discuss the implications of these findings for the development of novel therapeutic strategies, including targeted immunomodulation and cytokine blockade, which hold promise for treating granulomatous diseases while preserving host defence.
Collapse
Affiliation(s)
- Satoshi Nakamizo
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Alliance Laboratory for Advanced Medical Research, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- A*STAR Skin Research Labs (A*SRL), Singapore Immunology Network (SIgN), and Skin Research Institute of Singapore (SRIS), Agency for Science, Technology, and Research (A*STAR), Singapore
| |
Collapse
|
2
|
Froom ZSCS, Callaghan NI, Davenport Huyer L. Cellular crosstalk in fibrosis: insights into macrophage and fibroblast dynamics. J Biol Chem 2025:110203. [PMID: 40334985 DOI: 10.1016/j.jbc.2025.110203] [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: 03/13/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Pathological fibrosis, the excessive deposition of extracellular matrix and tissue stiffening that causes progressive organ dysfunction, underlies diverse chronic diseases. The fibrotic microenvironment is driven by the dynamic microenvironmental interaction between various cell types; macrophages and fibroblasts play central roles in fibrotic disease initiation, maintenance, and progression. Macrophage functional plasticity to microenvironmental stimuli modulates fibroblast functionality by releasing pro-inflammatory cytokines, growth factors, and matrix remodeling enzymes that promote fibroblast proliferation, activation, and differentiation into myofibroblasts. Activated fibroblasts and myofibroblasts serve as the fibrotic effector cells, secreting extracellular matrix components and initiating microenvironmental contracture. Fibroblasts also modulate macrophage function through the release of their own pro-inflammatory cytokines and growth factors, creating bidirectional crosstalk that reinforces the chronic fibrotic cycle. The intricate interplay between macrophages and fibroblasts, including their secretomes and signaling interactions, leads to tissue damage and pathological loss of tissue function. In this review, we examine macrophage-fibroblast reciprocal dynamic interactions in pathological fibrotic conditions. We discuss the specific lineages and functionality of macrophages and fibroblasts implicated in fibrotic progression, with focus on their signal transduction pathways and secretory signalling that enables their pro-fibrotic behaviour. We then finish with a set of recommendations for future experimentation with the goal of developing a set of potential targets for anti-fibrotic therapeutic candidates. Understanding the cellular interactions between macrophages and fibroblasts provides valuable insights into potential therapeutic strategies to mitigate fibrotic disease progression.
Collapse
Affiliation(s)
- Zachary S C S Froom
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Neal I Callaghan
- Department of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Locke Davenport Huyer
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS B3H 4R2, Canada; Nova Scotia Health, Halifax, NS B3S 0H6, Canada.
| |
Collapse
|
3
|
Stefanelli V, Lombardi J, Ferrer J, Gardocki-Sandor M. Vascularization of Human Acellular Dermal Matrices: A Comparative Study in a Nonhuman Primate Model. Tissue Eng Part A 2025; 31:419-432. [PMID: 39041614 DOI: 10.1089/ten.tea.2024.0059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024] Open
Abstract
Four human acellular dermal matrices (hADMs) were characterized in a nonhuman primate abdominal wall repair model by evaluating host immune response, vascularization, and incorporation into host tissues. AlloDerm™ (electron beam-sterilized hADM [e-hADM]), AlloMax™ (gamma beam-sterilized hADM, freeze-dried [g-hADM-FD]), DermaMatrix™ (hADM, freeze-dried [hADM-FD]), and FlexHD™ (ethanol-treated hADM [EtOH-hADM]) were each implanted in an abdominal wall-bridging defect in nonhuman primates (n = 3 animals/time point, n = 36 animals). Immunohistochemical and histological assessments were conducted on biopsies from each hADM at 1-, 3-, and 6-months postimplantation to assess vascularization (hematoxylin and eosin [H&E], CD31, alpha smooth muscle actin [αSMA], collagen IV), inflammatory/immune response (H&E, CD3, CD20, CD68), and collagen turnover (H&E, matrix metalloproteinase-9 [MMP-9]). MMP-9 immunolabeling was similar among different hADMs at 1 month; however, hADM-FD and EtOH-hADM showed higher total mean MMP-9-immunopositive areas at approximately 16% compared with <1% for e-hADM and g-hADM at 6 months postimplantation. Cells that stained positively for CD68, CD3, and CD20 were generally higher for hADM-FD and EtOH-hADM compared with other hADMs. The mean CD31-immunopositive area, CD31 vessel density, CD31 vessel diameter, and collagen IV-immunopositive area increased over time. Among all the hADM types, e-hADM had the highest mean (±standard deviation [SD]) CD31-immunopositive area at 1.54% ± 1.01%, vessel density at 7.86 × 10-5 ± 3.96 × 10-5 vessels/µm2, and collagen IV-immunopositive area at 2.55% ± 0.73% 1-month postimplantation. The pattern of αSMA immunolabeling varied among the hADMs. Histology showed that overall inflammation was mild at 1 month. Overall fibroblast repopulation and collagen remodeling increased over time from 1 to 6 months postimplantation. Fibroblast infiltration was minimal to mild at 1 month, with e-hADM showing the highest mean (±SD) score at 2.00 ± 0.00 compared with other hADMs. Only hADM-FD was not completely replaced by neotissue formation at 6 months postimplantation. All hADMs promoted vascularization, cell infiltration, and incorporation into host tissue, which were associated with acute inflammation and immune responses, within a 6-month period. A trend toward relatively enhanced early vascularization in e-hADM compared with other hADMs was observed. Immunogenic responses among the hADMs in the present study showed a slight distinction toward more quiescent terminally sterilized hADMs (e-hADM, g-hADM-FD) versus aseptically processed hADMs (EtOH-hADM, hADM-FD).
Collapse
Affiliation(s)
| | - Jared Lombardi
- Allergan Aesthetics, an AbbVie Company, Branchburg, New Jersey, USA
| | - Joselito Ferrer
- Allergan Aesthetics, an AbbVie Company, Branchburg, New Jersey, USA
| | | |
Collapse
|
4
|
Lösslein AK, Henneke P. Macrophage Differentiation and Metabolic Adaptation in Mycobacterial Infections. Annu Rev Immunol 2025; 43:423-450. [PMID: 40014665 DOI: 10.1146/annurev-immunol-082323-120757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
The adaptation of macrophages-the most common tissue-resident immune cells-to metabolic and microbial cues with high local variability is essential for the maintenance of organ integrity. In homeostasis, macrophages show largely predictable tissue-specific differentiation, as recently revealed by multidimensional methods. However, chronic infections with human-adapted pathogens substantially contribute to the differentiation complexity of tissue macrophages, which has been only partially resolved. Specifically, the response to mycobacterial species-which range from Mycobacterium tuberculosis (with highest specificity for humans, broad organ tropism, yet tissue-specific disease phenotypes) to environmental mycobacteria with humans as accidental hosts-may serve as a paradigm of tissue macrophage adaptation mechanisms. While mycobacterial species-specific tissue preferences are partially related to the mode of acquisition and pathogen characteristics, evolutionary convergence with macrophages driven by metabolic features of the target organ likely contributes to infection resistance and immunopathology. In this review, we unravel the mechanisms of tissue-specific macrophage differentiation and its limitations in mycobacterial infections.
Collapse
Affiliation(s)
- Anne Kathrin Lösslein
- Institute for Infection Prevention and Control, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany;
- Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp Henneke
- Institute for Infection Prevention and Control, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany;
- Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute for Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
5
|
Conner TS, Baaijens FPT, Bouten CVC, Angeloni L, Smits AIPM. A call for standardization: Evaluating different methodologies to induce in vitro foreign body giant cell formation for biomaterials research and design. Acta Biomater 2025; 194:20-37. [PMID: 39826854 DOI: 10.1016/j.actbio.2025.01.026] [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: 10/08/2024] [Revised: 12/20/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Foreign body giant cells (FBGCs) are crucial in the foreign body reaction at the biomaterial-tissue interface, forming through the fusion of cells from the monocyte/macrophage lineage and performing functions such as material degradation and fibrous encapsulation. Yet, their presence and role in biomaterials research is only slowly unveiled. This review analyzed existing FBGC literature identified through a search string and sources from FBGC articles to evaluate the most commonly used methods and highlight the challenges in establishing a standardized protocol. Our findings revealed a fragmented research landscape marked by significant variability in in vitro culture conditions, i.e., cell origin and type, culture media and sera, fusion-inducing factors, seeding density, culture surface, and inconsistencies in the read-outs. This complicates efforts toward standardization and hampers cross-study comparisons. Based on these results, we highlight the need and propose guidelines for standardized culture protocols for FBGC research. Overall, this review aims to underscore the relevance of improving reproducibility and reliability in FBGC research, facilitating effective cross-study comparisons and advancing understanding of FBGC formation and function, ultimately contributing to designing more effective biomaterial-based therapies. STATEMENT OF SIGNIFICANCE: Foreign body giant cells (FBGCs) are crucial in the body's response to implanted biomaterials. Yet, current research addressing their role and impact is highly fragmented. This review comprehensively and systematically examines the diverse methodologies and definitions used in FBGC research and identifies critical gaps and inconsistencies hindering the reproducibility and comparison of findings. By advocating for standardized protocols, we aim to enhance the reliability and equivalence of research, thus providing a stronger foundation for understanding biomaterial-driven FBGC formation and function. Establishing such a framework will impact biomaterial-based therapies, supporting their effectiveness and safety in medical applications, and is thus of relevance for scientists, companies, and clinicians in the biomaterial and medical device communities.
Collapse
Affiliation(s)
- Thijs S Conner
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Frank P T Baaijens
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Livia Angeloni
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands; Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, Rome, Italy
| | - Anthal I P M Smits
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands.
| |
Collapse
|
6
|
Paul K, Darzi S, O'Connell CD, Hennes DMZB, Rosamilia A, Gargett CE, Werkmeister JA, Mukherjee S. 3D Printed Mesh Geometry Modulates Immune Response and Interface Biology in Mouse and Sheep Model: Implications for Pelvic Floor Surgery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405004. [PMID: 39297316 PMCID: PMC11923936 DOI: 10.1002/advs.202405004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/13/2024] [Indexed: 03/21/2025]
Abstract
Pelvic organ prolapse (POP) is a highly prevalent yet neglected health burden for women. Strengthening the pelvic floor with bioactive tissue-engineered meshes is an emerging concept. This study investigates tissue regenerative design parameters, including degradability, porosity, and angulation, to develop alternative degradable melt electrowritten (MEW) constructs for surgical applications of POP. MEW constructs were fabricated in hierarchical geometries by two-way stacking of the fibers with three different inter layer angles of 90°, 45°, or 22.5°. Implants printed at 22.5° have higher tensile strength under dry conditions and show better vaginal fibroblast (VF) attachment in vitro. In vivo assessment using preclinical mouse and ovine models demonstrates more effective degradation and improved tissue integration in 22.5° angular meshes compared to 90° and 45° meshes, with evidence of neo-collagen deposition within implants at 6 weeks. The pattern and geometry of the layered MEW implants also influence the foreign body response, wherein the anti-inflammatory phenotype shows a greater ratio of anti-inflammatory CD206+ M2 macrophages/pro-inflammatory CCR7+ M1 macrophages. This presents an attractive strategy for improving the design and fabrication of next-generation vaginal implants for pelvic reconstructive surgery.
Collapse
Affiliation(s)
- Kallyanashis Paul
- The Ritchie CentreHudson Institute of Medical ResearchClayton3168Australia
- Department of Obstetrics and GynaecologyMonash UniversityClayton3168Australia
| | - Saeedeh Darzi
- The Ritchie CentreHudson Institute of Medical ResearchClayton3168Australia
- Department of Obstetrics and GynaecologyMonash UniversityClayton3168Australia
| | - Cathal D. O'Connell
- Biofab3D@ACMDSt Vincent's HospitalMelbourneVIC3065Australia
- Discipline of Electrical and Biomedical EngineeringSchool of EngineeringRMIT UniversityMelbourneVIC3000Australia
| | - David M. Z. B. Hennes
- The Ritchie CentreHudson Institute of Medical ResearchClayton3168Australia
- Department of Obstetrics and GynaecologyMonash UniversityClayton3168Australia
- Pelvic Floor Disorders UnitMonash HealthClaytonVIC3168Australia
| | - Anna Rosamilia
- The Ritchie CentreHudson Institute of Medical ResearchClayton3168Australia
- Department of Obstetrics and GynaecologyMonash UniversityClayton3168Australia
- Pelvic Floor Disorders UnitMonash HealthClaytonVIC3168Australia
| | - Caroline E. Gargett
- The Ritchie CentreHudson Institute of Medical ResearchClayton3168Australia
- Department of Obstetrics and GynaecologyMonash UniversityClayton3168Australia
| | - Jerome A Werkmeister
- The Ritchie CentreHudson Institute of Medical ResearchClayton3168Australia
- Department of Obstetrics and GynaecologyMonash UniversityClayton3168Australia
| | - Shayanti Mukherjee
- The Ritchie CentreHudson Institute of Medical ResearchClayton3168Australia
- Department of Obstetrics and GynaecologyMonash UniversityClayton3168Australia
| |
Collapse
|
7
|
Sabe H, Yahara Y, Ishii M. Cell fusion dynamics: mechanisms of multinucleation in osteoclasts and macrophages. Inflamm Regen 2024; 44:49. [PMID: 39605032 PMCID: PMC11600601 DOI: 10.1186/s41232-024-00360-3] [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: 08/05/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Cell-cell fusion is a vital biological process where the membranes of two or more cells merge to form a syncytium. This phenomenon is critical in various physiological and pathological contexts, including embryonic development, tissue repair, immune responses, and the progression of several diseases. Osteoclasts, which are cells from the monocyte/macrophage lineage responsible for bone resorption, have enhanced functionality due to cell fusion. Additionally, other multinucleated giant cells (MGCs) also arise from the fusion of monocytes and macrophages, typically during chronic inflammation and reactions to foreign materials such as prostheses or medical devices. Foreign body giant cells (FBGCs) and Langhans giant cells (LGCs) emerge only under pathological conditions and are involved in phagocytosis, antigen presentation, and the secretion of inflammatory mediators. This review provides a comprehensive overview of the mechanisms underlying the formation of multinucleated cells, with a particular emphasis on macrophages and osteoclasts. Elucidating the intracellular structures, signaling cascades, and fusion-mediating proteins involved in cell-cell fusion enhances our understanding of this fundamental biological process and helps identify potential therapeutic targets for disorders mediated by cell fusion.
Collapse
Affiliation(s)
- Hideaki Sabe
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yasuhito Yahara
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan.
- WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan.
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
8
|
Hancock TJ, Vlasyuk M, Foster JS, Macy S, Wooliver DC, Balachandran M, Williams AD, Martin EB, Kennel SJ, Heidel ER, Wall JS, Jackson JW. Neutrophils enhance the clearance of systemic amyloid deposits in a murine amyloidoma model. Front Immunol 2024; 15:1487250. [PMID: 39600710 PMCID: PMC11588727 DOI: 10.3389/fimmu.2024.1487250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Introduction Amyloid-specific antibodies have been shown to opsonize and enhance amyloid clearance in systemic amyloidosis mouse models. However, the immunological mechanisms by which amyloid is removed have not been clearly defined. Previous reports from preclinical in vivo studies suggest polymorphonuclear cells (i.e., neutrophils) can affect amyloid removal. Therefore, we sought to analyze how neutrophils may contribute to the clearance of human AL amyloid extracts, using a murine amyloidoma model. Methods Immunocompromised nude mice injected subcutaneously with patient-derived AL amyloid extract (generating a localized "amyloidoma") were used to circumvent confounding factors contributed by the adaptive immune system and served as the model system. Two representative AL amyloid extracts were used, ALλ(CLA), which is refractory to clearance, and ALκ(TAL), which is readily cleared in mice. Neutrophil recruitment to the amyloid masses, cellular activation, and propensity to engulf amyloid were assessed. Results Immunophenotyping of amyloidomas from animals implanted with 2 mg of either ALλ or ALκ revealed that more neutrophils were recruited to ALκ amyloid masses as compared to the ALλ material, which was generally devoid of neutrophils. Ex vivo analyses indicated neutrophils do not efficiently phagocytose amyloid directly. However, histological evaluation of the ALκ amyloidoma revealed the abundant presence of neutrophil extracellular traps, which were absent in the ALλ amyloidomas. Using neutrophil depletion experiments in mice, we determined that mice devoid of neutrophils cleared the human amyloid lesions less efficiently. Moreover, mice devoid of neutrophils also had significantly reduced intra-amyloid expression of inflammatory cytokines. Discussion Neutrophils may not directly mediate amyloid clearance through phagocytosis; however, these cells can be stimulated by the amyloid and may function to facilitate phagocytosis and amyloid clearance by professional phagocytes (e.g., macrophages).
Collapse
Affiliation(s)
- Trevor J. Hancock
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Marina Vlasyuk
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - James S. Foster
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Sallie Macy
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Daniel C. Wooliver
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Manasi Balachandran
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Angela D. Williams
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Emily B. Martin
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Stephen J. Kennel
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Eric R. Heidel
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Jonathan S. Wall
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Joseph W. Jackson
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| |
Collapse
|
9
|
Cai F, Jiang B, He F. Formation and biological activities of foreign body giant cells in response to biomaterials. Acta Biomater 2024; 188:1-26. [PMID: 39245307 DOI: 10.1016/j.actbio.2024.08.034] [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: 03/09/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 09/10/2024]
Abstract
The integration of biomaterials in medical applications triggers the foreign body response (FBR), a multi-stage immune reaction characterized by the formation of foreign body giant cells (FBGCs). Originating from the fusion of monocyte/macrophage lineage cells, FBGCs are pivotal participants during tissue-material interactions. This review provides an in-depth examination of the molecular processes during FBGC formation, highlighting signaling pathways and fusion mediators in response to both exogenous and endogenous stimuli. Moreover, a wide range of material-specific characteristics, such as surface chemical and physical properties, has been proven to influence the fusion of macrophages into FBGCs. Multifaceted biological activities of FBGCs are also explored, with emphasis on their phagocytic capabilities and extracellular secretory functions, which profoundly affect the vascularization, degradation, and encapsulation of the biomaterials. This review further elucidates the heterogeneity of FBGCs and their diverse roles during FBR, as demonstrated by their distinct behaviors in response to different materials. By presenting a comprehensive understanding of FBGCs, this review intends to provide strategies and insights into optimizing biocompatibility and the therapeutic potential of biomaterials for enhanced stability and efficacy in clinical applications. STATEMENT OF SIGNIFICANCE: As a hallmark of the foreign body response (FBR), foreign body giant cells (FBGCs) significantly impact the success of implantable biomaterials, potentially leading to complications such as chronic inflammation, fibrosis, and device failure. Understanding the role of FBGCs and modulating their responses are vital for successful material applications. This review provides a comprehensive overview of the molecules and signaling pathways guiding macrophage fusion into FBGCs. By elucidating the physical and chemical properties of materials inducing distinct levels of FBGCs, potential strategies of materials in modulating FBGC formation are investigated. Additionally, the biological activities of FBGCs and their heterogeneity in responses to different material categories in vivo are highlighted in this review, offering crucial insights for improving the biocompatibility and efficacy of biomaterials.
Collapse
Affiliation(s)
- Fangyuan Cai
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Bulin Jiang
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Fuming He
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
10
|
Fu X, Ye F, Wan Y, Xi G, Hua Y, Keep RF. The Role of Complement C1qa in Experimental Intracerebral Hemorrhage. Transl Stroke Res 2024:10.1007/s12975-024-01302-4. [PMID: 39370487 DOI: 10.1007/s12975-024-01302-4] [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: 08/08/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/08/2024]
Abstract
Evidence indicates that the complement system is activated and plays a role in brain injury after intracerebral hemorrhage (ICH). Most studies have focused on the role of C3, C5 and the membrane attack complex. The purpose of this study was to investigate the potential impact of complement C1q, a key upstream component of the classical pathway, on ICH-induced brain injury. Wild-type (WT) and C1qa knock out (KO) mice were compared using an autologous blood injection ICH model. Magnetic resonance imaging (MRI) was performed on days 1, 3 and 7 and brains harvested on days 3 and 7 for immunohistochemistry to examine brain injury mechanisms. WT and C1qa KO mice also received an intracerebral injection of thrombin, a key factor in ICH-induced brain injury. Following MRI scans, brains were harvested for immunohistochemistry on day 1. In comparison to WT mice, C1qa KO mice had reduced hematoma erythrolysis and neutrophil infiltration after ICH. However, they also had delayed hematoma clearance, which was associated with reduced induction of phagocytic multinuclear giant cells, and increased perihematomal neuronal damage. After thrombin injection, C1qa KO mice had smaller lesion volumes, less neuronal loss, reduced neutrophil infiltration, and less BBB damage. C1qa knockout has beneficial and detrimental effects on ICH-induced brain injury mechanisms, but a consistent beneficial effect after thrombin injection. Strategies to balance the roles of C1q after ICH may represent a promising therapeutic direction.
Collapse
Affiliation(s)
- Xiongjie Fu
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
- Department of Neurosurgery, the 2nd Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Fenghui Ye
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Yingfeng Wan
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, R5018 BSRB 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
11
|
Xia X, Yang Y, Liu P, Chen L, Dai X, Xue P, Wang Y. The senolytic drug ABT-263 accelerates ovarian aging in older female mice. Sci Rep 2024; 14:23178. [PMID: 39369073 PMCID: PMC11457520 DOI: 10.1038/s41598-024-73828-4] [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: 05/21/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024] Open
Abstract
Previous studies have reported that senolytic drugs can reverse obesity-mediated accumulation of senescent cells in the ovary and protect against cisplatin-induced ovarian injury by removing senescent cells. Early intervention with ABT-263 has been shown to mitigate ovarian aging. However, it remains unknown whether treatment with ABT-263 could rejuvenate the aged ovary in reproductively old females. Therefore, the current study was aimed to investigate whether advanced age intervention with ABT-263 could ameliorate age-related decline in ovarian function. Fourteen 16-month-old mice with a C57/BL6 background were treated with ABT-263 (N = 7) or vehicle (N = 7) for two weeks. Mice were initially treated with ABT-263 (60 mg/kg/d) or vehicle for 7 consecutive days. After a 7-day break, the treatment was repeated for another 7 consecutive days. Six 2-month-old mice with C57BL/6 were used as a young control. The hormonal levels, estrus cycles, ovarian reserve, ovarian cell proliferation and apoptosis, ovarian fibrosis, and steroidogenic gene expression of ovarian stromal cells were evaluated. ABT-263 treatment did not rescue abnormal estrus cycles and sex hormonal levels, or inhibit the formation of multinucleated giant cells and ovarian stromal cell apoptosis in aged ovaries. However, it reduced ovarian fibrosis and preserved the steroidogenic gene expression of ovarian stromal cells in aged ovaries. Importantly, ABT-263 treatment further depleted ovarian follicles in aged mice. In conclusion, ABT-263 treatment accelerated the depletion of ovarian follicles in aged mice, suggesting that senolytic drugs for reproductively old female may adversely affect female fertility.
Collapse
Affiliation(s)
- Xiyang Xia
- The Center for Reproductive Medicine, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Yingying Yang
- The Center for Reproductive Medicine, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Pengfei Liu
- The Department of Animal Center, Kebiao Medical Testing Center, Changzhou, Jiangsu, China
| | - Li Chen
- The Center for Reproductive Medicine, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Xiuliang Dai
- The Center for Reproductive Medicine, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China.
| | - Pingping Xue
- The Center for Reproductive Medicine, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China.
| | - Yufeng Wang
- The Center for Reproductive Medicine, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China.
| |
Collapse
|
12
|
Fu X, Wang M, Wan Y, Hua Y, Keep RF, Xi G. Formation of Multinucleated Giant Cells after Experimental Intracerebral Hemorrhage: Characteristics and Role of Complement C3. Biomedicines 2024; 12:1251. [PMID: 38927458 PMCID: PMC11201741 DOI: 10.3390/biomedicines12061251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/16/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Hematoma clearance is critical for mitigating intracerebral hemorrhage (ICH)-induced brain injury. Multinucleated giant cells (MGCs), a type of phagocyte, and the complement system may play a pivotal role in hematoma resolution, but whether the complement system regulates MGC formation after ICH remains unclear. The current study investigated the following: (1) the characteristics of MGC formation after ICH, (2) whether it was impacted by complement C3 deficiency in mice and (3) whether it also influenced hematoma degradation (hemosiderin formation). Young and aged male mice, young female mice and C3-deficient and -sufficient mice received a 30 μL injection of autologous whole blood into the right basal ganglia. Brain histology and immunohistochemistry were used to examine MGC formation on days 3 and 7. Hemosiderin deposition was examined by autofluorescence on day 28. Following ICH, MGCs were predominantly located in the peri-hematoma region exhibiting multiple nuclei and containing red blood cells or their metabolites. Aging was associated with a decrease in MGC formation after ICH, while sex showed no discernible effect. C3 deficiency reduced MGC formation and reduced hemosiderin formation. Peri-hematomal MGCs may play an important role in hematoma resolution. Understanding how aging and complement C3 impact MGCs may provide important insights into how to regulate hematoma resolution.
Collapse
Affiliation(s)
- Xiongjie Fu
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Neurosurgery, The 2nd Affiliated Hospital, Zhejiang University, Hangzhou 310027, China
| | - Ming Wang
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yingfeng Wan
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Richard F. Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
13
|
Santos-Ribeiro D, Cunha C, Carvalho A. Humoral pathways of innate immune regulation in granuloma formation. Trends Immunol 2024; 45:419-427. [PMID: 38762333 DOI: 10.1016/j.it.2024.04.008] [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: 03/30/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/20/2024]
Abstract
The humoral arm of mammalian innate immunity regulates several molecular mechanisms involved in resistance to pathogens, inflammation, and tissue repair. Recent studies highlight the crucial role played by humoral mediators in granulomatous inflammation. However the molecular mechanisms linking the function of these soluble molecules to the initiation and maintenance of granulomas remain elusive. We propose that humoral innate immunity coordinates fundamental physiological processes in macrophages which, in turn, initiate activation and transformation events that enable granuloma formation. We discuss the involvement of humoral mediators in processes such as immune activation, phagocytosis, metabolism, and tissue remodeling, and how these can dictate macrophage functionality during granuloma formation. These advances present opportunities for discovering novel disease factors and developing targeted, more effective treatments for granulomatous diseases.
Collapse
Affiliation(s)
- Diana Santos-Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
14
|
Verstappen K, Klymov A, Cicuéndez M, da Silva DM, Barroca N, Fernández-San-Argimiro FJ, Madarieta I, Casarrubios L, Feito MJ, Diez-Orejas R, Ferreira R, Leeuwenburgh SC, Portolés MT, Marques PA, Walboomers XF. Biocompatible adipose extracellular matrix and reduced graphene oxide nanocomposite for tissue engineering applications. Mater Today Bio 2024; 26:101059. [PMID: 38693996 PMCID: PMC11061343 DOI: 10.1016/j.mtbio.2024.101059] [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: 01/17/2024] [Revised: 03/30/2024] [Accepted: 04/13/2024] [Indexed: 05/03/2024] Open
Abstract
Despite the immense need for effective treatment of spinal cord injury (SCI), no successful repair strategy has yet been clinically implemented. Multifunctional biomaterials, based on porcine adipose tissue-derived extracellular matrix (adECM) and reduced graphene oxide (rGO), were recently shown to stimulate in vitro neural stem cell growth and differentiation. Nevertheless, their functional performance in clinically more relevant in vivo conditions remains largely unknown. Before clinical application of these adECM-rGO nanocomposites can be considered, a rigorous assessment of the cytotoxicity and biocompatibility of these biomaterials is required. For instance, xenogeneic adECM scaffolds could still harbour potential immunogenicity following decellularization. In addition, the toxicity of rGO has been studied before, yet often in experimental settings that do not bear relevance to regenerative medicine. Therefore, the present study aimed to assess both the in vitro as well as in vivo safety of adECM and adECM-rGO scaffolds. First, pulmonary, renal and hepato-cytotoxicity as well as macrophage polarization studies showed that scaffolds were benign invitro. Then, a laminectomy was performed at the 10th thoracic vertebra, and scaffolds were implanted directly contacting the spinal cord. For a total duration of 6 weeks, animal welfare was not negatively affected. Histological analysis demonstrated the degradation of adECM scaffolds and subsequent tissue remodeling. Graphene-based scaffolds showed a very limited fibrous encapsulation, while rGO sheets were engulfed by foreign body giant cells. Furthermore, all scaffolds were infiltrated by macrophages, which were largely polarized towards a pro-regenerative phenotype. Lastly, organ-specific histopathology and biochemical analysis of blood did not reveal any adverse effects. In summary, both adECM and adECM-rGO implants were biocompatible upon laminectomy while establishing a pro-regenerative microenvironment, which justifies further research on their therapeutic potential for treatment of SCI.
Collapse
Affiliation(s)
- Kest Verstappen
- Department of Dentistry-Regenerative Biomaterials, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 EX, Nijmegen, the Netherlands
| | - Alexey Klymov
- Department of Dentistry-Regenerative Biomaterials, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 EX, Nijmegen, the Netherlands
| | - Mónica Cicuéndez
- Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
| | - Daniela M. da Silva
- Centre for Mechanical Technology and Automation (TEMA), Intelligent Systems Associate Laboratory (LASI), Department of Mechanical Engineering, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Nathalie Barroca
- Centre for Mechanical Technology and Automation (TEMA), Intelligent Systems Associate Laboratory (LASI), Department of Mechanical Engineering, University of Aveiro, 3810-193, Aveiro, Portugal
| | | | - Iratxe Madarieta
- TECNALIA, Basque Research and Technology Alliance (BRTA), E20009, Donostia-San Sebastian, Spain
| | - Laura Casarrubios
- Department of Biochemistry and Molecular Biology, Faculty of Chemistry, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
| | - María José Feito
- Department of Biochemistry and Molecular Biology, Faculty of Chemistry, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
| | - Rosalía Diez-Orejas
- Department of Microbiology and Parasitology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
| | - Rita Ferreira
- Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology (LAQV-REQUIMTE), Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Sander C.G. Leeuwenburgh
- Department of Dentistry-Regenerative Biomaterials, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 EX, Nijmegen, the Netherlands
| | - María Teresa Portolés
- Department of Biochemistry and Molecular Biology, Faculty of Chemistry, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III (ISCIII), 28040, Madrid, Spain
| | - Paula A.A.P. Marques
- Centre for Mechanical Technology and Automation (TEMA), Intelligent Systems Associate Laboratory (LASI), Department of Mechanical Engineering, University of Aveiro, 3810-193, Aveiro, Portugal
| | - X. Frank Walboomers
- Department of Dentistry-Regenerative Biomaterials, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 EX, Nijmegen, the Netherlands
| |
Collapse
|
15
|
Nepon H, Allgayer R, Julien C, Petrecca S, Kalashnikov N, Safran T, Murphy A, Dionisopolous T, Davison P, Cerruti M, Vorstenbosch J. Altered Foreign Body Response at the Posterior Surface Compared to the Anterior Surface of Human Silicone Breast Implants. ACS Biomater Sci Eng 2024; 10:3006-3016. [PMID: 38640484 DOI: 10.1021/acsbiomaterials.3c01961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
Soft implantable devices are crucial to optimizing form and function for many patients. However, periprosthetic capsule fibrosis is one of the major challenges limiting the use of implants. Currently, little is understood about how spatial and temporal factors influence capsule physiology and how the local capsule environment affects the implant structure. In this work, we analyzed breast implant capsule specimens with staining, immunohistochemistry, and real-time polymerase chain reaction to investigate spatiotemporal differences in inflammation and fibrosis. We demonstrated that in comparison to the anterior capsule against the convex surface of breast implants, the posterior capsule against the flat surface of the breast implant displays several features of a dysregulated foreign body reaction including increased capsule thickness, abnormal extracellular remodeling, and infiltration of macrophages. Furthermore, the expression of pro-inflammatory cytokines increased in the posterior capsule across the lifespan of the device, but not in the anterior capsule. We also analyzed the surface oxidation of breast explant samples with XPS analysis. No significant differences in surface oxidation were identified either spatially or temporally. Collectively, our results support spatiotemporal heterogeneity in inflammation and fibrosis within the breast implant capsule. These findings presented here provide a more detailed picture of the complexity of the foreign body reaction surrounding implants destined for human use and could lead to key research avenues and clinical applications to treat periprosthetic fibrosis and improve device longevity.
Collapse
Affiliation(s)
- Hillary Nepon
- Division of Surgical and Interventional Sciences, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
| | - Raphaela Allgayer
- Department of Materials Engineering, McGill University, Wong Building, 3610 Rue University, Montreal, Quebec H3A 0C5, Canada
| | - Cedric Julien
- Research Institute of the McGill University Health Centre, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
| | - Sarah Petrecca
- Faculty of Medicine and Health Sciences, McGill University, 3605 de la Montagne, Montreal, Quebec H3G 1M1, Canada
| | - Nikita Kalashnikov
- Division of Surgical and Interventional Sciences, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
- Faculty of Medicine and Health Sciences, McGill University, 3605 de la Montagne, Montreal, Quebec H3G 1M1, Canada
| | - Tyler Safran
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
| | - Amanda Murphy
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
| | - Tassos Dionisopolous
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
| | - Peter Davison
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
| | - Marta Cerruti
- Department of Materials Engineering, McGill University, Wong Building, 3610 Rue University, Montreal, Quebec H3A 0C5, Canada
| | - Joshua Vorstenbosch
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
- Research Institute of the McGill University Health Centre, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
| |
Collapse
|
16
|
Hong J, Mukherjee B, Sanjoba C, Yamagishi J, Goto Y. Upregulation of ATP6V0D2 benefits intracellular survival of Leishmania donovani in erythrocytes-engulfing macrophages. Front Cell Infect Microbiol 2024; 14:1332381. [PMID: 38357442 PMCID: PMC10864549 DOI: 10.3389/fcimb.2024.1332381] [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: 11/02/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024] Open
Abstract
Visceral leishmaniasis (VL) is the most severe type of leishmaniasis which is caused by infection of Leishmania donovani complex. In the BALB/c mouse model of VL, multinucleated giant cells (MGCs) with heavy parasite infection consist of the largest population of hemophagocytes in the spleen of L. donovani-infected mice, indicating that MGCs provide the parasites a circumstance beneficial for their survival. Although ATP6V0D2 is a demonstrated factor inducing the formation of hemophagocytic MGCs during L. donovani infection, functions of this protein in shaping the infection outcome in macrophages remain unclear. Here we evaluated the influence of upregulated ATP6V0D2 on intracellular survival of the parasites. L. donovani infection-induced hemophagocytosis of normal erythrocytes by macrophages was suppressed by RNAi-based knockdown of Atp6v0d2. The knockdown of Atp6v0d2 did not improve the survival of amastigotes within macrophages when the cells were cultured in the absence of erythrocytes. On the other hand, reduced intracellular survival of amastigotes in macrophages by the knockdown was observed when macrophages were supplemented with antibody-opsonized erythrocytes before infection. There, increase in cytosolic labile iron pool was observed in the L. donovani-infected knocked-down macrophages. It suggests that ATP6V0D2 plays roles not only in upregulation of hemophagocytosis but also in iron trafficking within L. donovani-infected macrophages. Superior access to iron in macrophages may be how the upregulated expression of the molecule brings benefit to Leishmania for their intracellular survival in the presence of erythrocytes.
Collapse
Affiliation(s)
- Jing Hong
- Laboratory of Molecular Immunology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Budhaditya Mukherjee
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Chizu Sanjoba
- Laboratory of Molecular Immunology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Junya Yamagishi
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Yasuyuki Goto
- Laboratory of Molecular Immunology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
17
|
Gorgy A, Barone N, Nepon H, Dalfen J, Efanov JI, Davison P, Vorstenbosch J. Implant-based breast surgery and capsular formation: when, how and why?-a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:385. [PMID: 37970601 PMCID: PMC10632565 DOI: 10.21037/atm-23-131] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 06/28/2023] [Indexed: 11/17/2023]
Abstract
Background and Objective Implant-based breast surgery is a common procedure for both reconstructive and aesthetic purposes. Breast implants, like any foreign object, trigger the formation of a capsule around them. While generally harmless, the capsule can undergo fibrotic changes leading to capsular contracture, which can negatively impact surgical outcomes and patient well-being. Additionally, rare but serious complications, such as breast implant-associated anaplastic large cell lymphoma (BIA-ALCL) and capsule-associated squamous cell carcinoma, have been reported. This paper aims to review the physiology of capsular formation, identify factors contributing to capsule-related pathologies, and discuss their clinical implications. Methods A review of relevant literature was conducted by searching databases for articles published between inception and September 2022. The search included but not limited to terms such as "capsular formation" and "capsular contracture". Selected articles were critically analyzed to address the objectives of this review. Key Content and Findings Capsular formation involves interactions between the implant surface, surrounding tissues, and the immune system. Factors influencing pathological changes in the capsule include genetic predisposition, bacterial contamination, implant characteristics, and surgical techniques. Capsular contracture, characterized by tissue hardening, pain, and implant distortion, remains the most common complication. Rare but life-threatening conditions, such as BIA-ALCL and capsule-associated squamous cell carcinoma, necessitate vigilant monitoring and early detection. Conclusions Understanding the physiology of capsular formation and its associated pathologies is crucial for healthcare providers involved in implant-based breast surgery. Efforts should focus on minimizing the risk of capsular contracture through improved implant materials, surgical techniques, and infection prevention. The emergence of BIA-ALCL and capsule-associated squamous cell carcinoma underscores the importance of long-term surveillance and prompt diagnosis. Further research is needed to uncover underlying mechanisms and develop preventive measures and treatments for these complications. Enhancing our knowledge and clinical management of capsular formation will lead to safer and more successful outcomes in implant-based breast surgery.
Collapse
Affiliation(s)
- Andrew Gorgy
- Department of Plastic and Reconstructive Surgery, McGill University Health Center, Montreal, Quebec, Canada
| | - Natasha Barone
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Hillary Nepon
- Department of Plastic and Reconstructive Surgery, McGill University Health Center, Montreal, Quebec, Canada
| | - Jacquie Dalfen
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Johnny Ionut Efanov
- Plastic and Reconstructive Surgery Service, Department of Surgery, Centre Hospitalier de l’universite de Montreal, Montreal, Quebec, Canada
| | - Peter Davison
- Department of Plastic and Reconstructive Surgery, McGill University Health Center, Montreal, Quebec, Canada
| | - Joshua Vorstenbosch
- Department of Plastic and Reconstructive Surgery, McGill University Health Center, Montreal, Quebec, Canada
| |
Collapse
|
18
|
Isola JVV, Ocañas SR, Hubbart CR, Ko S, Mondal SA, Hense JD, Carter HNC, Schneider A, Kovats S, Alberola-Ila J, Freeman WM, Stout MB. A single-cell atlas of the aging murine ovary. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.29.538828. [PMID: 37162983 PMCID: PMC10168416 DOI: 10.1101/2023.04.29.538828] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Ovarian aging leads to diminished fertility, dysregulated endocrine signaling, and increased chronic disease burden. These effects begin to emerge long before follicular exhaustion. Around 35 years old, women experience a sharp decline in fertility, corresponding to declines in oocyte quality. Despite a growing body of work, the field lacks a comprehensive cellular map of the transcriptomic changes in the aging ovary to identify early drivers of ovarian decline. To fill this gap, we performed single-cell RNA sequencing on ovarian tissue from young (3-month-old) and reproductively aged (9-month-old) mice. Our analysis revealed a doubling of immune cells in the aged ovary, with lymphocyte proportions increasing the most, which was confirmed by flow cytometry. We also found an age-related downregulation of collagenase pathways in stromal fibroblasts, which corresponds to rises in ovarian fibrosis. Follicular cells displayed stress response, immunogenic, and fibrotic signaling pathway inductions with aging. This report raises provides critical insights into mechanisms responsible for ovarian aging phenotypes.
Collapse
Affiliation(s)
- José V. V. Isola
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Sarah R. Ocañas
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Chase R. Hubbart
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Sunghwan Ko
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Samim Ali Mondal
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Jessica D. Hense
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Nutrition College, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Hannah N. C. Carter
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Augusto Schneider
- Nutrition College, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Susan Kovats
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - José Alberola-Ila
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Willard M. Freeman
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Michael B. Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| |
Collapse
|
19
|
Hazra S, Kalyan Dinda S, Kumar Mondal N, Hossain SR, Datta P, Yasmin Mondal A, Malakar P, Manna D. Giant cells: multiple cells unite to survive. Front Cell Infect Microbiol 2023; 13:1220589. [PMID: 37790914 PMCID: PMC10543420 DOI: 10.3389/fcimb.2023.1220589] [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: 05/10/2023] [Accepted: 07/26/2023] [Indexed: 10/05/2023] Open
Abstract
Multinucleated Giant Cells (MGCs) are specialized cells that develop from the fusion of multiple cells, and their presence is commonly observed in human cells during various infections. However, MGC formation is not restricted to infections alone but can also occur through different mechanisms, such as endoreplication and abortive cell cycle. These processes lead to the formation of polyploid cells, eventually resulting in the formation of MGCs. In Entamoeba, a protozoan parasite that causes amoebic dysentery and liver abscesses in humans, the formation of MGCs is a unique phenomenon and not been reported in any other protozoa. This organism is exposed to various hostile environmental conditions, including changes in temperature, pH, and nutrient availability, which can lead to stress and damage to its cells. The formation of MGCs in Entamoeba is thought to be a survival strategy to cope with these adverse conditions. This organism forms MGCs through cell aggregation and fusion in response to osmotic and heat stress. The MGCs in Entamoeba are thought to have increased resistance to various stresses and can survive longer than normal cells under adverse conditions. This increased survival could be due to the presence of multiple nuclei, which could provide redundancy in case of DNA damage or mutations. Additionally, MGCs may play a role in the virulence of Entamoeba as they are found in the inflammatory foci of amoebic liver abscesses and other infections caused by Entamoeba. The presence of MGCs in these infections suggests that they may contribute to the pathogenesis of the disease. Overall, this article offers valuable insights into the intriguing phenomenon of MGC formation in Entamoeba. By unraveling the mechanisms behind this process and examining its implications, researchers can gain a deeper understanding of the complex biology of Entamoeba and potentially identify new targets for therapeutic interventions. The study of MGCs in Entamoeba serves as a gateway to exploring the broader field of cell fusion in various organisms, providing a foundation for future investigations into related cellular processes and their significance in health and disease.
Collapse
Affiliation(s)
- Shreyasee Hazra
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational and Research Institute (RKMVERI), Kolkata, India
| | - Suman Kalyan Dinda
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational and Research Institute (RKMVERI), Kolkata, India
| | - Naba Kumar Mondal
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational and Research Institute (RKMVERI), Kolkata, India
| | - Sk Rajjack Hossain
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational and Research Institute (RKMVERI), Kolkata, India
| | - Pratyay Datta
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational and Research Institute (RKMVERI), Kolkata, India
| | - Afsana Yasmin Mondal
- Institute of Health Sciences, Presidency University, Kolkata, West Bengal, India
| | - Pushkar Malakar
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational and Research Institute (RKMVERI), Kolkata, India
| | - Dipak Manna
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational and Research Institute (RKMVERI), Kolkata, India
| |
Collapse
|
20
|
Jackson JW, Foster JS, Martin EB, Macy S, Wooliver C, Balachandran M, Richey T, Heidel RE, Williams AD, Kennel SJ, Wall JS. Collagen inhibits phagocytosis of amyloid in vitro and in vivo and may act as a 'don't eat me' signal. Amyloid 2023; 30:249-260. [PMID: 36541892 DOI: 10.1080/13506129.2022.2155133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/04/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Systemic amyloidosis refers to a group of protein misfolding disorders characterized by the extracellular deposition of amyloid fibrils in organs and tissues. For reasons heretofore unknown, amyloid deposits are not recognized by the immune system, and progressive deposition leads to organ dysfunction. METHODS In vitro and in vivo phagocytosis assays were performed to elucidate the impact of collagen and other amyloid associated proteins (eg serum amyloid p component and apolipoprotein E) had on amyloid phagocytosis. Immunohistochemical and histopathological staining regimens were employed to analyze collagen-amyloid interactions and immune responses. RESULTS Histological analysis of amyloid-laden tissue indicated that collagen is intimately associated with amyloid deposits. We report that collagen inhibits phagocytosis of amyloid fibrils by macrophages. Treatment of 15 patient-derived amyloid extracts with collagenase significantly enhanced amyloid phagocytosis. Preclinical mouse studies indicated that collagenase treatment of amyloid extracts significantly enhanced clearance as compared to controls, coincident with increased immune cell infiltration of the subcutaneous amyloid lesion. CONCLUSIONS These data suggest that amyloid-associated collagen serves as a 'don't eat me' signal, thereby hindering clearance of amyloid. Targeted degradation of amyloid-associated collagen could result in innate immune cell recognition and clearance of pathologic amyloid deposits.
Collapse
Affiliation(s)
- Joseph W Jackson
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - James S Foster
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Emily B Martin
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Sallie Macy
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Craig Wooliver
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Manasi Balachandran
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Tina Richey
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - R Eric Heidel
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Angela D Williams
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Stephen J Kennel
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Jonathan S Wall
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| |
Collapse
|
21
|
Mofarrah M, Jafari-Gharabaghlou D, Farhoudi-Sefidan-Jadid M, Zarghami N. Potential application of inorganic nano-materials in modulation of macrophage function: Possible application in bone tissue engineering. Heliyon 2023; 9:e16309. [PMID: 37292328 PMCID: PMC10245018 DOI: 10.1016/j.heliyon.2023.e16309] [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: 12/30/2022] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 06/10/2023] Open
Abstract
Nanomaterials indicate unique physicochemical properties for drug delivery in osteogenesis. Benefiting from high surface area grades, high volume ratio, ease of functionalization by biological targeting moieties, and small size empower nanomaterials to pass through biological barriers for efficient targeting. Inorganic nanomaterials for bone regeneration include inorganic synthetic polymers, ceramic nanoparticles, metallic nanoparticles, and magnetic nanoparticles. These nanoparticles can effectively modulate macrophage polarization and function, as one of the leading players in osteogenesis. Bone healing procedures in close cooperation with the immune system. Inflammation is one of the leading triggers of the bone fracture healing barrier. Macrophages commence anti-inflammatory signaling along with revascularization in the damaged site to promote the formation of a soft callus, bone mineralization, and bone remodeling. In this review, we will discuss the role of macrophages in bone hemostasis and regeneration. Furthermore, we will summarize the influence of the various inorganic nanoparticles on macrophage polarization and function in the benefit of osteogenesis.
Collapse
Affiliation(s)
- Mohsen Mofarrah
- Department of Medical Biotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davoud Jafari-Gharabaghlou
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Farhoudi-Sefidan-Jadid
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Aydin University, Istanbul, Turkey
| |
Collapse
|
22
|
Hiéronimus L, Huaux F. B-1 cells in immunotoxicology: Mechanisms underlying their response to chemicals and particles. FRONTIERS IN TOXICOLOGY 2023; 5:960861. [PMID: 37143777 PMCID: PMC10151831 DOI: 10.3389/ftox.2023.960861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
Since their discovery nearly 40 years ago, B-1 cells have continued to challenge the boundaries between innate and adaptive immunity, as well as myeloid and lymphoid functions. This B-cell subset ensures early immunity in neonates before the development of conventional B (B-2) cells and respond to immune injuries throughout life. B-1 cells are multifaceted and serve as natural- and induced-antibody-producing cells, phagocytic cells, antigen-presenting cells, and anti-/pro-inflammatory cytokine-releasing cells. This review retraces the origin of B-1 cells and their different roles in homeostatic and infectious conditions before focusing on pollutants comprising contact-sensitivity-inducing chemicals, endocrine disruptors, aryl hydrocarbon receptor (AHR) ligands, and reactive particles.
Collapse
|
23
|
Ahmadzadeh K, Pereira M, Vanoppen M, Bernaerts E, Ko J, Mitera T, Maksoudian C, Manshian BB, Soenen S, Rose CD, Matthys P, Wouters C, Behmoaras J. Multinucleation resets human macrophages for specialized functions at the expense of their identity. EMBO Rep 2023; 24:e56310. [PMID: 36597777 PMCID: PMC9986822 DOI: 10.15252/embr.202256310] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 01/05/2023] Open
Abstract
Macrophages undergo plasma membrane fusion and cell multinucleation to form multinucleated giant cells (MGCs) such as osteoclasts in bone, Langhans giant cells (LGCs) as part of granulomas or foreign-body giant cells (FBGCs) in reaction to exogenous material. How multinucleation per se contributes to functional specialization of mature mononuclear macrophages remains poorly understood in humans. Here, we integrate comparative transcriptomics with functional assays in purified mature mononuclear and multinucleated human osteoclasts, LGCs and FBGCs. Strikingly, in all three types of MGCs, multinucleation causes a pronounced downregulation of macrophage identity. We show enhanced lysosome-mediated intracellular iron homeostasis promoting MGC formation. The transition from mononuclear to multinuclear state is accompanied by cell specialization specific to each polykaryon. Enhanced phagocytic and mitochondrial function associate with FBGCs and osteoclasts, respectively. Moreover, human LGCs preferentially express B7-H3 (CD276) and can form granuloma-like clusters in vitro, suggesting that their multinucleation potentiates T cell activation. These findings demonstrate how cell-cell fusion and multinucleation reset human macrophage identity as part of an advanced maturation step that confers MGC-specific functionality.
Collapse
Affiliation(s)
- Kourosh Ahmadzadeh
- Laboratory of Immunobiology, Department Microbiology, Immunology and Transplantation, Rega InstituteKU Leuven—University of LeuvenLeuvenBelgium
| | - Marie Pereira
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Hammersmith HospitalImperial College LondonLondonUK
| | - Margot Vanoppen
- Laboratory of Immunobiology, Department Microbiology, Immunology and Transplantation, Rega InstituteKU Leuven—University of LeuvenLeuvenBelgium
| | - Eline Bernaerts
- Laboratory of Immunobiology, Department Microbiology, Immunology and Transplantation, Rega InstituteKU Leuven—University of LeuvenLeuvenBelgium
| | - Jeong‐Hun Ko
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Hammersmith HospitalImperial College LondonLondonUK
| | - Tania Mitera
- Laboratory of Immunobiology, Department Microbiology, Immunology and Transplantation, Rega InstituteKU Leuven—University of LeuvenLeuvenBelgium
| | - Christy Maksoudian
- NanoHealth and Optical Imaging Group, Translational Cell and Tissue Research Unit, Department of Imaging and PathologyKU LeuvenLeuvenBelgium
| | - Bella B Manshian
- Translational Cell and Tissue Research Unit, Department of Imaging and PathologyKU LeuvenLeuvenBelgium
| | - Stefaan Soenen
- NanoHealth and Optical Imaging Group, Translational Cell and Tissue Research Unit, Department of Imaging and PathologyKU LeuvenLeuvenBelgium
| | - Carlos D Rose
- Division of Pediatric Rheumatology Nemours Children's HospitalThomas Jefferson UniversityPhiladelphiaPAUSA
| | - Patrick Matthys
- Laboratory of Immunobiology, Department Microbiology, Immunology and Transplantation, Rega InstituteKU Leuven—University of LeuvenLeuvenBelgium
| | - Carine Wouters
- Laboratory of Immunobiology, Department Microbiology, Immunology and Transplantation, Rega InstituteKU Leuven—University of LeuvenLeuvenBelgium
- Division Pediatric RheumatologyUZ LeuvenLeuvenBelgium
- European Reference Network for Rare ImmunodeficiencyAutoinflammatory and Autoimmune Diseases (RITA) at University Hospital LeuvenLeuvenBelgium
| | - Jacques Behmoaras
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Hammersmith HospitalImperial College LondonLondonUK
- Programme in Cardiovascular and Metabolic Disorders and Centre for Computational BiologyDuke‐NUS Medical School SingaporeSingaporeSingapore
| |
Collapse
|
24
|
Mao M, Liang N, Ren R, Zhao Y, Ma D, Liu H. A Nomogram Model for Predicting the Postoperative Recurrence of Localized Laryngeal Amyloidosis. Ann Otol Rhinol Laryngol 2023; 132:259-265. [PMID: 35403442 DOI: 10.1177/00034894221086990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To analyze the factors related to postoperative recurrence in patients with localized laryngeal amyloidosis (LocLA) and to construct a nomogram prediction model (NPM). METHODS We collected the data for LocLA patients diagnosed from March 2000 to May 2019 and clinical characteristics data were extracted. Factors related to recurrence were analyzed using multivariate logistic regression. The NPM was constructed for predicting the recurrence risk of LocLA. The receiver operating characteristic (ROC) curve evaluated the distinguishing ability using the area under curve (AUC). The calibration curve was created to evaluate the consistency of the NPM. RESULTS A total of 226 confirmed LocLA cases were included. One hundred seventy-five cases (77.4%) had localized single nodule, and 51 cases had more than one lesions. Sixty-three (27.9%) cases had no multinucleated giant cell (MGC) around amyloid, and 163 (72.1%) cases had MGC around amyloid. Multivariate logistic regression analysis showed that more than one lesions (odds ratio [OR] = 3.206 and 95% confidence interval [CI]: 1.492-6.888; P value: .003), subglottic involvement (OR = 2.926 and 95% CI: 1.300-6.585; P = .010), and no multinucleated giant cell (MGC) around amyloid (OR = 2.503 and 95% CI: 1.173-5.342; P = .018) had a statistically significant effect on postoperative LocLA recurrence (P < .05). The AUC of the ROC curve was 0.753 (95% CI: 0.667-0.832). The bias-corrected curve approached the ideal curve, with an average absolute error of 0.037. CONCLUSIONS More than one lesions, subglottic involvement, and no MGC around amyloid are risk factors for postoperative recurrence of LocLA. The NPM constructed has good applicability.
Collapse
Affiliation(s)
- Meiling Mao
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University; Beijing Key Laboratory of Head and Neck Molecular Diagnostic Pathology, Beijing, China
| | - Na Liang
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University; Beijing Key Laboratory of Head and Neck Molecular Diagnostic Pathology, Beijing, China
| | - Ran Ren
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University; Beijing Key Laboratory of Head and Neck Molecular Diagnostic Pathology, Beijing, China
| | - Yihua Zhao
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University; Beijing Key Laboratory of Head and Neck Molecular Diagnostic Pathology, Beijing, China
| | - Donglin Ma
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University; Beijing Key Laboratory of Head and Neck Molecular Diagnostic Pathology, Beijing, China
| | - Honggang Liu
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University; Beijing Key Laboratory of Head and Neck Molecular Diagnostic Pathology, Beijing, China
| |
Collapse
|
25
|
The Contribution of the Sheep and the Goat Model to the Study of Ovarian Ageing. BIOLOGY 2023; 12:biology12020270. [PMID: 36829547 PMCID: PMC9953374 DOI: 10.3390/biology12020270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/20/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
Ovarian ageing stands as the major contributor towards fertility loss. As such, there is an urge for studies addressing the mechanisms that promote ovarian ageing and new strategies aiming to delay it. Recently, the presence of a unique population of multinucleated giant cells has been identified in the ovaries of reproductively aged mice. These cells have been considered hallmarks of ovarian ageing. However, up to date multinucleated giant cells have only been described in the ovaries of the mice. Therefore, the aim of the present work was to evaluate and characterize the presence of such hallmarks of ovarian ageing in the sheep and the goat. In this study, ovaries from juvenile (6 months) and mature animals (18-24 months) were used. The hematoxylin and eosin technique was performed to describe the ovarian morphology and evaluate the ovarian follicle reserve pool. Sudan black B staining and the detection of autofluorescence emission were used to identify and characterize the presence of multinucleated giant cells. Statistical analyses were performed with GraphPad Prism 9.0.0. A decrease in the follicle reserve pool and the presence of multinucleated giant cells, with lipofuscin accumulation and the emission of autofluorescence, were observed in the ovaries of the mature animals of both species. Our results support the interest in the use of the ovine and the caprine model, that share physiological and pathophysiological characteristics with humans, in future studies addressing ovarian ageing.
Collapse
|
26
|
Schloesser D, Lindenthal L, Sauer J, Chung KJ, Chavakis T, Griesser E, Baskaran P, Maier-Habelsberger U, Fundel-Clemens K, Schlotthauer I, Watson CK, Swee LK, Igney F, Park JE, Huber-Lang MS, Thomas MJ, El Kasmi KC, Murray PJ. Senescent cells suppress macrophage-mediated corpse removal via upregulation of the CD47-QPCT/L axis. J Cell Biol 2023; 222:213731. [PMID: 36459066 PMCID: PMC9723804 DOI: 10.1083/jcb.202207097] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/19/2022] [Accepted: 11/04/2022] [Indexed: 12/03/2022] Open
Abstract
Progressive accrual of senescent cells in aging and chronic diseases is associated with detrimental effects in tissue homeostasis. We found that senescent fibroblasts and epithelia were not only refractory to macrophage-mediated engulfment and removal, but they also paralyzed the ability of macrophages to remove bystander apoptotic corpses. Senescent cell-mediated efferocytosis suppression (SCES) was independent of the senescence-associated secretory phenotype (SASP) but instead required direct contact between macrophages and senescent cells. SCES involved augmented senescent cell expression of CD47 coinciding with increased CD47-modifying enzymes QPCT/L. SCES was reversible by interfering with the SIRPα-CD47-SHP-1 axis or QPCT/L activity. While CD47 expression increased in human and mouse senescent cells in vitro and in vivo, another ITIM-containing protein, CD24, contributed to SCES specifically in human epithelial senescent cells where it compensated for genetic deficiency in CD47. Thus, CD47 and CD24 link the pathogenic effects of senescent cells to homeostatic macrophage functions, such as efferocytosis, which we hypothesize must occur efficiently to maintain tissue homeostasis.
Collapse
Affiliation(s)
| | | | - Julia Sauer
- Boehringer Ingelheim, Biberach an der Riß, Germany
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory of Medicine, Faculty of Medicine at University Hospital, Technische Universität Dresden, Dresden, Germany
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory of Medicine, Faculty of Medicine at University Hospital, Technische Universität Dresden, Dresden, Germany
| | - Eva Griesser
- Boehringer Ingelheim, Biberach an der Riß, Germany
| | | | | | | | | | | | - Lee Kim Swee
- Boehringer Ingelheim, Biberach an der Riß, Germany
| | | | | | - Markus S Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital Ulm, Ulm, Germany
| | | | | | - Peter J Murray
- Max Planck Institute of Biochemistry, Martinsried, Germany
| |
Collapse
|
27
|
Wang CJ, Noble PB, Elliot JG, James AL, Wang KCW. From Beneath the Skin to the Airway Wall: Understanding the Pathological Role of Adipose Tissue in Comorbid Asthma-Obesity. Compr Physiol 2023; 13:4321-4353. [PMID: 36715283 DOI: 10.1002/cphy.c220011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This article provides a contemporary report on the role of adipose tissue in respiratory dysfunction. Adipose tissue is distributed throughout the body, accumulating beneath the skin (subcutaneous), around organs (visceral), and importantly in the context of respiratory disease, has recently been shown to accumulate within the airway wall: "airway-associated adipose tissue." Excessive adipose tissue deposition compromises respiratory function and increases the severity of diseases such as asthma. The mechanisms of respiratory impairment are inflammatory, structural, and mechanical in nature, vary depending on the anatomical site of deposition and adipose tissue subtype, and likely contribute to different phenotypes of comorbid asthma-obesity. An understanding of adipose tissue-driven pathophysiology provides an opportunity for diagnostic advancement and patient-specific treatment. As an exemplar, the potential impact of airway-associated adipose tissue is highlighted, and how this may change the management of a patient with asthma who is also obese. © 2023 American Physiological Society. Compr Physiol 13:4321-4353, 2023.
Collapse
Affiliation(s)
- Carolyn J Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Peter B Noble
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - John G Elliot
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Alan L James
- Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,Medical School, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Kimberley C W Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Telethon Kids Institute, The University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
28
|
Mamilos A, Winter L, Schmitt VH, Barsch F, Grevenstein D, Wagner W, Babel M, Keller K, Schmitt C, Gürtler F, Schreml S, Niedermair T, Rupp M, Alt V, Brochhausen C. Macrophages: From Simple Phagocyte to an Integrative Regulatory Cell for Inflammation and Tissue Regeneration-A Review of the Literature. Cells 2023; 12:276. [PMID: 36672212 PMCID: PMC9856654 DOI: 10.3390/cells12020276] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/29/2022] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
The understanding of macrophages and their pathophysiological role has dramatically changed within the last decades. Macrophages represent a very interesting cell type with regard to biomaterial-based tissue engineering and regeneration. In this context, macrophages play a crucial role in the biocompatibility and degradation of implanted biomaterials. Furthermore, a better understanding of the functionality of macrophages opens perspectives for potential guidance and modulation to turn inflammation into regeneration. Such knowledge may help to improve not only the biocompatibility of scaffold materials but also the integration, maturation, and preservation of scaffold-cell constructs or induce regeneration. Nowadays, macrophages are classified into two subpopulations, the classically activated macrophages (M1 macrophages) with pro-inflammatory properties and the alternatively activated macrophages (M2 macrophages) with anti-inflammatory properties. The present narrative review gives an overview of the different functions of macrophages and summarizes the recent state of knowledge regarding different types of macrophages and their functions, with special emphasis on tissue engineering and tissue regeneration.
Collapse
Affiliation(s)
- Andreas Mamilos
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
| | - Lina Winter
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
| | - Volker H. Schmitt
- Department of Cardiology, University Medical Centre, Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, 55131 Mainz, Germany
| | - Friedrich Barsch
- Medical Center, Faculty of Medicine, Institute for Exercise and Occupational Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - David Grevenstein
- Clinic and Polyclinic for Orthopedics and Trauma Surgery, University Hospital of Cologne, 50937 Cologne, Germany
| | - Willi Wagner
- Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Translational Lung Research Centre Heidelberg (TLRC), German Lung Research Centre (DZL), 69120 Heidelberg, Germany
| | - Maximilian Babel
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
| | - Karsten Keller
- Department of Cardiology, University Medical Centre, Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Department of Sports Medicine, Medical Clinic VII, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Christine Schmitt
- Department of Internal Medicine, St. Vincenz and Elisabeth Hospital of Mainz (KKM), 55131 Mainz, Germany
| | - Florian Gürtler
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
| | - Stephan Schreml
- Department of Dermatology, University Medical Centre Regensburg, 93053 Regensburg, Germany
| | - Tanja Niedermair
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
| | - Markus Rupp
- Department for Trauma Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Volker Alt
- Department for Trauma Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Christoph Brochhausen
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
- Institute of Pathology, University Medical Centre Mannheim, Ruprecht-Karls-University Heidelberg, 68167 Mannheim, Germany
| |
Collapse
|
29
|
Kang MS, Lee GH, Kwon IH, Yang MJ, Heo MB, Choi JW, Lee TG, Yoon CH, Baek B, Sung MC, Kim DW, Park EJ. Uptake and toxicity of cerium dioxide nanoparticles with different aspect ratio. Toxicol Lett 2022; 373:196-209. [PMID: 36464203 DOI: 10.1016/j.toxlet.2022.11.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/18/2022] [Accepted: 11/21/2022] [Indexed: 12/05/2022]
Abstract
Cerium dioxide nanoparticles (CeONPs) have been extensively applied in research for future energy development due to two common oxidation states on their surface. Considering that shape (aspect ratio) is a key determinant of NPs-induced toxicity, we compared the toxicity of hexagonal (H)- and rod-shaped (R)-CeONPs in mice. At 24 h after pharyngeal aspiration, both types of CeONPs recruited surrounding immune cells (monocytes and neutrophils) into the lung, and R-CeONPs induced a more severe pulmonary inflammatory response compared with H-CeONPs. To identify an indicator to predict pulmonary inflammatory responses at the cellular level, we also investigated their responses in alveolar macrophage cells. At 24 h after treatment, both types of CeONPs were mainly located within the vacuoles (partially, in the lysosome) in the cytoplasm. Mitochondrial damage, intracellular calcium accumulation, and increased NO production were observed in cells exposed to both types of CeONPs, ultimately resulting in a decrease in cell viability. More interestingly, both types of CeONPs formed multinucleated giant cells. Meanwhile, contrary to when suspended in deionized water, R-CeONPs were strongly aggregated with a negative charge in cell culture media, whereas H-CeONPs were relatively well-dispersed with a positive charge. R-CeONPs-induced lysosomal extension was also recovered by premix with negatively charged DNA, and even NPs suspended in cell culture media without cells were detected under the FACS system, suggesting interference by protein corona. Therefore, we suggest that shape (aspect ratio) is an important factor determining inhaled NPs-induced pathology and that the effect of the surface charge and protein corona should be carefully considered in interpreting results derived from in vitro tests. Furthermore, we propose that the relationship between the formation of multinucleated giant cells and the inflammatory response of inhaled CeONPs should be further studied.
Collapse
Affiliation(s)
- Min-Sung Kang
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, 02447, Republic of Korea; Jeonbuk Branch Institute, Korea Institute of Toxicology, 56212, Republic of Korea
| | - Gwang-Hee Lee
- School of Civil, Environmental and Architectural Engineering, Korea University, 02841, Republic of Korea
| | - Ik Hwan Kwon
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 34113, Republic of Korea
| | - Mi-Jin Yang
- Jeonbuk Branch Institute, Korea Institute of Toxicology, 56212, Republic of Korea
| | - Min Beom Heo
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 34113, Republic of Korea
| | - Jae-Won Choi
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, 02447, Republic of Korea; Safety Measurement Institute, Korea Research Institute of Standards and Science, 34113, Republic of Korea
| | - Tae Geol Lee
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 34113, Republic of Korea
| | - Cheol-Ho Yoon
- Environmental Analysis Team, Korea Basic Science Institute, Seoul 28119, Republic of Korea
| | - Bosung Baek
- Toxicity Evaluation Center, Keyprime Research Company, 28161, Republic of Korea; Department of Biochemistry and Molecular Biology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Myeong-Chang Sung
- School of Civil, Environmental and Architectural Engineering, Korea University, 02841, Republic of Korea
| | - Dong-Wan Kim
- School of Civil, Environmental and Architectural Engineering, Korea University, 02841, Republic of Korea.
| | - Eun-Jung Park
- Department of Biochemistry and Molecular Biology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Human Health and Environmental Toxins Research Center, Kyung Hee University, 02447, Republic of Korea.
| |
Collapse
|
30
|
Different Molecular Features of Epithelioid and Giant Cells in Foreign Body Reaction Identified by Single-Cell RNA Sequencing. J Invest Dermatol 2022; 142:3232-3242.e16. [PMID: 35853485 DOI: 10.1016/j.jid.2022.06.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/18/2022] [Accepted: 06/21/2022] [Indexed: 01/05/2023]
Abstract
Although macrophage‒epithelioid cell (EPC)‒giant cell (GC) differentiation is acknowledged in foreign body reaction (FBR), the exact molecular features remain elusive. To discover the molecular profiles of EPC and GC, we analyzed mouse sponge and silk FBRs by integrating single-cell RNA sequencing and spatial sequencing, which identified seven cell types, including macrophages and fibroblasts. Macrophages comprised three subsets with a trajectory from M2-like cell to EPC to GC. They were different in many aspects, including cytokine, extracellular matrix organization/degradation, epithelial modules, and glycolysis that were consistent in both sponge and silk FBRs. EPCs exhibited epithelial modules and extracellular matrix organization, and GCs showed glycolysis, extracellular matrix degradation, and cell fusion signatures. Cellular interactions in GCs and M2-like cells were predicted to be higher than that in EPCs. High expression of inflammation or fusion-related (GPNMB, matrix metalloproteinase 12 gene MMP12, DCSTAMP) and glycolysis-related (PGAM1, ALDOA) genes was identified in GCs of human/mouse tissues, suggesting them as GC-specific markers. Our study identified unique signatures of EPCs and GCs in FBR. Importantly, GCs showed strong glycolysis signatures and cellular interactions, suggesting their activation in FBR. Our data on EPC and GC refinement and GC-specific markers enable the understanding of FBR and help to explore preventive and therapeutic management strategies for skin FBRs.
Collapse
|
31
|
Monocyte-Macrophage Lineage Cell Fusion. Int J Mol Sci 2022; 23:ijms23126553. [PMID: 35742997 PMCID: PMC9223484 DOI: 10.3390/ijms23126553] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 02/06/2023] Open
Abstract
Cell fusion (fusogenesis) occurs in natural and pathological conditions in prokaryotes and eukaryotes. Cells of monocyte–macrophage lineage are highly fusogenic. They create syncytial multinucleated giant cells (MGCs) such as osteoclasts (OCs), MGCs associated with the areas of infection/inflammation, and foreign body-induced giant cells (FBGCs). The fusion of monocytes/macrophages with tumor cells may promote cancer metastasis. We describe types and examples of monocyte–macrophage lineage cell fusion and the role of actin-based structures in cell fusion.
Collapse
|
32
|
Ahmad F, Rani A, Alam A, Zarin S, Pandey S, Singh H, Hasnain SE, Ehtesham NZ. Macrophage: A Cell With Many Faces and Functions in Tuberculosis. Front Immunol 2022; 13:747799. [PMID: 35603185 PMCID: PMC9122124 DOI: 10.3389/fimmu.2022.747799] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 03/30/2022] [Indexed: 01/16/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) is the causative agent of human tuberculosis (TB) which primarily infects the macrophages. Nearly a quarter of the world's population is infected latently by Mtb. Only around 5%-10% of those infected develop active TB disease, particularly during suppressed host immune conditions or comorbidity such as HIV, hinting toward the heterogeneity of Mtb infection. The aerosolized Mtb first reaches the lungs, and the resident alveolar macrophages (AMs) are among the first cells to encounter the Mtb infection. Evidence suggests that early clearance of Mtb infection is associated with robust innate immune responses in resident macrophages. In addition to lung-resident macrophage subsets, the recruited monocytes and monocyte-derived macrophages (MDMs) have been suggested to have a protective role during Mtb infection. Mtb, by virtue of its unique cell surface lipids and secreted protein effectors, can evade killing by the innate immune cells and preferentially establish a niche within the AMs. Continuous efforts to delineate the determinants of host defense mechanisms have brought to the center stage the crucial role of macrophage phenotypical variations for functional adaptations in TB. The morphological and functional heterogeneity and plasticity of the macrophages aid in confining the dissemination of Mtb. However, during a suppressed or hyperactivated immune state, the Mtb virulence factors can affect macrophage homeostasis which may skew to favor pathogen growth, causing active TB. This mini-review is aimed at summarizing the interplay of Mtb pathomechanisms in the macrophages and the implications of macrophage heterogeneity and plasticity during Mtb infection.
Collapse
Affiliation(s)
- Faraz Ahmad
- Laboratory of Infection Biology and Cell Signaling, Indian Council of Medical Research (ICMR)-National Institute of Pathology, New Delhi, India
| | - Anshu Rani
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi (IIT-D), New Delhi, India
| | - Anwar Alam
- Laboratory of Infection Biology and Cell Signaling, Indian Council of Medical Research (ICMR)-National Institute of Pathology, New Delhi, India
| | - Sheeba Zarin
- Laboratory of Infection Biology and Cell Signaling, Indian Council of Medical Research (ICMR)-National Institute of Pathology, New Delhi, India
| | - Saurabh Pandey
- Department of Biochemistry, Jamia Hamdard, New Delhi, India
| | - Hina Singh
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi (IIT-D), New Delhi, India
| | - Seyed Ehtesham Hasnain
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi (IIT-D), New Delhi, India
- Department of Life Science, School of Basic Sciences and Research, Sharda University, Greater Noida, India
| | - Nasreen Zafar Ehtesham
- Laboratory of Infection Biology and Cell Signaling, Indian Council of Medical Research (ICMR)-National Institute of Pathology, New Delhi, India
| |
Collapse
|
33
|
Ahmadzadeh K, Vanoppen M, Rose CD, Matthys P, Wouters CH. Multinucleated Giant Cells: Current Insights in Phenotype, Biological Activities, and Mechanism of Formation. Front Cell Dev Biol 2022; 10:873226. [PMID: 35478968 PMCID: PMC9035892 DOI: 10.3389/fcell.2022.873226] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/17/2022] [Indexed: 12/21/2022] Open
Abstract
Monocytes and macrophages are innate immune cells with diverse functions ranging from phagocytosis of microorganisms to forming a bridge with the adaptive immune system. A lesser-known attribute of macrophages is their ability to fuse with each other to form multinucleated giant cells. Based on their morphology and functional characteristics, there are in general three types of multinucleated giant cells including osteoclasts, foreign body giant cells and Langhans giant cells. Osteoclasts are bone resorbing cells and under physiological conditions they participate in bone remodeling. However, under pathological conditions such as rheumatoid arthritis and osteoporosis, osteoclasts are responsible for bone destruction and bone loss. Foreign body giant cells and Langhans giant cells appear only under pathological conditions. While foreign body giant cells are found in immune reactions against foreign material, including implants, Langhans giant cells are associated with granulomas in infectious and non-infectious diseases. The functionality and fusion mechanism of osteoclasts are being elucidated, however, our knowledge on the functions of foreign body giant cells and Langhans giant cells is limited. In this review, we describe and compare the phenotypic aspects, biological and functional activities of the three types of multinucleated giant cells. Furthermore, we provide an overview of the multinucleation process and highlight key molecules in the different phases of macrophage fusion.
Collapse
Affiliation(s)
- Kourosh Ahmadzadeh
- Laboratory of Immunobiology, Department Microbiology and Immunology, Rega Institute, KU Leuven – University of Leuven, Leuven, Belgium
- *Correspondence: Kourosh Ahmadzadeh, ; Carine Helena Wouters,
| | - Margot Vanoppen
- Laboratory of Immunobiology, Department Microbiology and Immunology, Rega Institute, KU Leuven – University of Leuven, Leuven, Belgium
| | - Carlos D. Rose
- Division of Pediatric Rheumatology Nemours Children’s Hospital, Thomas Jefferson University, Philadelphia, PA, United States
| | - Patrick Matthys
- Laboratory of Immunobiology, Department Microbiology and Immunology, Rega Institute, KU Leuven – University of Leuven, Leuven, Belgium
| | - Carine Helena Wouters
- Laboratory of Immunobiology, Department Microbiology and Immunology, Rega Institute, KU Leuven – University of Leuven, Leuven, Belgium
- Division Pediatric Rheumatology, UZ Leuven, Leuven, Belgium
- European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) at University Hospital Leuven, Leuven, Belgium
- *Correspondence: Kourosh Ahmadzadeh, ; Carine Helena Wouters,
| |
Collapse
|
34
|
Kakanj P, Bhide S, Moussian B, Leptin M. Autophagy-mediated plasma membrane removal promotes the formation of epithelial syncytia. EMBO J 2022; 41:e109992. [PMID: 35262206 PMCID: PMC9194749 DOI: 10.15252/embj.2021109992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 01/18/2023] Open
Abstract
Epithelial wound healing in Drosophila involves the formation of multinucleate cells surrounding the wound. We show that autophagy, a cellular degradation process often deployed in stress responses, is required for the formation of a multinucleated syncytium during wound healing, and that autophagosomes that appear near the wound edge acquire plasma membrane markers. In addition, uncontrolled autophagy in the unwounded epidermis leads to the degradation of endo‐membranes and the lateral plasma membrane, while apical and basal membranes and epithelial barrier function remain intact. Proper functioning of TORC1 is needed to prevent destruction of the larval epidermis by autophagy, in a process that depends on phagophore initiation and expansion but does not require autophagosomes fusion with lysosomes. Autophagy induction can also affect other sub‐cellular membranes, as shown by its suppression of experimentally induced laminopathy‐like nuclear defects. Our findings reveal a function for TORC1‐mediated regulation of autophagy in maintaining membrane integrity and homeostasis in the epidermis and during wound healing.
Collapse
Affiliation(s)
- Parisa Kakanj
- Institute for Genetics, University of Cologne, Cologne, Germany.,Director's Research Unit, European Molecular Biology Laboratory, Heidelberg, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Sourabh Bhide
- Director's Research Unit, European Molecular Biology Laboratory, Heidelberg, Germany.,Faculty of Biosciences, Collaboration for Joint PhD degree between EMBL and Heidelberg University, Heidelberg, Germany
| | | | - Maria Leptin
- Institute for Genetics, University of Cologne, Cologne, Germany.,Director's Research Unit, European Molecular Biology Laboratory, Heidelberg, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| |
Collapse
|
35
|
Thermal treatment at 500°C significantly reduces the reaction to irregular tricalcium phosphate granules as foreign bodies: An in vivo study. Acta Biomater 2022. [DOI: 10.1016/j.actbio.2022.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
36
|
Mouse innate-like B-1 lymphocytes promote inhaled particle-induced in vitro granuloma formation and inflammation in conjunction with macrophages. Arch Toxicol 2021; 96:585-599. [PMID: 34935064 PMCID: PMC8837577 DOI: 10.1007/s00204-021-03200-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/09/2021] [Indexed: 11/20/2022]
Abstract
The current paradigm for explaining lung granulomatous diseases induced by inhaled particles is mainly based on macrophages. This mechanism is now challenging because B lymphocytes also infiltrate injured tissue, and the deficiency in B lymphocytes is associated with limited lung granulomas in silica-treated mice. Here, we investigated how B lymphocytes respond to micro- and nanoparticles by combining in vivo and in vitro mouse models. We first demonstrated that innate-like B-1 lymphocytes (not conventional B-2 lymphocytes or plasma cells) specifically accumulated during granuloma formation in mice instilled with crystalline silica (DQ12, 2.5 mg/mouse) and carbon nanotubes (CNT Mitsui, 0.2 mg/mouse). In comparison to macrophages, peritoneal B-1 lymphocytes purified from naïve mice were resistant to the pyroptotic activity of reactive particles (up to 1 mg/mL) but clustered to establish in vitro cell/particle aggregates. Mouse B-1 lymphocytes (not B-2 lymphocytes) in coculture with macrophages and CNT (0.1 µg/mL) organized three-dimensional spheroid structures in Matrigel and stimulated the release of TIMP-1. Furthermore, purified B-1 lymphocytes are sensitive to nanosilica toxicity through radical generation in culture. Nanosilica-exposed B-1 lymphocytes released proinflammatory cytokines and alarmins. In conclusion, our data indicate that in addition to macrophages, B-1 lymphocytes participate in micrometric particle-induced granuloma formation and display inflammatory functions in response to nanoparticles.
Collapse
|
37
|
Abstract
Cell membrane fusion and multinucleation in macrophages are associated with physiologic homeostasis as well as disease. Osteoclasts are multinucleated macrophages that resorb bone through increased metabolic activity resulting from cell fusion. Fusion of macrophages also generates multinucleated giant cells (MGCs) in white adipose tissue (WAT) of obese individuals. For years, our knowledge of MGCs in WAT has been limited to their description as part of crown-like structures (CLS) surrounding damaged adipocytes. However, recent evidence indicates that these cells can phagocytose oversized lipid remnants, suggesting that, as in osteoclasts, cell fusion and multinucleation are required for specialized catabolic functions. We thus reason that WAT MGCs can be viewed as functionally analogous to osteoclasts and refer to them in this article as adipoclasts. We first review current knowledge on adipoclasts and their described functions. In view of recent advances in single cell genomics, we describe WAT macrophages from a ‘fusion perspective’ and speculate on the ontogeny of adipoclasts. Specifically, we highlight the role of CD9 and TREM2, two plasma membrane markers of lipid-associated macrophages in WAT, which have been previously described as regulators of fusion and multinucleation in osteoclasts and MGCs. Finally, we consider whether strategies aiming to target WAT macrophages can be more selectively directed against adipoclasts.
Collapse
|
38
|
Ma J, Liu Q, White JR. Novel methods to determine complement activation in human serum induced by the complex of Dezamizumab and serum amyloid P. J Biol Chem 2021; 297:101136. [PMID: 34461096 PMCID: PMC8463879 DOI: 10.1016/j.jbc.2021.101136] [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: 03/08/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 12/02/2022] Open
Abstract
Lack of simple and robust methods to determine complement activation in human serum induced by antigen–antibody complexes is a major hurdle for monitoring therapeutic antibody drug quality and stability. Dezamizumab is a humanized IgG1 monoclonal antibody that binds to serum amyloid P component (SAP) for potential treatment of systemic amyloidosis. The mechanism of action of Dezamizumab includes the binding of SAP, complement activation through classical pathway, and phagocytosis; however, the steps in this process cannot be easily monitored. We developed two novel methods to determine Dezamizumab-SAP complex-induced complement activation. Complement component 3 (C3) depletion was detected by homogeneous time-resolved fluorescence (HTRF), and C3a desArg fragment, formed after the cleavage of C3 to yield C3a followed by removal of its C-terminal arginine residue, was determined using Meso Scale Discovery (MSD) technology. We found that the presence of both Dezamizumab and SAP was required for complement activation via both methods. The optimal molar ratio of Dezamizumab:SAP was 6:1 in order to obtain maximal complement activation. The relative potency from both methods showed a good correlation to Dezamizumab-SAP-dependent complement component 1q (C1q) binding activity in Dezamizumab thermal-stressed samples. Both SAP and C1q binding, as determined by surface plasmon resonance and the two complement activation potency methods described here, reflect the mechanism of action of Dezamizumab. We conclude that these methods can be used to monitor Dezamizumab quality for drug release and stability testing, and the novel potency methods reported here can be potentially used to evaluate complement activity induced by other antigen–antibody complexes.
Collapse
Affiliation(s)
- Jianhong Ma
- GlaxoSmithKline, Structure Function Characterization, CMCA, Collegeville, Pennsylvania, USA.
| | - Qi Liu
- GlaxoSmithKline, Structure Function Characterization, CMCA, Collegeville, Pennsylvania, USA
| | - John R White
- GlaxoSmithKline, Structure Function Characterization, CMCA, Collegeville, Pennsylvania, USA
| |
Collapse
|
39
|
Qiao W, Xie H, Fang J, Shen J, Li W, Shen D, Wu J, Wu S, Liu X, Zheng Y, Cheung KMC, Yeung KWK. Sequential activation of heterogeneous macrophage phenotypes is essential for biomaterials-induced bone regeneration. Biomaterials 2021; 276:121038. [PMID: 34339925 DOI: 10.1016/j.biomaterials.2021.121038] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 06/23/2021] [Accepted: 07/21/2021] [Indexed: 02/06/2023]
Abstract
Macrophage has been gradually recognized as a central regulator in tissue regeneration, and the study of how macrophage mediates biomaterials-induced bone regeneration through immunomodulatory pathway becomes popular. However, the current understanding on the roles of different macrophage phenotypes in regulating bone tissue regeneration remains controversial. In this study, we demonstrate that sequential infiltration of heterogeneous phenotypes of macrophages triggered by bio-metal ions effectively facilitates bone healing in bone defect. Indeed, M1 macrophages promote the recruitment and early commitment of osteogenic and angiogenic progenitors, while M2 macrophages and osteoclasts support the deposition and mineralization of the bone matrix, as well as the maturation of blood vessels. Moreover, we have identified a group of bone biomaterial-related multinucleated cells that behave similarly to M2 macrophages with wound-healing features rather than participate in the bone resorption cascade similarly to osteoclasts. Our study shows how sequential activation of macrophage-osteoclast lineage contribute to a highly orchestrated immune response in the bone tissue microenvironment around biomaterials to regulate the complex biological process of bone healing. Therefore, we believe that the temporal activation pattern of heterogeneous macrophage phenotypes should be considered when the next generation of biomaterials for bone regeneration is engineered.
Collapse
Affiliation(s)
- Wei Qiao
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, 999077, China; Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China
| | - Huizhi Xie
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, 999077, China; Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China
| | - Jinghan Fang
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, 999077, China; Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China
| | - Jie Shen
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, 999077, China; Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China
| | - Wenting Li
- State Key Laboratory for Turbulence and Complex System and Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing, 100871, PR China
| | - Danni Shen
- State Key Laboratory for Turbulence and Complex System and Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing, 100871, PR China
| | - Jun Wu
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China
| | - Shuilin Wu
- School of Materials Science and Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, PR China; Cixi Center of Biomaterials Surface Engineering, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Ningbo, PR China
| | - Yufeng Zheng
- State Key Laboratory for Turbulence and Complex System and Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing, 100871, PR China
| | - Kenneth M C Cheung
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, 999077, China; Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China
| | - Kelvin W K Yeung
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, 999077, China; Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China; Cixi Center of Biomaterials Surface Engineering, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Ningbo, PR China.
| |
Collapse
|
40
|
Dufrançais O, Mascarau R, Poincloux R, Maridonneau-Parini I, Raynaud-Messina B, Vérollet C. Cellular and molecular actors of myeloid cell fusion: podosomes and tunneling nanotubes call the tune. Cell Mol Life Sci 2021; 78:6087-6104. [PMID: 34296319 PMCID: PMC8429379 DOI: 10.1007/s00018-021-03875-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/25/2021] [Accepted: 06/05/2021] [Indexed: 12/22/2022]
Abstract
Different types of multinucleated giant cells (MGCs) of myeloid origin have been described; osteoclasts are the most extensively studied because of their importance in bone homeostasis. MGCs are formed by cell-to-cell fusion, and most types have been observed in pathological conditions, especially in infectious and non-infectious chronic inflammatory contexts. The precise role of the different MGCs and the mechanisms that govern their formation remain poorly understood, likely due to their heterogeneity. First, we will introduce the main populations of MGCs derived from the monocyte/macrophage lineage. We will then discuss the known molecular actors mediating the early stages of fusion, focusing on cell-surface receptors involved in the cell-to-cell adhesion steps that ultimately lead to multinucleation. Given that cell-to-cell fusion is a complex and well-coordinated process, we will also describe what is currently known about the evolution of F-actin-based structures involved in macrophage fusion, i.e., podosomes, zipper-like structures, and tunneling nanotubes (TNT). Finally, the localization and potential role of the key fusion mediators related to the formation of these F-actin structures will be discussed. This review intends to present the current status of knowledge of the molecular and cellular mechanisms supporting multinucleation of myeloid cells, highlighting the gaps still existing, and contributing to the proposition of potential disease-specific MGC markers and/or therapeutic targets.
Collapse
Affiliation(s)
- Ophélie Dufrançais
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Rémi Mascarau
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina
| | - Renaud Poincloux
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Isabelle Maridonneau-Parini
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France
| | - Brigitte Raynaud-Messina
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France.
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina.
| | - Christel Vérollet
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France.
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina.
| |
Collapse
|
41
|
Multinucleated Giant Cells Induced by a Silk Fibroin Construct Express Proinflammatory Agents: An Immunohistological Study. MATERIALS 2021; 14:ma14144038. [PMID: 34300957 PMCID: PMC8307820 DOI: 10.3390/ma14144038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022]
Abstract
Multinucleated giant cells (MNGCs) are frequently observed in the implantation areas of different biomaterials. The main aim of the present study was to analyze the long-term polarization pattern of the pro- and anti-inflammatory phenotypes of macrophages and MNGCs for 180 days to better understand their role in the success or failure of biomaterials. For this purpose, silk fibroin (SF) was implanted in a subcutaneous implantation model of Wistar rats as a model for biomaterial-induced MNGCs. A sham operation was used as a control for physiological wound healing. The expression of different inflammatory markers (proinflammatory M1: CCR-7, iNos; anti-inflammatory M2: CD-206, CD-163) and tartrate-resistant acid phosphatase (TRAP) and CD-68 were identified using immunohistochemical staining. The results showed significantly higher numbers of macrophages and MNGCs within the implantation bed of SF-expressed M1 markers, compared to M2 markers. Interestingly, the expression of proinflammatory markers was sustained over the long observation period of 180 days. By contrast, the control group showed a peak of M1 macrophages only on day 3. Thereafter, the inflammatory pattern shifted to M2 macrophages. No MNGCs were observed in the control group. To the best of our knowledge, this is study is the first to outline the persistence of pro-inflammatory MNGCs within the implantation bed of SF and to describe their long-term kinetics over 180 days. Clinically, these results are highly relevant to understand the role of biomaterial-induced MNGCs in the long term. These findings suggest that tailored physicochemical properties may be a key to avoiding extensive inflammatory reactions and achieving clinical success. Therefore, further research is needed to elucidate the correlation between proinflammatory MNGCs and the physicochemical characteristics of the implanted biomaterial.
Collapse
|
42
|
Implant Fibrosis and the Underappreciated Role of Myofibroblasts in the Foreign Body Reaction. Cells 2021; 10:cells10071794. [PMID: 34359963 PMCID: PMC8304203 DOI: 10.3390/cells10071794] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
Body implants and implantable medical devices have dramatically improved and prolonged the life of countless patients. However, our body repair mechanisms have evolved to isolate, reject, or destroy any object that is recognized as foreign to the organism and inevitably mounts a foreign body reaction (FBR). Depending on its severity and chronicity, the FBR can impair implant performance or create severe clinical complications that will require surgical removal and/or replacement of the faulty device. The number of review articles discussing the FBR seems to be proportional to the number of different implant materials and clinical applications and one wonders, what else is there to tell? We will here take the position of a fibrosis researcher (which, coincidentally, we are) to elaborate similarities and differences between the FBR, normal wound healing, and chronic healing conditions that result in the development of peri-implant fibrosis. After giving credit to macrophages in the inflammatory phase of the FBR, we will mainly focus on the activation of fibroblastic cells into matrix-producing and highly contractile myofibroblasts. While fibrosis has been discussed to be a consequence of the disturbed and chronic inflammatory milieu in the FBR, direct activation of myofibroblasts at the implant surface is less commonly considered. Thus, we will provide a perspective how physical properties of the implant surface control myofibroblast actions and accumulation of stiff scar tissue. Because formation of scar tissue at the surface and around implant materials is a major reason for device failure and extraction surgeries, providing implant surfaces with myofibroblast-suppressing features is a first step to enhance implant acceptance and functional lifetime. Alternative therapeutic targets are elements of the myofibroblast mechanotransduction and contractile machinery and we will end with a brief overview on such targets that are considered for the treatment of other organ fibroses.
Collapse
|
43
|
Ghate A, Sharma S, Agrawal P, Sahu A. Differential expression of complement receptors CR1/2 and CR4 by murine M1 and M2 macrophages. Mol Immunol 2021; 137:75-83. [PMID: 34229135 DOI: 10.1016/j.molimm.2021.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 05/20/2021] [Accepted: 06/06/2021] [Indexed: 11/26/2022]
Abstract
Macrophages polarize into functionally divergent phenotypes - M1 and M2 - which express distinct receptors. These cells are known to express complement receptors, including CR1, CR3, and CR4. However, whether these complement receptors are differentially expressed on M1 and M2 macrophages is not yet known. Herein, we have examined the expression of CR1 to CR4 on murine bone marrow-derived M1 (stimulated with IFN-γ or LPS) and M2 (stimulated with IL-4 or IL-4 + IL-13) macrophages. We show that M1 cells exhibit increased expression of CR1/2, whereas M2 cells display the higher expression of CR4; CR3 is equally expressed on both the phenotypes. Thus, M1 cells are CR1/2+CR4+, and M2 are CR1/2-CR4+. Functional probing of these cells for their phagocytic ability indicates that M1 cells, which express higher CR1/2, internalize a significantly greater number of C3b-opsonized erythrocytes. Both M1 and M2 cells, on the other hand, internalize iC3b-opsonized erythrocytes to a similar extent. Interestingly, the phagocytic receptor involved in phagocytosis of iC3b-opsonized erythrocytes is only CR3 with no contribution of CR4. We, thus, propose that complement receptor expression can be used in combination with the expression of other known polarization markers to better locate a macrophage along its phenotypic spectrum.
Collapse
Affiliation(s)
- Arya Ghate
- Complement Biology Laboratory, National Centre for Cell Science, S. P. Pune University Campus, Pune - 411007, India
| | - Samriddhi Sharma
- Complement Biology Laboratory, National Centre for Cell Science, S. P. Pune University Campus, Pune - 411007, India
| | - Palak Agrawal
- Complement Biology Laboratory, National Centre for Cell Science, S. P. Pune University Campus, Pune - 411007, India
| | - Arvind Sahu
- Complement Biology Laboratory, National Centre for Cell Science, S. P. Pune University Campus, Pune - 411007, India.
| |
Collapse
|
44
|
Lösslein AK, Lohrmann F, Scheuermann L, Gharun K, Neuber J, Kolter J, Forde AJ, Kleimeyer C, Poh YY, Mack M, Triantafyllopoulou A, Dunlap MD, Khader SA, Seidl M, Hölscher A, Hölscher C, Guan XL, Dorhoi A, Henneke P. Monocyte progenitors give rise to multinucleated giant cells. Nat Commun 2021; 12:2027. [PMID: 33795674 PMCID: PMC8016882 DOI: 10.1038/s41467-021-22103-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 02/23/2021] [Indexed: 01/12/2023] Open
Abstract
The immune response to mycobacteria is characterized by granuloma formation, which features multinucleated giant cells as a unique macrophage type. We previously found that multinucleated giant cells result from Toll-like receptor-induced DNA damage and cell autonomous cell cycle modifications. However, the giant cell progenitor identity remained unclear. Here, we show that the giant cell-forming potential is a particular trait of monocyte progenitors. Common monocyte progenitors potently produce cytokines in response to mycobacteria and their immune-active molecules. In addition, common monocyte progenitors accumulate cholesterol and lipids, which are prerequisites for giant cell transformation. Inducible monocyte progenitors are so far undescribed circulating common monocyte progenitor descendants with high giant cell-forming potential. Monocyte progenitors are induced in mycobacterial infections and localize to granulomas. Accordingly, they exhibit important immunological functions in mycobacterial infections. Moreover, their signature trait of high cholesterol metabolism may be piggy-backed by mycobacteria to create a permissive niche.
Collapse
Affiliation(s)
- Anne Kathrin Lösslein
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
- MOTI-VATE Graduate School, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Florens Lohrmann
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Pediatrics and Adolescent Medicine, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Spemann Graduate School for Biology and Medicine (SGBM) and IMM-PACT Clinician Scientist Program, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Kourosh Gharun
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Jana Neuber
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Julia Kolter
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Aaron James Forde
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Christian Kleimeyer
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ying Yee Poh
- Nanyang Technological University, Lee Kong Chian School of Medicine, Singapore, Singapore
| | - Matthias Mack
- University Hospital Regensburg, Internal Medicine II, Nephrology, Regensburg, Germany
| | - Antigoni Triantafyllopoulou
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Rheumatism Research Centre Berlin, Leibniz Association, Berlin, Germany
| | - Micah D Dunlap
- Department of Molecular Microbiology, Washington University in St. Louis School of Medicine, Saint Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, Saint Louis, MO, USA
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University in St. Louis School of Medicine, Saint Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, Saint Louis, MO, USA
| | - Maximilian Seidl
- Center for Chronic Immunodeficiency and Institute for Clinical Pathology, Department of Pathology, Medical Center and Faculty of Medicine, Freiburg, Germany and Institute of Pathology, Heinrich Heine University and University Hospital of Duesseldorf, Duesseldorf, Germany
| | | | - Christoph Hölscher
- Forschungszentrum Borstel, Leibniz Lungenzentrum, Borstel, Germany
- Deutsches Zentrum für Infektionsforschung, Standort Borstel, Borstel, Germany
| | - Xue Li Guan
- Nanyang Technological University, Lee Kong Chian School of Medicine, Singapore, Singapore
| | - Anca Dorhoi
- Max Planck Institute for Infection Biology, Berlin, Germany
- Institute of Immunology, Federal Research Institute for Animal Health, Friedrich-Loeffler-Institut (FLI), Insel Riems, Germany
- Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany
| | - Philipp Henneke
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Center for Pediatrics and Adolescent Medicine, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
45
|
Weavers H, Martin P. The cell biology of inflammation: From common traits to remarkable immunological adaptations. J Cell Biol 2021; 219:151857. [PMID: 32539109 PMCID: PMC7337495 DOI: 10.1083/jcb.202004003] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023] Open
Abstract
Tissue damage triggers a rapid and robust inflammatory response in order to clear and repair a wound. Remarkably, many of the cell biology features that underlie the ability of leukocytes to home in to sites of injury and to fight infection—most of which are topics of intensive current research—were originally observed in various weird and wonderful translucent organisms over a century ago by Elie Metchnikoff, the “father of innate immunity,” who is credited with discovering phagocytes in 1882. In this review, we use Metchnikoff’s seminal lectures as a starting point to discuss the tremendous variety of cell biology features that underpin the function of these multitasking immune cells. Some of these are shared by other cell types (including aspects of motility, membrane trafficking, cell division, and death), but others are more unique features of innate immune cells, enabling them to fulfill their specialized functions, such as encapsulation of invading pathogens, cell–cell fusion in response to foreign bodies, and their self-sacrifice as occurs during NETosis.
Collapse
Affiliation(s)
- Helen Weavers
- School of Biochemistry, Biomedical Sciences, University of Bristol, Bristol UK
| | - Paul Martin
- School of Biochemistry, Biomedical Sciences, University of Bristol, Bristol UK.,School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, UK.,School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
46
|
Queval CJ, Fearns A, Botella L, Smyth A, Schnettger L, Mitermite M, Wooff E, Villarreal-Ramos B, Garcia-Jimenez W, Heunis T, Trost M, Werling D, Salguero FJ, Gordon SV, Gutierrez MG. Macrophage-specific responses to human- and animal-adapted tubercle bacilli reveal pathogen and host factors driving multinucleated cell formation. PLoS Pathog 2021; 17:e1009410. [PMID: 33720986 PMCID: PMC7993774 DOI: 10.1371/journal.ppat.1009410] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/25/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023] Open
Abstract
The Mycobacterium tuberculosis complex (MTBC) is a group of related pathogens that cause tuberculosis (TB) in mammals. MTBC species are distinguished by their ability to sustain in distinct host populations. While Mycobacterium bovis (Mbv) sustains transmission cycles in cattle and wild animals and causes zoonotic TB, M. tuberculosis (Mtb) affects human populations and seldom causes disease in cattle. The host and pathogen determinants underlying host tropism between MTBC species are still unknown. Macrophages are the main host cell that encounters mycobacteria upon initial infection, and we hypothesised that early interactions between the macrophage and mycobacteria influence species-specific disease outcome. To identify factors that contribute to host tropism, we analysed blood-derived primary human and bovine macrophages (hMϕ or bMϕ, respectively) infected with Mbv and Mtb. We show that Mbv and Mtb reside in different cellular compartments and differentially replicate in hMϕ whereas both Mbv and Mtb efficiently replicate in bMϕ. Specifically, we show that out of the four infection combinations, only the infection of bMϕ with Mbv promoted the formation of multinucleated giant cells (MNGCs), a hallmark of tuberculous granulomas. Mechanistically, we demonstrate that both MPB70 from Mbv and extracellular vesicles released by Mbv-infected bMϕ promote macrophage multinucleation. Importantly, we extended our in vitro studies to show that granulomas from Mbv-infected but not Mtb-infected cattle contained higher numbers of MNGCs. Our findings implicate MNGC formation in the contrasting pathology between Mtb and Mbv for the bovine host and identify MPB70 from Mbv and extracellular vesicles from bMϕ as mediators of this process. The identification of host and pathogen factors contributing to host-pathogen interaction is crucial to understand the pathogenesis and dissemination of tuberculosis. This is particularly the case in deciphering the mechanistic basis for host-tropism across the MTBC. Here, we show that in vitro, M. bovis but not M. tuberculosis induces multinucleated cell formation in bovine macrophages. We identified host and pathogen mechanistic drivers of multinucleated cell formation: MPB70 as the M. bovis factor and bovine macrophage extracellular vesicles. Using a cattle experimental infection model, we confirmed differential multinucleated cell formation in vivo. Thus, we have identified host and pathogen factors that contribute to host tropism in human/bovine tuberculosis. Additionally, this work provides an explanation for the long-standing association of multinucleated cells with tuberculosis pathogenesis.
Collapse
Affiliation(s)
| | | | | | - Alicia Smyth
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Dublin, Ireland
| | | | - Morgane Mitermite
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Esen Wooff
- Animal and Plant Health Agency, Addlestone, United Kingdom
| | - Bernardo Villarreal-Ramos
- Animal and Plant Health Agency, Addlestone, United Kingdom
- Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Aberystwyth, United Kingdom
| | - Waldo Garcia-Jimenez
- Department of Pathology an Infectious Diseases. School of Veterinary Medicine. University of Surrey, Guildford, United Kingdom
| | - Tiaan Heunis
- Biosciences Institute, Newcastle University, Newcastle, United Kingdom
| | - Matthias Trost
- Biosciences Institute, Newcastle University, Newcastle, United Kingdom
| | - Dirk Werling
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, University of London, Hatfield Hertfordshire, United Kingdom
| | - Francisco J. Salguero
- Department of Pathology an Infectious Diseases. School of Veterinary Medicine. University of Surrey, Guildford, United Kingdom
- National Infection Service, Public Health England (PHE), Porton Down, Salisbury, United Kingdom
| | - Stephen V. Gordon
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Dublin, Ireland
| | | |
Collapse
|
47
|
Amengual-Peñafiel L, Córdova LA, Constanza Jara-Sepúlveda M, Brañes-Aroca M, Marchesani-Carrasco F, Cartes-Velásquez R. Osteoimmunology drives dental implant osseointegration: A new paradigm for implant dentistry. JAPANESE DENTAL SCIENCE REVIEW 2021; 57:12-19. [PMID: 33737990 PMCID: PMC7946347 DOI: 10.1016/j.jdsr.2021.01.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/30/2020] [Accepted: 01/14/2021] [Indexed: 01/08/2023] Open
Abstract
There is a complex interaction between titanium dental implants, bone, and the immune system. Among them, specific immune cells, macrophages play a crucial role in the osseointegration dynamics. Infiltrating macrophages and resident macrophages (osteomacs) contribute to achieving an early pro-regenerative peri-implant environment. Also, multinucleated giant cells (MNGCs) in the bone-implant interface and their polarization ability, maintain a peri-implant immunological balance to preserve osseointegration integrity. However, dental implants can display cumulative levels of antigens (ions, nano and microparticles and bacterial antigens) at the implant–tissue interface activating an immune-inflammatory response. If the inflammation is not resolved or reactivated due to the stress signals and the immunogenicity of elements present, this could lead implants to aseptic loosening, infections, and subsequent bone loss. Therefore, to maintain osseointegration and prevent bone loss of implants, a better understanding of the osteoimmunology of the peri-implant environment would lead to the development of new therapeutic approaches. In this line, depicting osteoimmunological mechanisms, we discuss immunomodulatory strategies to improve and preserve a long-term functional integration between dental implants and the human body. Scientific field of dental science: implant dentistry.
Collapse
Affiliation(s)
| | - Luis A Córdova
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Chile, Chile.,Department of Oral and Maxillofacial Surgery, Clínica Las Condes, Santiago, Chile.,Department of Oral and Maxillofacial Surgery, Complejo Hospitalario San José. Craneofacial Translational Research Laboratory, Faculty of Dentistry, University of Chile, Santiago, Chile
| | | | | | | | | |
Collapse
|
48
|
Thermal treatment at 500 °C significantly reduces the reaction to irregular tricalcium phosphate granules as foreign bodies: An in vivo study. Acta Biomater 2021; 121:621-636. [PMID: 33249227 DOI: 10.1016/j.actbio.2020.11.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 10/28/2020] [Accepted: 11/19/2020] [Indexed: 12/17/2022]
Abstract
Evaporation of phosphate species during thermal treatment (> 400 °C) of calcium phosphates leads to the formation of an alkaline layer on their surface. The aim of this study was to evaluate the hypothesis that the biological response of thermally treated calcium phosphates is modified by the presence of such an alkaline layer on their surface. For this purpose, 0.125-0.180 mm α- and β-tricalcium phosphate (TCP) granules were obtained by crushing and size classification, with some being subjected to thermal treatment at 500 °C. The four types of granules (α-TCP, β-TCP, α-TCP-500 °C, and β-TCP-500 °C) were implanted subcutaneously and orthotopically in rats. Sham operations served as control. Subcutaneously, α-TCP and β-TCP induced significantly more multinucleated giant cells (MNGCs) than calcined granules. Most of the induced MNGCs were TRAP-negative, CD-68 positive and cathepsin K-negative, reflecting a typical indication of a reaction with a foreign body. The vessel density was significantly higher in the α-TCP and β-TCP groups than it was in the α-TCP-500 °C and β-TCP-500 °C groups. In the femur model, β-TCP-500 °C induced significantly more new bone formation than that induced by β-TCP. The granule size was also significantly larger in the β-TCP-500 °C group, making it more resistant to degradation than β-TCP. The MNGC density was higher in the α-TCP and β-TCP groups than in the α-TCP-500 °C and β-TCP-500 °C groups, including cathepsin-positive, CD-68 positive, TRAP-positive and TRAP-negative MNGCs. In conclusion, this study confirms that the biological response of calcium phosphates was affected by the presence of an alkaline layer on their surface. Thermally-treated α-TCP and β-TCP granules produced significantly fewer MNGCs and were significantly less degraded than non-thermally-treated α-TCP and β-TCP granules. Thermally treating α-TCP and β-TCP granules shifts the reaction from a foreign body reaction towards a physiological reaction by downregulating the number of induced MNGCs and enhancing degradation resistance.
Collapse
|
49
|
Braune J, Lindhorst A, Fröba J, Hobusch C, Kovacs P, Blüher M, Eilers J, Bechmann I, Gericke M. Multinucleated Giant Cells in Adipose Tissue Are Specialized in Adipocyte Degradation. Diabetes 2021; 70:538-548. [PMID: 33158932 DOI: 10.2337/db20-0293] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 10/30/2020] [Indexed: 11/13/2022]
Abstract
Obesity is associated with chronic low-grade inflammation of visceral adipose tissue (AT) characterized by an increasing number of AT macrophages (ATMs) and linked to type 2 diabetes. AT inflammation is histologically indicated by the formation of so-called crown-like structures, as ATMs accumulate around dying adipocytes, and the occurrence of multinucleated giant cells (MGCs). However, to date, the function of MGCs in obesity is unknown. Therefore, the aim of this study was to characterize MGCs in AT and unravel the function of these cells. We demonstrated that MGCs occurred in obese patients and after 24 weeks of a high-fat diet in mice, accompanying signs of AT inflammation and then representing ∼3% of ATMs in mice. Mechanistically, we found evidence that adipocyte death triggered MGC formation. Most importantly, MGCs in obese AT had a higher capacity to phagocytize oversized particles, such as adipocytes, as shown by live imaging of AT, 45-µm bead uptake ex vivo, and higher lipid content in vivo. Finally, we showed that interleukin-4 treatment was sufficient to increase the number of MGCs in AT, whereas other factors may be more important for endogenous MGC formation in vivo. Most importantly, our data suggest that MGCs are specialized for clearance of dead adipocytes in obesity.
Collapse
Affiliation(s)
- Julia Braune
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Andreas Lindhorst
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Janine Fröba
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | | | - Peter Kovacs
- Medical Department III, Leipzig University, Leipzig, Germany
| | - Matthias Blüher
- Medical Department III, Leipzig University, Leipzig, Germany
| | - Jens Eilers
- Carl-Ludwig Institute of Physiology, Leipzig University, Leipzig, Germany
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Martin Gericke
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
50
|
Foley KG, Pritchard MT, Duncan FE. Macrophage-derived multinucleated giant cells: hallmarks of the aging ovary. Reproduction 2021; 161:V5-V9. [PMID: 33258461 DOI: 10.1530/rep-20-0489] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/20/2020] [Indexed: 12/31/2022]
Abstract
Inflammaging is a state of chronic, low-grade inflammation associated with aging which contributes to age-related diseases. Recently, an age-associated increase in inflammation has been documented in the mammalian ovary, which is accompanied by a shift in the immune cell profile. In this Point of View article, we consider a unique population of macrophage-derived multinucleated giant cells, found in reproductively old mouse ovaries, as potential markers or functional drivers of inflammation in ovarian aging.
Collapse
Affiliation(s)
- K Grace Foley
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Michele T Pritchard
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|