1
|
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
|
2
|
Raftery AL, O'Brien CA, Shad A, L'Estrange-Stranieri E, Hsu AT, Jacobsen EA, Harris NL, Tsantikos E, Hibbs ML. Activated eosinophils in early life impair lung development and promote long-term lung damage. Mucosal Immunol 2024; 17:871-891. [PMID: 38901764 DOI: 10.1016/j.mucimm.2024.06.003] [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: 12/04/2023] [Revised: 05/21/2024] [Accepted: 06/05/2024] [Indexed: 06/22/2024]
Abstract
Exaggeration of type 2 immune responses promotes lung inflammation and altered lung development; however, eosinophils, despite expansion in the postnatal lung, have not been specifically assessed in the context of neonatal lung disease. Furthermore, early life factors including prematurity and respiratory infection predispose infants to chronic obstructive pulmonary disease later in life. To assess eosinophils in the developing lung and how they may contribute to chronic lung disease, we generated mice harboring eosinophil-specific deletion of the negative regulatory enzyme SH2 domain-containing inositol 5' phosphatase-1. This increased the activity and number of pulmonary eosinophils in the developing lung, which was associated with impaired lung development, expansion of activated alveolar macrophages (AMφ), multinucleated giant cell formation, enlargement of airspaces, and fibrosis. Despite regression of eosinophils following completion of lung development, AMφ-dominated inflammation persisted, alongside lung damage. Bone marrow chimera studies showed that SH2 domain-containing inositol 5' phosphatase-1-deficient eosinophils were not sufficient to drive inflammatory lung disease in adult steady-state mice but once inflammation and damage were present, it could not be resolved. Depletion of eosinophils during alveolarization alleviated pulmonary inflammation and lung pathology, demonstrating an eosinophil-intrinsic effect. These results show that the presence of activated eosinophils during alveolarization aggravates AMφs and promotes sustained inflammation and long-lasting lung pathology.
Collapse
Affiliation(s)
- April L Raftery
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Caitlin A O'Brien
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Ali Shad
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Elan L'Estrange-Stranieri
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Amy T Hsu
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Elizabeth A Jacobsen
- Division of Allergy, Asthma and Clinical Immunology, Department of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Nicola L Harris
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Evelyn Tsantikos
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Margaret L Hibbs
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
3
|
Mechanisms of Foreign Body Giant Cell Formation in Response to Implantable Biomaterials. Polymers (Basel) 2023; 15:polym15051313. [PMID: 36904554 PMCID: PMC10007405 DOI: 10.3390/polym15051313] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
Long term function of implantable biomaterials are determined by their integration with the host's body. Immune reactions against these implants could impair the function and integration of the implants. Some biomaterial-based implants lead to macrophage fusion and the formation of multinucleated giant cells, also known as foreign body giant cells (FBGCs). FBGCs may compromise the biomaterial performance and may lead to implant rejection and adverse events in some cases. Despite their critical role in response to implants, there is a limited understanding of cellular and molecular mechanisms involved in forming FBGCs. Here, we focused on better understanding the steps and mechanisms triggering macrophage fusion and FBGCs formation, specifically in response to biomaterials. These steps included macrophage adhesion to the biomaterial surface, fusion competency, mechanosensing and mechanotransduction-mediated migration, and the final fusion. We also described some of the key biomarkers and biomolecules involved in these steps. Understanding these steps on a molecular level would lead to enhance biomaterials design and improve their function in the context of cell transplantation, tissue engineering, and drug delivery.
Collapse
|
4
|
Villarreal-Leal RA, Healey GD, Corradetti B. Biomimetic immunomodulation strategies for effective tissue repair and restoration. Adv Drug Deliv Rev 2021; 179:113913. [PMID: 34371087 DOI: 10.1016/j.addr.2021.113913] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/21/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022]
Abstract
Inflammation plays a central role in wound healing following injury or disease and is mediated by a precise cascade of cellular and molecular events. Unresolved inflammatory processes lead to chronic inflammation and fibrosis, which can result in prolonged wound healing lasting months or years that hampers tissue function. Therapeutic interventions mediated by immunomodulatory drugs, cells, or biomaterials, are therefore most effective during the inflammatory phase of wound healing when a pro-regenerative environment is essential. In this review, we discuss the advantages of exploiting knowledge of the native tissue microenvironment to develop therapeutics capable of modulating the immune response and promoting functional tissue repair. In particular, we provide examples of the most recent biomimetic platforms proposed to accomplish this goal, with an emphasis on those able to induce macrophage polarization towards a pro-regenerative phenotype.
Collapse
|
5
|
Fibrin polymer on the surface of biomaterial implants drives the foreign body reaction. Biomaterials 2021; 277:121087. [PMID: 34478933 DOI: 10.1016/j.biomaterials.2021.121087] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/09/2021] [Accepted: 08/23/2021] [Indexed: 12/18/2022]
Abstract
Implantation of biomaterials and medical devices in the body triggers the foreign body reaction (FBR) which is characterized by macrophage fusion at the implant surface leading to the formation of foreign body giant cells and the development of the fibrous capsule enveloping the implant. While adhesion of macrophages to the surface is an essential step in macrophage fusion and implanted biomaterials are known to rapidly acquire a layer of host proteins, a biological substrate that is responsible for this process in vivo is unknown. Here we show that mice with genetically imposed fibrinogen deficiency display a dramatic reduction of macrophage fusion on biomaterials implanted intraperitoneally and subcutaneously and are protected from the formation of the fibrin-containing fibrous capsule. Furthermore, macrophage fusion on biomaterials implanted in FibAEK mice that express a mutated form of fibrinogen incapable of thrombin-mediated polymerization was strongly reduced. Despite the lack of fibrin, the capsule was formed in FibAEK mice, although it had a different composition and distinct mechanical properties than that in wild-type mice. Specifically, while mononuclear α-SMA-expressing macrophages embedded in the capsule of both strains of mice secreted collagen, the amount of collagen and its density in the tissue of FibAEK mice was reduced. These data identify fibrin polymer as a key biological substrate driving the development of the FBR.
Collapse
|
6
|
Balansky R, La Maestra S, Kancheva VD, Trofimov AV, Djongov L, De Flora S. Clastogenic effects of cigarette smoke and urethane and their modulation by olive oil, curcumin and carotenoids in adult mice and foetuses. Food Chem Toxicol 2021; 155:112383. [PMID: 34224802 DOI: 10.1016/j.fct.2021.112383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/16/2021] [Accepted: 07/01/2021] [Indexed: 10/20/2022]
Abstract
In spite of the overwhelming epidemiological evidence for cigarette smoke (CS) carcinogenicity, less attention has been paid to the effects of CS as a complex mixture. As assessed in a series of experiments in murine models, the whole-body exposure to mainstream CS induced significant increases of micronucleated cells in the respiratory tract, bone marrow and peripheral blood of adult mice as well as in the liver and peripheral blood of foetuses whose mothers had been exposed throughout pregnancy. Urethane was potently clastogenic in the same cells when injected intraperitoneally. The daily administration of extra-virgin olive oil by gavage produced evident and consistent protective effects in all monitored experimental systems. In contrast, sunflower oil exhibited some adverse effects. Curcumin did not produce any significant effect in the bone marrow of both CS-exposed adults and foetuses but it elicited a dose-dependent protective effect traceable in blood erythrocytes. However, the higher curcumin dose further increased the frequency of micronucleated pulmonary alveolar macrophages. The apparent protective effects produced by lycopene and by a carotenoid mix were overwhelmed by those produced by olive oil, and lycopene even exhibited a worsening effect on the frequency of micronucleated erythroblasts in the bone marrow of urethane-treated adult mice.
Collapse
Affiliation(s)
- Roumen Balansky
- University Specialized Hospital for Active Treatment in Oncology, Sofia, 1756, Bulgaria
| | | | - Vessela D Kancheva
- Institute of Organic Chemistry, Center of Phytochemistry, Bulgarian Academy of Sciences, Sofia, 1113, Bulgaria
| | - Aleksei V Trofimov
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Lachezar Djongov
- University Specialized Hospital for Active Treatment in Oncology, Sofia, 1756, Bulgaria
| | - Silvio De Flora
- Department of Health Sciences, University of Genoa, 16139, Genoa, Italy.
| |
Collapse
|
7
|
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: 15] [Impact Index Per Article: 3.8] [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
|
8
|
Yu LC, Miao JK, Li WB, Chen N, Chen QX. Intranasal IL-4 Administration Alleviates Functional Deficits of Periventricular Leukomalacia in Neonatal Mice. Front Neurol 2020; 11:930. [PMID: 32982939 PMCID: PMC7492203 DOI: 10.3389/fneur.2020.00930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/17/2020] [Indexed: 01/23/2023] Open
Abstract
Background: Periventricular leukomalacia (PVL) is the major form of brain injury in premature infants. Currently, there are no therapies to treat PVL. Several studies suggested that polarization of microglia, a resident macrophage-like immune cell in the central nervous system, plays a vital role in brain injury and recovery. As an important mediator of immunity, interleukin-4 (IL-4) has critical effects on many immune cells, such as astrocytes and microglia. Increasing evidence shows that IL-4 plays a well-established role in attenuating inflammation in neurological disorders. Additionally, as a noninvasive and highly effective method, intranasal drug administration is gaining increasing attention. Therefore, in our study, we hypothesized that intranasal IL-4 administration is a promising strategy for PVL treatment. Methods: The therapeutic effects of IL-4 on neuroprotection were evaluated using a Control group, Hypoxia group, and Hypoxia + IL-4 treatment group. The PVL mouse model was established by a severe acute hypoxia (SAH) protocol. Exogenous IL-4 was intranasally administered to investigate its neuroprotective effects. A functional study was used to investigate neurological deficits, immunohistochemical technology and Western blotting were used to detect protein levels, and electron microscopy was used to evaluate myelination. Results: The results suggested that hypoxia stimulated Iba1+ microglial activation, downregulated myelin-related gene (NG2, MAG, and MBP) expression, reduced MBP protein levels, and caused neurological deficits. However, the intranasal administration of exogenous IL-4 partially inhibited Iba1+ microglial activation, improved myelination, and alleviated neurological deficits. The mechanistic study showed that IL-4 improved myelination possibly through the IL-4Ra-mediated polarization of microglia from the M1 phenotype to the M2 phenotype. Conclusion: In summary, our findings demonstrated that the intranasal administration of exogenous IL-4 improves myelination and attenuates functional deficits in a hypoxia-induced PVL model. Intranasal IL-4 administration may be a promising strategy for PVL treatment, for which further mechanistic studies are urgent.
Collapse
Affiliation(s)
- Lin-Chao Yu
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Chongqing Key Laboratory of Child Health and Nutrition, Chongqing, China
| | - Jing-Kun Miao
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Health Center for Women and Children, Chongqing, China
| | - Wei-Bin Li
- Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Na Chen
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Qi-Xiong Chen
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| |
Collapse
|
9
|
The effects of IL-4 and RANKL on viability of giant cell granuloma patients' monocytes. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
10
|
Balabiyev A, Podolnikova NP, Mursalimov A, Lowry D, Newbern JM, Roberson RW, Ugarova TP. Transition of podosomes into zipper-like structures in macrophage-derived multinucleated giant cells. Mol Biol Cell 2020; 31:2002-2020. [PMID: 32579434 PMCID: PMC7543064 DOI: 10.1091/mbc.e19-12-0707] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Macrophage fusion resulting in the formation of multinucleated giant cells (MGCs) is a multistage process that requires many adhesion-dependent steps and involves the rearrangement of the actin cytoskeleton. The diversity of actin-based structures and their role in macrophage fusion is poorly understood. In this study, we revealed hitherto unrecognized actin-based zipper-like structures (ZLSs) that arise between MGCs formed on the surface of implanted biomaterials. We established an in vitro model for the induction of these structures in mouse macrophages undergoing IL-4–mediated fusion. Using this model, we show that over time MGCs develop cell–cell contacts containing ZLSs. Live-cell imaging using macrophages isolated from mRFP- or eGFP-LifeAct mice demonstrated that ZLSs are dynamic formations undergoing continuous assembly and disassembly and that podosomes are precursors of these structures. Immunostaining experiments showed that vinculin, talin, integrin αMβ2, and other components of podosomes are present in ZLSs. Macrophages deficient in WASp or Cdc42, two key molecules involved in actin core organization in podosomes, as well as cells treated with the inhibitors of the Arp2/3 complex, failed to form ZLSs. Furthermore, E-cadherin and nectin-2 were found between adjoining membranes, suggesting that the transition of podosomes into ZLSs is induced by bridging plasma membranes by junctional proteins.
Collapse
Affiliation(s)
- Arnat Balabiyev
- School of Life Sciences, Arizona State University, Tempe, AZ 85287
| | | | - Aibek Mursalimov
- School of Life Sciences, Arizona State University, Tempe, AZ 85287
| | - David Lowry
- Eyring Materials Center, Arizona State University, Tempe, AZ 85287
| | - Jason M Newbern
- School of Life Sciences, Arizona State University, Tempe, AZ 85287
| | | | | |
Collapse
|
11
|
Dutra WO, Barbosa DF, de Souza PEA, Morgan D, Poetker S, Guimarães LH, Bacelar O, Gollob KJ, Carvalho EM. A Th2-Type Response Is Associated With Exuberant Lesions in Pregnant Women Infected With Leishmania braziliensis. J Infect Dis 2019; 219:480-488. [PMID: 30165577 DOI: 10.1093/infdis/jiy510] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/23/2018] [Indexed: 11/14/2022] Open
Abstract
Background Cutaneous leishmaniasis (CL) is characterized by an exaggerated inflammatory response. During pregnancy there is a decreased inflammatory response, and we have shown that pregnant women with CL develop exuberant lesions. Methods Cytokine production by peripheral blood mononuclear cells and the frequency of cells expressing cytokines in lesions from pregnant and nonpregnant women with CL were evaluated. Results We observed that CL lesions from pregnant women displayed a more intense cellular infiltrate, associated with an increase in neutrophils and CD4+ cells. While no difference was observed regarding the number of interferon-gamma (IFN-γ)+ cells in lesions from pregnant compared to nonpregnant women with CL, interleukin-10 (IL-10) and IL-4 expression were approximately 3-times higher in lesions in pregnant women. Main sources of IL-4 and IL-10 were CD4+ and CD68+ cells, respectively. Expression of IL-4, but not IFN-γ or IL-10, was positively correlated with the intensity of inflammatory infiltrate in lesions from pregnant women. Conclusions These results provide evidence of an IL-4-mediated pathology in Leishmania braziliensis-infected pregnant women. These differences in lesion pathogenesis in pregnant and nonpregnant women may open possibilities for new therapies for CL treatment during pregnancy, which are currently lacking.
Collapse
Affiliation(s)
- Walderez O Dutra
- Laboratory of Cell-Cell Interactions, Department of Morphology, Biological Sciences Institute, Universidade Federal de Minas Gerais, Belo Horizonte
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Salvador, Bahia, Brazil
| | - Daniela Faria Barbosa
- Laboratory of Cell-Cell Interactions, Department of Morphology, Biological Sciences Institute, Universidade Federal de Minas Gerais, Belo Horizonte
- Centro Universitário de Formiga, Minas Gerais
| | | | - Daniel Morgan
- Division of International Medicine and Infectious Diseases, Department of Medicine, Weill Cornell Medical College, New York, NewYork
| | - Shelene Poetker
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medical College, New York, NewYork
| | - Luiz Henrique Guimarães
- Immunology Service, Hospital Universitário Prof. Edgar Santos, Universidade Federal da Bahia, Salvador
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Salvador, Bahia, Brazil
| | - Olívia Bacelar
- Immunology Service, Hospital Universitário Prof. Edgar Santos, Universidade Federal da Bahia, Salvador
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Salvador, Bahia, Brazil
| | - Kenneth J Gollob
- A.C. Camargo Cancer Center, São Paulo, Universidade Federal da Bahia, Salvador
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Salvador, Bahia, Brazil
| | - Edgar M Carvalho
- Immunology Service, Hospital Universitário Prof. Edgar Santos, Universidade Federal da Bahia, Salvador
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Salvador, Bahia, Brazil
| |
Collapse
|
12
|
Faust JJ, Balabiyev A, Heddleston JM, Podolnikova NP, Baluch DP, Chew TL, Ugarova TP. An actin-based protrusion originating from a podosome-enriched region initiates macrophage fusion. Mol Biol Cell 2019; 30:2254-2267. [PMID: 31242090 PMCID: PMC6743464 DOI: 10.1091/mbc.e19-01-0009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/15/2019] [Accepted: 06/17/2019] [Indexed: 01/24/2023] Open
Abstract
Macrophage fusion resulting in the formation of multinucleated giant cells occurs in a variety of chronic inflammatory diseases, yet the mechanism responsible for initiating this process is unknown. Here, we used live cell imaging to show that actin-based protrusions at the leading edge initiate macrophage fusion. Phase-contrast video microscopy demonstrated that in the majority of events, short protrusions (∼3 µm) between two closely apposed cells initiated fusion, but occasionally we observed long protrusions (∼12 µm). Using macrophages isolated from LifeAct mice and imaging with lattice light sheet microscopy, we further found that fusion-competent protrusions formed at sites enriched in podosomes. Inducing fusion in mixed populations of GFP- and mRFP-LifeAct macrophages showed rapid spatial overlap between GFP and RFP signal at the site of fusion. Cytochalasin B strongly reduced fusion and when rare fusion events occurred, protrusions were not observed. Fusion of macrophages deficient in Wiskott-Aldrich syndrome protein and Cdc42, key molecules involved in the formation of actin-based protrusions and podosomes, was also impaired both in vitro and in vivo. Finally, inhibiting the activity of the Arp2/3 complex decreased fusion and podosome formation. Together these data suggest that an actin-based protrusion formed at the leading edge initiates macrophage fusion.
Collapse
Affiliation(s)
- James J. Faust
- School of Life Sciences, Arizona State University, Tempe, AZ 85287
| | - Arnat Balabiyev
- School of Life Sciences, Arizona State University, Tempe, AZ 85287
| | - John M. Heddleston
- Advanced Imaging Center, HHMI Janelia Research Campus, Ashburn, VA 20147
| | | | - D. Page Baluch
- School of Life Sciences, Arizona State University, Tempe, AZ 85287
| | - Teng-Leong Chew
- Advanced Imaging Center, HHMI Janelia Research Campus, Ashburn, VA 20147
| | | |
Collapse
|
13
|
Pereira M, Petretto E, Gordon S, Bassett JHD, Williams GR, Behmoaras J. Common signalling pathways in macrophage and osteoclast multinucleation. J Cell Sci 2018; 131:131/11/jcs216267. [PMID: 29871956 DOI: 10.1242/jcs.216267] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Macrophage cell fusion and multinucleation are fundamental processes in the formation of multinucleated giant cells (MGCs) in chronic inflammatory disease and osteoclasts in the regulation of bone mass. However, this basic cell phenomenon is poorly understood despite its pathophysiological relevance. Granulomas containing multinucleated giant cells are seen in a wide variety of complex inflammatory disorders, as well as in infectious diseases. Dysregulation of osteoclastic bone resorption underlies the pathogenesis of osteoporosis and malignant osteolytic bone disease. Recent reports have shown that the formation of multinucleated giant cells and osteoclast fusion display a common molecular signature, suggesting shared genetic determinants. In this Review, we describe the background of cell-cell fusion and the similar origin of macrophages and osteoclasts. We specifically focus on the common pathways involved in osteoclast and MGC fusion. We also highlight potential approaches that could help to unravel the core mechanisms underlying bone and granulomatous disorders in humans.
Collapse
Affiliation(s)
- Marie Pereira
- Centre for Inflammatory Disease, Imperial College London, London W12 0NN, UK
| | - Enrico Petretto
- Duke-NUS Medical School, Singapore 169857, Republic of Singapore
| | - Siamon Gordon
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan City 33302, Taiwan.,Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - J H Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Graham R Williams
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Jacques Behmoaras
- Centre for Inflammatory Disease, Imperial College London, London W12 0NN, UK
| |
Collapse
|
14
|
Abstract
Granulomas are organized aggregates of macrophages, often with characteristic morphological changes, and other immune cells. These evolutionarily ancient structures form in response to persistent particulate stimuli-infectious or noninfectious-that individual macrophages cannot eradicate. Granulomas evolved as protective responses to destroy or sequester particles but are frequently pathological in the context of foreign bodies, infections, and inflammatory diseases. We summarize recent findings that suggest that the granulomatous response unfolds in a stepwise program characterized by a series of macrophage activations and transformations that in turn recruit additional cells and produce structural changes. We explore why different granulomas vary and the reasons that granulomas are protective and pathogenic. Understanding the mechanisms and role of granuloma formation may uncover new therapies for the multitude of granulomatous diseases that constitute serious medical problems while enhancing the protective function of granulomas in infections.
Collapse
Affiliation(s)
- Antonio J Pagán
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, United Kingdom; , .,MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Lalita Ramakrishnan
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, United Kingdom; , .,MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| |
Collapse
|
15
|
Horn V, Triantafyllopoulou A. DNA damage signaling and polyploid macrophages in chronic inflammation. Curr Opin Immunol 2017; 50:55-63. [PMID: 29202328 DOI: 10.1016/j.coi.2017.11.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 10/30/2017] [Accepted: 11/17/2017] [Indexed: 01/01/2023]
Abstract
Whole genome duplications, an important step in cancer development, also occur in the macrophage lineage in disease: large multinucleated macrophages found within compact, ordered aggregates of immune cells, called granulomas, are a well-known histologic entity. Very recent work suggests that granuloma macrophages remarkably acquire epithelial cell features and the genotoxic stress response instructs granuloma macrophage genome duplications, suggesting that granuloma macrophages and pre-malignant epithelial cells may share common mechanisms of adaptation to chronic genotoxic stress. Exploring these mechanisms is key for a better understanding of the pathogenesis of chronic inflammatory diseases. Here we review the mechanisms of macrophage polyploidization, the role of DNA damage signaling in this process and the function of polyploid macrophages, with a focus on chronic inflammation.
Collapse
Affiliation(s)
- Veronika Horn
- Department of Rheumatology and Clinical Immunology, Charité University Medical Center, D-10117 Berlin, Germany; Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, D-79106 Freiburg, Germany
| | - Antigoni Triantafyllopoulou
- Department of Rheumatology and Clinical Immunology, Charité University Medical Center, D-10117 Berlin, Germany; Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, D-79106 Freiburg, Germany; German Rheumatism Research Center, A Leibniz Institute, D-10117 Berlin, Germany; Institute of Microbiology, Charité University Medical Center, D-12203 Berlin, Germany.
| |
Collapse
|
16
|
CD300f:IL-5 cross-talk inhibits adipose tissue eosinophil homing and subsequent IL-4 production. Sci Rep 2017; 7:5922. [PMID: 28725048 PMCID: PMC5517555 DOI: 10.1038/s41598-017-06397-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/09/2017] [Indexed: 12/22/2022] Open
Abstract
Eosinophils and their associated cytokines IL-4 and IL-5 are emerging as central orchestrators of the immune-metabolic axis. Herein, we demonstrate that cross-talk between the Ig-superfamily receptor CD300f and IL-5 is a key checkpoint that modifies the ability of eosinophils to regulate metabolic outcomes. Generation of Il5 Tg /Cd300f -/- mice revealed marked and distinct increases in eosinophil levels and their production of IL-4 in the white and brown adipose tissues. Consequently, Il5 Tg /Cd300f -/- mice had increased alternatively activated macrophage accumulation in the adipose tissue. Cd300f -/- mice displayed age-related accumulation of eosinophils and macrophages in the adipose tissue and decreased adipose tissue weight, which was associated with decreased diet-induced weight gain and insulin resistance. Notably, Il5 Tg /CD300f -/- were protected from diet-induced weight gain and glucose intolerance. These findings highlight the cross-talk between IL-5 receptor and CD300f as a novel pathway regulating adipose tissue eosinophils and offer new entry points for therapeutic intervention for obesity and its complications.
Collapse
|
17
|
Development of fusogenic glass surfaces that impart spatiotemporal control over macrophage fusion: Direct visualization of multinucleated giant cell formation. Biomaterials 2017; 128:160-171. [PMID: 28340410 DOI: 10.1016/j.biomaterials.2017.02.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 02/21/2017] [Accepted: 02/26/2017] [Indexed: 01/07/2023]
Abstract
Implantation of synthetic material, including vascular grafts, pacemakers, etc. results in the foreign body reaction and the formation of multinucleated giant cells (MGCs) at the exterior surface of the implant. Despite the long-standing premise that fusion of mononucleated macrophages results in the formation of MGCs, to date, no published study has shown fusion in context with living specimens. This is due to the fact that optical-quality glass, which is required for the majority of live imaging techniques, does not promote macrophage fusion. Consequently, the morphological changes that macrophages undergo during fusion as well as the mechanisms that govern this process remain ill-defined. In this study, we serendipitously identified a highly fusogenic glass surface and discovered that the capacity to promote fusion was due to oleamide contamination. When adsorbed on glass, oleamide and other molecules that contain long-chain hydrocarbons promoted high levels of macrophage fusion. Adhesion, an essential step for macrophage fusion, was apparently mediated by Mac-1 integrin (CD11b/CD18, αMβ2) as determined by single cell force spectroscopy and adhesion assays. Micropatterned glass further increased fusion and enabled a remarkable degree of spatiotemporal control over MGC formation. Using these surfaces, we reveal the kinetics that govern MGC formation in vitro. We anticipate that the spatiotemporal control afforded by these surfaces will expedite studies designed to identify the mechanism(s) of macrophage fusion and MGC formation with implication for the design of novel biomaterials.
Collapse
|
18
|
Klopfleisch R. Macrophage reaction against biomaterials in the mouse model - Phenotypes, functions and markers. Acta Biomater 2016; 43:3-13. [PMID: 27395828 DOI: 10.1016/j.actbio.2016.07.003] [Citation(s) in RCA: 229] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 06/08/2016] [Accepted: 07/05/2016] [Indexed: 02/07/2023]
Abstract
UNLABELLED The foreign body reaction (FBR) is a response of the host tissue against more or less degradation-resistant foreign macromolecular material. The reaction is divided into five different phases which involve most aspects of the innate and the adaptive immune system: protein adsorption, acute and chronic inflammation, foreign body giant cell formation and fibrosis. It is long known, that macrophages play a central role in all of these phases except for protein adsorption. Initially it was believed that the macrophage driven FBR has a complete negative effect on biocompatibility. Recent progress in biomaterial and macrophage research however describe macrophages as more than pure antigen phagocytosing and presenting cells and thus pro-inflammatory cells involved in biomaterial encapsulation and failure. Quite contrary, both, pro-inflammatory M1 macrophages, the diverse regulatory M2 macrophage subtypes and even foreign body giant cells (FBGC) are after necessary for integration of non-degradable biomaterials and degradation and replacement of degradable biomaterials. This review gives a comprehensive overview on the taxonomy of the currently known macrophage subtypes. Their diverging functions, metabolism and markers are summarized and the relevance of this more diverse macrophage picture for the design of biomaterials is shortly discussed. STATEMENT OF SIGNIFICANCE The view on role of macrophages in the foreign body reaction against biomaterials is rapidly changing. Despite the initial idea that macrophage are mainly involved in undesired degradation and biomaterial rejection it becomes now clear that they are nevertheless necessary for proper integration of non-degradable biomaterials and degradation of placeholder, degradable biomaterials. As a pathologist I experienced a lack on a good summary on the current taxonomy, functions and phenotypes of macrophages in my recent projects on the biocompatibility of biomaterials in the mouse model. The submitted review therefore intends to gives a comprehensive overview on the taxonomy of the currently known macrophage subtypes. Their diverging functions, metabolism and markers are summarized and the relevance of this more diverse macrophage picture for the design of biomaterials is shortly discussed.
Collapse
Affiliation(s)
- R Klopfleisch
- Institute of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Straße 15, Berlin 14163, Germany.
| |
Collapse
|
19
|
McClean CM, Tobin DM. Macrophage form, function, and phenotype in mycobacterial infection: lessons from tuberculosis and other diseases. Pathog Dis 2016; 74:ftw068. [PMID: 27402783 DOI: 10.1093/femspd/ftw068] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2016] [Indexed: 02/07/2023] Open
Abstract
Macrophages play a central role in mycobacterial pathogenesis. Recent work has highlighted the importance of diverse macrophage types and phenotypes that depend on local environment and developmental origins. In this review, we highlight how distinct macrophage phenotypes may influence disease progression in tuberculosis. In addition, we draw on work investigating specialized macrophage populations important in cancer biology and atherosclerosis in order to suggest new areas of investigation relevant to mycobacterial pathogenesis. Understanding the mechanisms controlling the repertoire of macrophage phenotypes and behaviors during infection may provide opportunities for novel control of disease through modulation of macrophage form and function.
Collapse
Affiliation(s)
- Colleen M McClean
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, DUMC 3020, Durham, NC 27710, USA Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA Medical Scientist Training Program, Duke University School of Medicine, Durham, NC 27710, USA
| | - David M Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, DUMC 3020, Durham, NC 27710, USA Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
20
|
Abstract
No survey of clinically important immunological phenomena would be complete without consideration of the functions of phagocytic cells. They play a pivotal role in the immune response by kiling microbes, by presenting antigens to lymphocytes and by serving as supportive, accessory cells to lymphocytes, at least partly by releasing soluble factors. The phagocytes of the body, professional and non professional, consist of two specialized groups of cells: granulocytes, which can be mobilized rapidly and which reach inflamed sites quickly and in large numbers, and which are highly efficient at dealing with many types of injury and infection but which have no capacity for differentiation and live only a short time; and the mononuclear phagocyte system consisting partly of motile cells which respond initially more slowly than neutrophils but which can differentiate in sites of inflammation into cells which are more efficient in various functions than the cells from which they originated. Many mononuclear phagocytes are fixed cells located in tissues where they act as trays or filters for material circulating through the tissue. Phagocytes, which usually function as the primary defender in infections, have also been implicated as effector cells in several conditions characterized by a destructive inflammatory response.
Collapse
Affiliation(s)
- G. Ricevuti
- Department of Internal Medicine and Therapeutics, Section of Medical Pathology, University of Pavia, IRCCS Policlinico S. Matteo, 27100 Pavia, Italy
| | - A. Mazzone
- Department of Internal Medicine and Therapeutics, Section of Medical Pathology, University of Pavia, IRCCS Policlinico S. Matteo, 27100 Pavia, Italy
| | - A. Notario
- Department of Internal Medicine and Therapeutics, Section of Medical Pathology, University of Pavia, IRCCS Policlinico S. Matteo, 27100 Pavia, Italy
| |
Collapse
|
21
|
Gibon E, Córdova LA, Lu L, Lin TH, Yao Z, Hamadouche M, Goodman SB. The biological response to orthopedic implants for joint replacement. II: Polyethylene, ceramics, PMMA, and the foreign body reaction. J Biomed Mater Res B Appl Biomater 2016; 105:1685-1691. [PMID: 27080740 DOI: 10.1002/jbm.b.33676] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 02/02/2016] [Accepted: 03/20/2016] [Indexed: 11/10/2022]
Abstract
Novel evidence-based prosthetic designs and biomaterials facilitate the performance of highly successful joint replacement (JR) procedures. To achieve this goal, constructs must be durable, biomechanically sound, and avoid adverse local tissue reactions. Different biomaterials such as metals and their alloys, polymers, ceramics, and composites are currently used for JR implants. This review focuses on (1) the biological response to the different biomaterials used for TJR and (2) the chronic inflammatory and foreign-body response induced by byproducts of these biomaterials. A homeostatic state of bone and surrounding soft tissue with current biomaterials for JR can be achieved with mechanically stable, infection free and intact (as opposed to the release of particulate or ionic byproducts) implants. Adverse local tissue reactions (an acute/chronic inflammatory reaction, periprosthetic osteolysis, loosening and subsequent mechanical failure) may evolve when the latter conditions are not met. This article (Part 2 of 2) summarizes the biological response to the non-metallic materials commonly used for joint replacement including polyethylene, ceramics, and polymethylmethacrylate (PMMA), as well as the foreign body reaction to byproducts of these materials. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 1685-1691, 2017.
Collapse
Affiliation(s)
- Emmanuel Gibon
- Department of Orthopedic Surgery, Stanford University, Stanford, California.,Laboratoire de Biomécanique et Biomatériaux Ostéo-Articulaires-UMR CNRS 7052, Faculté de Médecine-Université Paris 7, Paris, France.,Department of Orthopedic Surgery, Hopital Cochin, APHP, Paris, France
| | - Luis A Córdova
- Department of Orthopedic Surgery, Stanford University, Stanford, California.,Department of Oral and Maxillofacial Surgery, University of Chile-Conicyt, Santiago, Chile
| | - Laura Lu
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Tzu-Hua Lin
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Zhenyu Yao
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Moussa Hamadouche
- Laboratoire de Biomécanique et Biomatériaux Ostéo-Articulaires-UMR CNRS 7052, Faculté de Médecine-Université Paris 7, Paris, France.,Department of Orthopedic Surgery, Hopital Cochin, APHP, Paris, France
| | - Stuart B Goodman
- Department of Orthopedic Surgery, Stanford University, Stanford, California.,Department of Bioengineering, Stanford University, Stanford, California
| |
Collapse
|
22
|
Macrophage and Multinucleated Giant Cell Classification. CURRENT TOPICS IN ENVIRONMENTAL HEALTH AND PREVENTIVE MEDICINE 2016. [DOI: 10.1007/978-4-431-55732-6_1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
23
|
Kumarguru BN, Natarajan M, Biligi DS, Raghupathi AR. Giant Cell Lesions of Lungs: A Histopathological and Morphometric Study of Seven Autopsy Cases. J Clin Diagn Res 2015; 9:EC12-6. [PMID: 26673670 DOI: 10.7860/jcdr/2015/15035.6786] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 09/03/2015] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Macrophages undergo fusion to form multinucleated giant cells (MGC) in several pathologic conditions. The exact mechanism of their generation is still unclear. MGC are a common feature of granulomas that develop during various inflammatory reactions. AIM To study the histopathological features of giant cell lesions in lungs and correlate the characteristics of giant cells with other histopathological findings. Also, to determine the utility of morphometry to differentiate foreign body and Langhans MGC. MATERIALS AND METHODS Seven cases were analysed. Specimen of lungs was grossed, sectioned and processed. Routinely, tissue sections were stained by Haematoxylin and Eosin (H&E) stain. Polarizing microscopy and special stains were employed in selected cases. Granulomas and MGC were counted and measured. Several other parameters like location, distribution, type and number of MGC, associated predominant inflammatory component and nature of granulomas were analysed. RESULTS Five patterns of lesions were observed in seven cases. Aspiration pneumonia was seen in three cases (42.85%) and constituted the most common pattern. However, aspiration pneumonia as the only cause of MGC was seen in only one case (14.28%). Pulmonary tuberculosis and asteroid bodies constituted two cases (28.57%) each. Cryptococcal pneumonia and cholesterol clefts constituted one case (14.28%) each. Crypococci were demonstrated to be positively birefringent by polarized microscopy on Ziehl-Neelsen stained sections. Based on statistical analysis of morphometric data, a new index (NP index) was proposed to statistically categorize MGC into foreign body type and Langhans type. NP index value of ≤0.016 was found to be statistically significant (p<0.005) in foreign body MGC. It had high sensitivity and efficacy. CONCLUSION MGC may not be always associated with granulomas. The mechanisms that lead to the occurrence of MGC, independent of granuloma needs to be elucidated. Morphometry may serve as a useful aid. But a pathologist has to rely on the morphological details to categorize MGC.
Collapse
Affiliation(s)
- B N Kumarguru
- Assistant Professor, Department of Pathology, PES Institute of Medical sciences and Research , Kuppam, Andhra Pradesh, India
| | - M Natarajan
- Professor, Department of Pathology, Bangalore Medical College and Research Institute , Bangalore, Karnataka, India
| | - Dayananda S Biligi
- Professor, Department of Pathology, Bangalore Medical College and Research Institute , Bangalore, Karnataka, India
| | - A R Raghupathi
- Professor, Department of Pathology, Bangalore Medical College and Research Institute , Bangalore, Karnataka, India
| |
Collapse
|
24
|
Levaot N, Ottolenghi A, Mann M, Guterman-Ram G, Kam Z, Geiger B. Osteoclast fusion is initiated by a small subset of RANKL-stimulated monocyte progenitors, which can fuse to RANKL-unstimulated progenitors. Bone 2015; 79:21-8. [PMID: 26008608 DOI: 10.1016/j.bone.2015.05.021] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 05/09/2015] [Accepted: 05/15/2015] [Indexed: 10/23/2022]
Abstract
Osteoclasts are multinucleated, bone-resorbing cells formed via fusion of monocyte progenitors, a process triggered by prolonged stimulation with RANKL, the osteoclast master regulator cytokine. Monocyte fusion into osteoclasts has been shown to play a key role in bone remodeling and homeostasis; therefore, aberrant fusion may be involved in a variety of bone diseases. Indeed, research in the last decade has led to the discovery of genes regulating osteoclast fusion; yet the basic cellular regulatory mechanism underlying the fusion process is poorly understood. Here, we applied a novel approach for tracking the fusion processes, using live-cell imaging of RANKL-stimulated and non-stimulated progenitor monocytes differentially expressing dsRED or GFP, respectively. We show that osteoclast fusion is initiated by a small (~2.4%) subset of precursors, termed "fusion founders", capable of fusing either with other founders or with non-stimulated progenitors (fusion followers), which alone, are unable to initiate fusion. Careful examination indicates that the fusion between a founder and a follower cell consists of two distinct phases: an initial pairing of the two cells, typically lasting 5-35 min, during which the cells nevertheless maintain their initial morphology; and the fusion event itself. Interestingly, during the initial pre-fusion phase, a transfer of the fluorescent reporter proteins from nucleus to nucleus was noticed, suggesting crosstalk between the founder and follower progenitors via the cytoplasm that might directly affect the fusion process, as well as overall transcriptional regulation in the developing heterokaryon.
Collapse
Affiliation(s)
- Noam Levaot
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | - Aner Ottolenghi
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Mati Mann
- Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Gali Guterman-Ram
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Zvi Kam
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Benjamin Geiger
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
25
|
Interleukin-4 Expressed By Neoplastic Cells Provokes an Anti-Metastatic Myeloid Immune Response. JOURNAL OF CLINICAL & CELLULAR IMMUNOLOGY 2015; 6:1-9. [PMID: 27563494 PMCID: PMC4995104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Interleukin-4 (IL-4) can induce macrophages to undergo alternative activation and polarize toward an M2-like or wound healing phenotype. Tumor associated macrophages (TAMs) are thought to assume M2-like properties, and it has been suggested they promote tumor growth and metastasis through effects on the tumor stroma, including extracelluar matrix remodeling and angiogenesis. IL-4 also promotes macrophage survival and formation of multinucleated giant cells, which have enhanced phagocytic behavior. This study was designed to explore the effect of cancer cell derived IL-4 on the tumor immune stroma and metastasis. METHODS The metastatic mouse mammary carcinoma cell line AC2M2 was transduced with control or IL-4 encoding retroviruses and employed in orthotopic engraftment models. Tumor growth and metastasis were assessed. The cellular composition and biomarker expression of tumors were examined by immunohistochemical staining and flow cytometry; the transcriptome of the immune stroma was analyzed by nanoString based transcript quantitation; and in vivo and in vitro interactions between cancer cells and macrophages were assessed by flow cytometry and co-culture with video-time lapse microscopy, respectively. RESULTS Unexpectedly, tumors from IL-4 expressing AC2M2 engrafted cells grew at reduced rates, and most surprising, they lost all metastatic potential relative to tumors from control AC2M2 cells. Myeloid cell numbers were not increased in IL-4 expressing tumors, but their expression of the M2 marker arginase I was elevated. Transcriptome analysis revealed an immune signature consistent with IL-4 induced M2 polarization of the tumor microenvironment and a generalized increase in myeloid involvement in the tumor stroma. Flow cytometry analysis indicated enhanced cancer cell phagocytosis by TAMs from IL-4 expressing tumors, and co-culture studies showed that IL-4 expressing cancer cells supported the survival and promoted the in vitro phagocytic behavior of macrophages. CONCLUSIONS Although M2-like TAMs have been linked to enhanced tumorigenesis, this study shows that IL-4 production by cancer cells is associated with suppressed tumor growth and loss of metastatic potential as well as enhanced phagocytic behavior of TAMs.
Collapse
|
26
|
Sheikh F, Dickensheets H, Pedras-Vasconcelos J, Ramalingam T, Helming L, Gordon S, Donnelly RP. The Interleukin-13 Receptor-α1 Chain Is Essential for Induction of the Alternative Macrophage Activation Pathway by IL-13 but Not IL-4. J Innate Immun 2015; 7:494-505. [PMID: 25766112 DOI: 10.1159/000376579] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/29/2015] [Indexed: 12/17/2022] Open
Abstract
Macrophages coexpress both the interleukin (IL)-2Rγ chain (γ(c)) and IL-13Rα1. These receptor chains can heterodimerize with IL-4Rα to form type I or type II IL-4 receptor complexes, respectively. We used macrophages derived from Il2rg and Il13ra1 knockout (KO) mice to evaluate the requirements for these receptor chains for induction of the alternative macrophage activation (AMA) pathway by IL-4 and IL-13. Absence of γ(c) significantly decreased activation of STAT6 by IL-4 but not IL-13. However, although activation of STAT6 by IL-4 was markedly reduced in γ(c) KO macrophages, it was not abolished, indicating that IL-4 can still signal through type II IL-4 receptors via the IL-13Rα1 chain. IL-13 failed to activate STAT6 in macrophages derived from Il13ra1 KO mice; however, these cells remained fully responsive to IL-4. The inability of IL-13 but not IL-4 to signal in Il13ra1(-/-) macrophages correlated with the inability of IL-13 but not IL-4 to induce expression of genes such as Arg1, Retnla and Ccl11 that are characteristically expressed by alternatively activated macrophages. In addition, IL-13 but not IL-4 failed to induce membrane fusion and giant cell formation by Il13ra1 KO macrophages. These findings demonstrate that the IL-13Rα1 chain is essential for induction of the AMA pathway by IL-13 but not IL-4.
Collapse
Affiliation(s)
- Faruk Sheikh
- Division of Therapeutic Proteins, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Md., USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Kweon H, Kim SG, Choi JY. Inhibition of foreign body giant cell formation by 4- hexylresorcinol through suppression of diacylglycerol kinase delta gene expression. Biomaterials 2014; 35:8576-84. [DOI: 10.1016/j.biomaterials.2014.06.050] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 06/26/2014] [Indexed: 12/22/2022]
|
28
|
Pegoraro G, Eaton BP, Ulrich RL, Lane DJ, Ojeda JF, Bavari S, DeShazer D, Panchal RG. A high-content imaging assay for the quantification of the Burkholderia pseudomallei induced multinucleated giant cell (MNGC) phenotype in murine macrophages. BMC Microbiol 2014; 14:98. [PMID: 24750902 PMCID: PMC4077104 DOI: 10.1186/1471-2180-14-98] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 04/11/2014] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Burkholderia pseudomallei (Bp), a Gram-negative, motile, facultative intracellular bacterium is the causative agent of melioidosis in humans and animals. The Bp genome encodes a repertoire of virulence factors, including the cluster 3 type III secretion system (T3SS-3), the cluster 1 type VI secretion system (T6SS-1), and the intracellular motility protein BimA, that enable the pathogen to invade both phagocytic and non-phagocytic cells. A unique hallmark of Bp infection both in vitro and in vivo is its ability to induce cell-to-cell fusion of macrophages to form multinucleated giant cells (MNGCs), which to date are semi-quantitatively reported following visual inspection. RESULTS In this study we report the development of an automated high-content image acquisition and analysis assay to quantitate the Bp induced MNGC phenotype. Validation of the assay was performed using T6SS-1 (∆hcp1) and T3SS-3 (∆bsaZ) mutants of Bp that have been previously reported to exhibit defects in their ability to induce MNGCs. Finally, screening of a focused small molecule library identified several Histone Deacetylase (HDAC) inhibitors that inhibited Bp-induced MNGC formation of macrophages. CONCLUSIONS We have successfully developed an automated HCI assay to quantitate MNGCs induced by Bp in macrophages. This assay was then used to characterize the phenotype of the Bp mutants for their ability to induce MNGC formation and identify small molecules that interfere with this process. Successful application of chemical genetics and functional reverse genetics siRNA approaches in the MNGC assay will help gain a better understanding of the molecular targets and cellular mechanisms responsible for the MNGC phenotype induced by Bp, by other bacteria such as Mycobacterium tuberculosis, or by exogenously added cytokines.
Collapse
Affiliation(s)
- Gianluca Pegoraro
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702-5011, USA
- Perkin Elmer, Waltham, MA 02451, USA
- Present Address: Center for Cancer Research, National Cancer Institute/NIH, Bethesda, MD 20892, USA
| | - Brett P Eaton
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702-5011, USA
| | - Ricky L Ulrich
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702-5011, USA
| | - Douglas J Lane
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702-5011, USA
| | - Jenifer F Ojeda
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702-5011, USA
| | - Sina Bavari
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702-5011, USA
| | - David DeShazer
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702-5011, USA
| | - Rekha G Panchal
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702-5011, USA
| |
Collapse
|
29
|
|
30
|
Hu Y, Ek-Rylander B, Wendel M, Andersson G. Reciprocal effects of Interferon-γ and IL-4 on differentiation to osteoclast-like cells by RANKL or LPS. Oral Dis 2013; 20:682-92. [PMID: 24118341 DOI: 10.1111/odi.12189] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 09/18/2013] [Accepted: 09/21/2013] [Indexed: 01/25/2023]
Abstract
OBJECTIVE LPS can induce differentiation to osteoclast-like cells independent of RANKL. In comparison with RANKL, the effects of Th1 and Th2 cytokines on LPS-induced osteoclastogenesis have not been extensively studied. In this study, we investigated the effects of IFN-γ and IL-4 on RANKL- or LPS-induced osteoclastogenesis. MATERIALS AND METHODS RAW 264.7 cells were induced to differentiate into osteoclast-like cells by RANKL or LPS, in the absence or presence of IFN-γ or IL-4. The number of TRAP-positive, multinucleated (≥ 3 nuclei) cells (MNCs) was counted. mRNA and protein levels of TRAP and cathepsin K were determined by quantitative RT-PCR and Western immunoblot, respectively. Expression of other genes implicated in osteoclast and macrophage differentiation and inflammation was also quantitated and was subsequently assessed in bone marrow-derived macrophages (BMMs). Phagocytic capacity of differentiated RAW264.7 was investigated by the uptake of pHrodo S. aureus bioparticles conjugates. RESULTS In contrast to the RANKL-treated cell population that gained more macrophage-like properties at the level of gene and protein expression as well as phagocytosis in the presence of IFN-γ or IL-4, the LPS-induced population gained more osteoclast-like properties by the addition of the same factors. CONCLUSION These data suggest that the adaptive immune system, through either Th1 or Th2 cytokines, is able to modify the differentiation process of osteoclasts in inflammatory situations. Moreover, the study provides an example of different regulation of osteoclast differentiation during physiological and inflammatory conditions.
Collapse
Affiliation(s)
- Y Hu
- Institution of Dental Medicine, Qilu Hospital, Shandong University, Jinan, China; Department of Dental Medicine, Division of Oral Biology, Karolinska Institutet, Huddinge, Sweden
| | | | | | | |
Collapse
|
31
|
King JG, Hillyer JF. Spatial and temporal in vivo analysis of circulating and sessile immune cells in mosquitoes: hemocyte mitosis following infection. BMC Biol 2013; 11:55. [PMID: 23631603 PMCID: PMC3660217 DOI: 10.1186/1741-7007-11-55] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 04/17/2013] [Indexed: 11/10/2022] Open
Abstract
Background Mosquitoes respond to infection by mounting immune responses. The primary regulators of these immune responses are cells called hemocytes, which kill pathogens via phagocytosis and via the production of soluble antimicrobial factors. Mosquito hemocytes are circulated throughout the hemocoel (body cavity) by the swift flow of hemolymph (blood), and data show that some hemocytes also exist as sessile cells that are attached to tissues. The purpose of this study was to create a quantitative physical map of hemocyte distribution in the mosquito, Anopheles gambiae, and to describe the cellular immune response in an organismal context. Results Using correlative imaging methods we found that the number of hemocytes in a mosquito decreases with age, but that regardless of age, approximately 75% of the hemocytes occur in circulation and 25% occur as sessile cells. Infection induces an increase in the number of hemocytes, and tubulin and nuclear staining showed that this increase is primarily due to mitosis and, more specifically, autonomous cell division, by circulating granulocytes. The majority of sessile hemocytes are present on the abdominal wall, although significant numbers of hemocytes are also present in the thorax, head, and several of the appendages. Within the abdominal wall, the areas of highest hemocyte density are the periostial regions (regions surrounding the valves of the heart, or ostia), which are ideal locations for pathogen capture as these are areas of high hemolymph flow. Conclusions These data describe the spatial and temporal distribution of mosquito hemocytes, and map the cellular response to infection throughout the hemocoel.
Collapse
Affiliation(s)
- Jonas G King
- Department of Biological Sciences, Vanderbilt University, VU Station B 35-1634, Nashville, TN 37235, USA
| | | |
Collapse
|
32
|
Miyamoto T. STATs and macrophage fusion. JAKSTAT 2013; 2:e24777. [PMID: 24069561 PMCID: PMC3772113 DOI: 10.4161/jkst.24777] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 04/23/2013] [Accepted: 04/23/2013] [Indexed: 01/17/2023] Open
Abstract
Macrophages play a pivotal role in host defense against multiple foreign materials such as bacteria, parasites and artificial devices. Some macrophage lineage cells, namely osteoclasts and foreign body giant cells (FBGCs), form multi-nuclear giant cells by the cell-cell fusion of mono-nuclear cells. Osteoclasts are bone-resorbing cells, and are formed in the presence of RANKL on the surface of bones, while FBGCs are formed in the presence of IL-4 or IL-13 on foreign materials such as artificial joints, catheters and parasites. Recently, fusiogenic mechanisms and the molecules required for the cell-cell fusion of these macrophage lineage cells were, at least in part, clarified. Dendritic cell specific transmembrane protein (DC-STAMP) and osteoclast stimulatory transmembrane protein (OC-STAMP), both of which comprise seven transmembrane domains, are required for both osteoclast and FBGC cell-cell fusion. STAT6 was demonstrated to be required for the cell-cell fusion of FBGCs but not osteoclasts. In this review, advances in macrophage cell-cell fusion are discussed.
Collapse
Affiliation(s)
- Takeshi Miyamoto
- Department of Orthopedic Surgery; Department of Integrated Bone Metabolism and Immunology; Keio Kanrinmaru Project; Keio University School of Medicine; Tokyo, Japan
| |
Collapse
|
33
|
de Melo JF, da Costa TB, da Costa Lima TD, Chaves MEC, Vayssade M, Nagel MD, de Castro CMMB. Long-term effects of a neonatal low-protein diet in rats on the number of macrophages in culture and the expression/production of fusion proteins. Eur J Nutr 2012; 52:1475-82. [DOI: 10.1007/s00394-012-0453-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 09/27/2012] [Indexed: 01/21/2023]
|
34
|
Holt DJ, Grainger DW. Senescence and quiescence induced compromised function in cultured macrophages. Biomaterials 2012; 33:7497-507. [PMID: 22809642 DOI: 10.1016/j.biomaterials.2012.06.099] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 06/30/2012] [Indexed: 10/28/2022]
Abstract
Implants are predisposed to infection even years after implantation, despite ostensibly being surrounded by innumerable macrophages as part of the host foreign body response. The local implant environment could adversely influence the implant-associated macrophage phenotype, proliferative capacity, activation states, and ability to neutralize pathogens. This study monitored cultured macrophage proliferative states and phagocytotic competence on tissue culture plastic to address the hypothesis that extended contact with foreign materials alters macrophage phenotype. That such macrophage alterations might also occur around implants has significance to the foreign body response, infection, cancer, autoimmune and other diseases. Specifically, multiple indicators of macrophage proliferation in various culture conditions, including cell confluence, long-term culture (21 days), lipopolysaccharide (LPS) stimulation, passaging, and mitogenic stimulation are reported. Importantly, primary murine macrophages became quiescent at high confluence and senescent during long-term culture. Senescent macrophages significantly reduced their ability to phagocytose particles, while quiescent macrophages did not. Cell senescence and quiescence were not observed with repeated passaging. Primary macrophage stimulation with LPS delayed senescence but did not eliminate it. These results prompt the conclusion that both cell quiescence and senescence are observed under common macrophage culture conditions and could alter macrophage behavior and phenotypes in extended in vitro culture, such as the ability to phagocytose. Such macrophage transitions around foreign bodies in vivo are not documented: quiescence and senescence reported here in macrophage culture could be relevant to macrophage behavior both in vitro in bioassays and in vivo in the foreign body response and implant-centered infection.
Collapse
Affiliation(s)
- Dolly J Holt
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84112-5820, USA
| | | |
Collapse
|
35
|
Malhotra A, Nair P, Dhawan DK. Premature mitochondrial senescence and related ultrastructural changes during lung carcinogenesis modulation by curcumin and resveratrol. Ultrastruct Pathol 2012; 36:179-184. [PMID: 22559045 DOI: 10.3109/01913123.2011.652765] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The present study attempted to explore the efficacy of curcumin and resveratrol in modulating premature mitochondria senescence and ultrastructural changes during lung carcinogenesis. The mice were segregated into 5 groups, which included normal control, benzo[a]pyrene (BP) treated, BP + curcumin (C) treated, BP + resveratrol (R) treated, and BP + C + R treated groups. Animals were given a single ip injection of benzo[a]pyrene in corn oil at a dose level of 100 mg/kg body weight. Treatments of curcumin and resveratrol were given orally in drinking water at a dose level of 60 mg/kg body weight and 5.7 µg/mL drinking water, respectively, 3 times a week for a total duration of 22 weeks. Ultrastructure of BP-treated mice revealed disruptions in cellular integrity along with nuclear deformation and premature mitochondrial senescence. Interestingly, supplementation of curcumin and resveratrol individually resulted in improvement of ultrahistoarchitecture of BP-treated mice but the improvement was much greater with combined supplementation of phytochemicals. Further, benzo[a]pyrene treatment revealed alterations in lung histoarchitecture, which, however, was improved appreciably following combined supplementation with curcumin and resveratrol. The present study concludes that combined supplementation with curcumin and resveratrol effectively modulates histoarchitecture as well as ultrahistoarchitecture during benzo[a]pyrene-induced lung carcinogenesis in mice. Cancer is a public health problem worldwide. Lung cancer is a major cause of mortality throughout the world and is responsible for the deaths of more than one million people annually. Phytochemicals have shown great potential in preventing the occurrence of cancer and other chronic diseases that result from oxidative stress induced by free radicals. Phytochemicals are nonnutritive products of plants and, being nontoxic, are presently being studied the world over for their chemopreventive actions in controlling various diseases, including cancer. In the present study, curcumin and resveratrol are the phytochemicals of interest. Curcumin, a polyphenol, has been reported to have anti-invasive properties. Further, curcumin has been shown to activate apoptotic machinery in patients with lung cancer. On the other hand, resveratrol (trans-3,4,5- thihydroxystibene) is a phytoalexin that is present naturally in grapes as well as in a variety of medicinal plants and has been shown to exhibit antioxidant activity with a potential to induce apoptosis.
Collapse
Affiliation(s)
- Anshoo Malhotra
- Department of Biophysics, Panjab University, Chandigarh, India
| | | | | |
Collapse
|
36
|
Khandwekar A, Rho CK. Modulation of cellular responses on engineered polyurethane implants. J Biomed Mater Res A 2012; 100:2211-22. [PMID: 22492665 DOI: 10.1002/jbm.a.34146] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 01/30/2012] [Accepted: 02/09/2012] [Indexed: 12/18/2022]
Abstract
An in vivo rat cage implant system was used to study the effect of polyurethane surface chemistries on protein adsorption, macrophage adhesion, foreign-body giant cell formation (FBGCs), cellular apoptosis, and cytokine response. Polyurethanes with zwitterionic, anionic, and cationic chemistries were developed. The changes in the surface topography of the materials were determined using atomic force microscopy and the wettability by dynamic contact angle measurements. The in vitro protein adsorption studies revealed higher protein adsorption on cationic surfaces when compared with the base, while adsorption was significantly reduced on zwitterionic (**p < 0.01) and anionic (*p < 0.05) polyurethanes. Analysis of the exudates surrounding the materials revealed no differences between surfaces in the types or levels of cells present. Conversely, the proportion of adherent cells undergoing apoptosis, as determined by annexin V-FITC staining, increased significantly on anionic followed by zwitterionic surfaces (60 + 5.0 and 38 + 3.7%) when compared with the base. Additionally, zwitterionic and anionic substrates provided decreased rates of macrophage adhesion and fusion into FBGCs, whereas cationic surfaces promoted macrophage adhesion and FBGC formation. Visualization of the F-actin cytoskeleton by Alexa Fluor 488 phalloidin showed a significant delay in the cytoskeletal fusion response on zwitterionic and the anionic surfaces. The real-time polymerase chain reaction (PCR) analysis of proinflammatory cytokines (tumor necrosis factor (TNF)-α and interleukin (IL)-10) and pro-wound healing cytokines (IL-4 and TGF-β) revealed differential cytokine responses. Cationic substrates that triggered stimulation of TNF-α and IL-4 were associated with more spread cells and higher FBGCs, whereas zwitterionic and anionic substrates that suppressed these cytokines levels were associated with less spread cells and few FBGCs. These studies have revealed that zwitterionic and anionic polyurethane surface chemistries can not only reduce nonspecific adhesion, fusion, and inflammatory events but also effectively promote cellular apoptosis in vivo.
Collapse
Affiliation(s)
- Anand Khandwekar
- Department of Bioengineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.
| | | |
Collapse
|
37
|
Balansky R, D'Agostini F, Micale RT, La Maestra S, Steele VE, De Flora S. Dose-related cytogenetic damage in pulmonary alveolar macrophages from mice exposed to cigarette smoke early in life. Arch Toxicol 2012; 86:509-16. [PMID: 21989788 DOI: 10.1007/s00204-011-0765-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 09/29/2011] [Indexed: 12/01/2022]
Abstract
The micronucleus test detects both structural and numerical chromosomal aberrations caused by environmental agents. However, this test is poorly sensitive to detect the clastogenicity of cigarette smoke (CS) in human peripheral blood lymphocytes. At variance with peripheral blood lymphocytes and other cells outside the lower respiratory tract, pulmonary alveolar macrophages (PAM) are selectively affected by inhalable carcinogens and have been used to evaluate the modulation of CS-related cytogenetic alterations in vivo. The present study was aimed at evaluating (1) the cytogenetic response in PAM isolated from the lung of mice exposed to CS during the first 4 weeks of life and (2) the dose dependence of MN and polynucleated (PN) PAM formation in CS-exposed mice. To this purpose, ICR(CD-1) mice were exposed whole body to mainstream CS for 4 weeks, starting immediately after birth. Bronchoalveolar lavage (BAL) was performed to evaluate the cellularity of this fluid and the frequency of PN and MN PAM. At the doses of 119, 292, and 438 mg/m(3) total particulate matter, CS significantly increased both the proportion of PAM in the BAL fluid and the frequencies of PN and MN PAM. The cytogenetic effects were significantly correlated with the CS dose. In conclusion, PAM are suitable to detect induction by CS of clastogenic and aneugenic effects in mice during a developmental period corresponding to infancy, childhood, and early adolescence in humans. These surrogate cells, providing an important defense mechanism of the respiratory tract, are proposed as indicators of CS-related DNA damage in youngsters.
Collapse
Affiliation(s)
- Roumen Balansky
- National Center of Oncology, Str. Plovdivsko Pole 6, 1756 Sofia, Bulgaria.
| | | | | | | | | | | |
Collapse
|
38
|
Nascimento MPPD, Bannwart CF, Nakaira-Takahagi E, Peraçoli MTS. Granulocyte macrophage colony-stimulating factor enhances the modulatory effect of cytokines on monocyte-derived multinucleated giant cell formation and fungicidal activity against Paracoccidioides brasiliensis. Mem Inst Oswaldo Cruz 2011; 106:735-41. [PMID: 22012229 DOI: 10.1590/s0074-02762011000600014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 08/09/2011] [Indexed: 11/22/2022] Open
Abstract
Multinucleated giant cells (MGC) are cells present in characteristic granulomatous inflammation induced by intracellular infectious agents or foreign materials. The present study evaluated the modulatory effect of granulocyte macrophage colony-stimulating factor (GM-CSF) in association with other cytokines such as interferon-gamma (IFN-γ), tumour necrosis factor-alpha, interleukin (IL)-10 or transforming growth factor beta (TGF-β1) on the formation of MGC from human peripheral blood monocytes stimulated with Paracoccidioides brasiliensis antigen (PbAg). The generation of MGC was determined by fusion index (FI) and the fungicidal activity of these cells was evaluated after 4 h of MGC co-cultured with viable yeast cells of P. brasiliensis strain 18 (Pb18). The results showed that monocytes incubated with PbAg and GM-CSF plus IFN-γ had a significantly higher FI than in all the other cultures, while the addition of IL-10 or TGF-β1 had a suppressive effect on MGC generation. Monocytes incubated with both pro and anti-inflammatory cytokines had a higher induction of foreign body-type MGC rather than Langhans-type MGC. MGC stimulated with PbAg and GM-CSF in association with the other cytokines had increased fungicidal activity and the presence of GM-CSF also partially inhibited the suppressive effects of IL-10 and TGF-β1. Together, these results suggest that GM-CSF is a positive modulator of PbAg-stimulated MGC generation and on the fungicidal activity against Pb18.
Collapse
Affiliation(s)
- Magda Paula Pereira do Nascimento
- Departamento de Doenças Tropicais, Faculdade de Medicina, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brasil
| | | | | | | |
Collapse
|
39
|
Yu M, Qi X, Moreno JL, Farber DL, Keegan AD. NF-κB signaling participates in both RANKL- and IL-4-induced macrophage fusion: receptor cross-talk leads to alterations in NF-κB pathways. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:1797-806. [PMID: 21734075 PMCID: PMC3150418 DOI: 10.4049/jimmunol.1002628] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
NF-κB activation is essential for receptor activator for NF-κB ligand (RANKL)-induced osteoclast formation. IL-4 is known to inhibit the RANKL-induced osteoclast differentiation while at the same time promoting macrophage fusion to form multinucleated giant cells (MNG). Several groups have proposed that IL-4 inhibition of osteoclastogenesis is mediated by suppressing the RANKL-induced activation of NF-κB. However, we found that IL-4 did not block proximal, canonical NF-κB signaling. Instead, we found that IL-4 inhibited alternative NF-κB signaling and induced p105/50 expression. Interestingly, in nfκb1(-/-) bone marrow-derived macrophages (BMM), the formation of both multinucleated osteoclast and MNG induced by RANKL or IL-4, respectively, was impaired. This suggests that NF-κB signaling also plays an important role in IL-4-induced macrophage fusion. Indeed, we found that the RANKL-induced and IL-4-induced macrophage fusion were both inhibited by the NF-κB inhibitors IκB kinase 2 inhibitor and NF-κB essential modulator inhibitory peptide. Furthermore, overexpression of p50, p65, p52, and RelB individually in nfκb1(-/-) or nfκb1(+/+) BMM enhanced both giant osteoclast and MNG formation. Interestingly, knockdown of nfκb2 in wild-type BMM dramatically enhanced both osteoclast and MNG formation. In addition, both RANKL- and IL-4-induced macrophage fusion were impaired in NF-κB-inducing kinase(-/-) BMM. These results suggest IL-4 influences NF-κB pathways by increasing p105/p50 and suppressing RANKL-induced p52 translocation and that NF-κB pathways participate in both RANKL- and IL-4-induced giant cell formation.
Collapse
Affiliation(s)
- Minjun Yu
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201
- Columbia Center for Translational Immunology, Columbia University, New York, NY 10032
| | - Xiulan Qi
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Jose L. Moreno
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201
- Food and Drug Administration, Besthesda, MD, 20014
| | - Donna L. Farber
- Columbia Center for Translational Immunology, Columbia University, New York, NY 10032
| | - Achsah D. Keegan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201
- Program in Oncology, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
40
|
Molecular and cellular mechanisms of mammalian cell fusion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 713:33-64. [PMID: 21432013 DOI: 10.1007/978-94-007-0763-4_4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The fusion of one cell with another occurs in development, injury and disease. Despite the diversity of fusion events, five steps in sequence appear common. These steps include programming fusion-competent status, chemotaxis, membrane adhesion, membrane fusion, and post-fusion resetting. Recent advances in the field start to reveal the molecules involved in each step. This review focuses on some key molecules and cellular events of cell fusion in mammals. Increasing evidence demonstrates that membrane lipid rafts, adhesion proteins and actin rearrangement are critical in the final step of membrane fusion. Here we propose a new model for the formation and expansion of membrane fusion pores based on recent observations on myotube formation. In this model, membrane lipid rafts first recruit adhesion molecules and align with opposing membranes, with the help of a cortical actin "wall" as a rigid supportive platform. Second, the membrane adhesion proteins interact with each other and trigger actin rearrangement, which leads to rapid dispersion of lipid rafts and flow of a highly fluidic phospholipid bilayer into the site. Finally, the opposing phospholipid bilayers are then pushed into direct contact leading to the formation of fusion pores by the force generated through actin polymerization. The actin polymerization generated force also drives the expansion of the fusion pores. However, several key questions about the process of cell fusion still remain to be explored. The understanding of the mechanisms of cell fusion may provide new opportunities in correcting development disorders or regenerating damaged tissues by inhibiting or promoting molecular events associated with fusion.
Collapse
|
41
|
Estrella JL, Kan-Sutton C, Gong X, Rajagopalan M, Lewis DE, Hunter RL, Eissa NT, Jagannath C. A Novel in vitro Human Macrophage Model to Study the Persistence of Mycobacterium tuberculosis Using Vitamin D(3) and Retinoic Acid Activated THP-1 Macrophages. Front Microbiol 2011; 2:67. [PMID: 21747789 PMCID: PMC3128978 DOI: 10.3389/fmicb.2011.00067] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Accepted: 03/25/2011] [Indexed: 11/13/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) replicates within the human macrophages and we investigated the activating effects of retinoic acid (RA) and vitamin D(3) (VD) on macrophages in relation to the viability of intracellular Mtb. A combination of these vitamins (RAVD) enhanced the levels of DC-SIGN and mannose receptors on THP-1 macrophages that increased mycobacterial uptake but inhibited the subsequent intracellular growth of Mtb by inducing reactive oxygen species and autophagy. RAVD also enhanced antigen presenting and chemotactic receptors on THPs suggesting an activated phenotype for RAVD activated THPs. RAVD mediated activation was also associated with a marked phenotypic change in Mtb infected THPs that fused with adjacent THPs to form multinucleated giant cells (MNGCs). Typically, MNGCs occurred over 30 days of in vitro culture and contained non-replicating persisting Mtb for more than 60 days in culture. Latent tuberculosis occurs in over a third of mankind and we propose that RAVD mediated induction of persistent Mtb within human macrophages provides a novel model to develop therapeutic approaches and investigate pathogenesis of latency.
Collapse
Affiliation(s)
- Jaymie L. Estrella
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences CenterHouston, TX, USA
| | - Celestine Kan-Sutton
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences CenterHouston, TX, USA
| | - Xing Gong
- Department of Internal Medicine, Baylor College of MedicineHouston, TX, USA
| | - Malini Rajagopalan
- Department of Biochemistry, University of Texas Health Sciences CenterTyler, TX, USA
| | - Dorothy E. Lewis
- Department of Internal Medicine and Infectious Disease, University of Texas Health Sciences CenterHouston, TX, USA
| | - Robert L. Hunter
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences CenterHouston, TX, USA
| | - N. Tony Eissa
- Department of Internal Medicine, Baylor College of MedicineHouston, TX, USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences CenterHouston, TX, USA
| |
Collapse
|
42
|
Macrophage fusion and multinucleated giant cells of inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 713:97-111. [PMID: 21432016 DOI: 10.1007/978-94-007-0763-4_7] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Macrophages undergo fusion with other macrophages to form the hallmark multinucleated giant cells of chronic inflammation. However, neither the existence of distinct morphological types of giant cells, the signaling pathways that induce their formation, the molecular mechanism(s) of macrophage fusion, nor the significance of macrophage multinucleation at chronic inflammatory sites are well understood. Our efforts have been focused on these unknowns, particularly as they relate to the foreign body-type giant cells that form on implanted biomaterials and biomedical devices. We have pursued the discoveries of human macrophage fusion factors (interleukin-4, interleukin-13, α-tocopherol) with emphasis on foreign body giant cells, and identified adhesion receptors and signaling intermediates, as well as an adhesion protein substrate (vitronectin) that supports macrophage fusion. Studies on the molecular mechanism of macrophage fusion have revealed it to be a mannose receptor-mediated phagocytic process with participation of the endoplasmic reticulum. Further phenotypic and functional investigations will foster new perspectives on these remarkable multinucleated cells and their physiological significances in multiple inflammatory processes.
Collapse
|
43
|
Wang HW, Joyce JA. Alternative activation of tumor-associated macrophages by IL-4: priming for protumoral functions. Cell Cycle 2010; 9:4824-35. [PMID: 21150330 PMCID: PMC3047808 DOI: 10.4161/cc.9.24.14322] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2010] [Accepted: 11/30/2010] [Indexed: 01/21/2023] Open
Abstract
Although macrophages were originally recognized as major immune effector cells, it is now appreciated that they also play many important roles in the maintenance of tissue homeostasis, and are involved in a variety of pathological conditions including cancer. Several studies have demonstrated the contributions of tumor-associated macrophages (TAMs) to tumor initiation, progression, and metastasis. However, the detailed mechanisms underlying how TAMs differ molecularly from their normal counterparts and how the conversion to TAMs occurs have only just begun to be understood. TAMs have been proposed to exhibit phenotypes of 'alternatively activated' macrophages, though there has been limited evidence directly linking the phenotypes of TAMs to the alternative activation of macrophages. This review will focus on IL-4, the prototypic cytokine that induces the alternative activation of macrophages, and review current knowledge regarding the contributions of IL-4 to the phenotypes of TAMs and its effects on tumorigenesis.
Collapse
Affiliation(s)
- Hao-Wei Wang
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, Cornell University, New York, NY, USA
| | | |
Collapse
|
44
|
Freisinger E, Cramer C, Xia X, Murthy SN, Slakey DP, Chiu E, Newsome ER, Alt EU, Izadpanah R. Characterization of hematopoietic potential of mesenchymal stem cells. J Cell Physiol 2010; 225:888-97. [DOI: 10.1002/jcp.22299] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
45
|
Lyons JA, Haring JS, Biga PR. Myostatin expression, lymphocyte population, and potential cytokine production correlate with predisposition to high-fat diet induced obesity in mice. PLoS One 2010; 5:e12928. [PMID: 20877574 PMCID: PMC2943928 DOI: 10.1371/journal.pone.0012928] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Accepted: 08/27/2010] [Indexed: 12/21/2022] Open
Abstract
A strong relationship exists between increased inflammatory cytokines and muscle insulin resistance in obesity. This study focused on identifying a relationship between metabolic propensity and myostatin expression in muscle and spleen cells in response to high-fat diet intake. Using a comparative approach, we analyzed the effects of high-fat diet intake on myostatin and follistatin expression, spleen cell composition, and potential cytokine expression in high-fat diet induced obesity (HFDIO) resistant (SWR/J) and susceptible (C57BL/6) mice models. Results demonstrated overall increased myostatin expression in muscle following high-fat diet intake in HFDIO-susceptible mice, while myostatin expression levels decreased initially in muscle from high-fat diet fed resistant mice. In HFDIO-resistant mice, myostatin expression decreased in spleen, while myostatin increased in spleen tissue from HFDIO-susceptible mice. Proinflammatory cytokine (IL-17, IL-1β, and IFNγ) potential increased in splenocytes from HFDIO-susceptible mice. In comparison, C57BL/6 mice fed a high-fat diet exhibited higher frequencies of CD4+/CD44hi and CD8+/CD44hi cells in the spleen compared to control fed mice. Together, these results suggest that susceptibility to high-fat diet induced obesity could be influenced by local myostatin activity in a tissue-specific manner and that splenocytes exhibit differential cytokine production in a strain-dependent manner. This study sets the stage for future investigations into the interactions between growth, inflammation, and metabolism.
Collapse
Affiliation(s)
- Jeri-Anne Lyons
- Department of Clinical Health Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, United States of America
| | - Jodie S. Haring
- Department of Chemistry and Molecular Biology, North Dakota State University, Fargo, North Dakota, United States of America
| | - Peggy R. Biga
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota, United States of America
- * E-mail:
| |
Collapse
|
46
|
Shankar SP, Babensee JE. Comparative characterization of cultures of primary human macrophages or dendritic cells relevant to biomaterial studies. J Biomed Mater Res A 2010; 92:791-800. [PMID: 19274715 DOI: 10.1002/jbm.a.32406] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Macrophages are central mediators of biomaterial-associated wound healing. Dendritic cells (DCs) link innate and adaptive immunity and are important in the context of the host response to combination products. Starting with human peripheral blood mononuclear cells (PBMCs), DCs were derived from monocytes upon culture with granulocyte macrophage colony-stimulating factor and interleukin-4; macrophages were derived from monocytes upon culture without cytokines. Macrophage or DC cultures were characterized at relevant timepoints in both adherent and nonadherent fractions on control Primaria surfaces to characterize and define these inflammatory/immune cells as a prequel to their use in in vitro test biomaterial-host response studies. At day 10 (typical time for harvesting macrophages for subsequent treatment with test biomaterials), macrophages were CD11c+, macrophage mannose receptor (MMR)+, CD14+, and CD64+. At day 6 (typical time for harvesting of DCs after 24-h treatment with test biomaterials), DCs were CD1c+, CD11c+, CD123+, MMR+, CD14+, and CD64-. Furthermore, CD3+ and CD4+ T lymphocytes and CD19+ and CD24+ B lymphocytes were present in both cultures at all timepoints, although to different extents. Immature DCs (approximately 15 microm), were rounded but presented extensive dendritic processes upon maturation with lipopolysaccharide. Alternatively, adherent macrophages (approximately 15-20 microm) displayed internalized lipids and exhibited few membrane processes. The characterization and comparison of existing techniques to establish reliable, reproducible primary cultures of DCs or macrophages provides an important basis for examining and interpreting complex macrophage/DC-lymphocyte-orchestrated host responses in future studies with equivalent cell populations on test biomaterials.
Collapse
Affiliation(s)
- Sucharita P Shankar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive Atlanta, Georgia 30332, USA
| | | |
Collapse
|
47
|
Yasui K, Yashiro M, Nagaoka Y, Manki A, Wada T, Tsuge M, Kondo Y, Morishima T. Thalidomide prevents formation of multinucleated giant cells (Langhans-type cells) from cultured monocytes: possible pharmaceutical applications for granulomatous disorders. Int J Immunopathol Pharmacol 2009; 22:707-14. [PMID: 19822087 DOI: 10.1177/039463200902200316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Thalidomide is an effective drug for chronic inflammatory diseases, but the mechanism underlying its immunomodulatory action remains uncertain. Thalidomide has been reported to clinically improve chronic inflammatory granulomatous disorders. In such disorders, the granulomas consist of epithelioid cells, scattered lymphocytes and multinucleated giant cells (MNGC; Langhans-type cells). The present experimental approach permitted the reproduction of MNGC formation from peripheral blood monocytes and examination of thalidomides effect on it. MNGC can be effectively generated from monocytes cultured in the presence of interleukin-4 (IL-4) and macrophage colony-stimulating factor(M-CSF) for 14 days. Thalidomide can inhibit the formation of MNGC in a dose-dependent manner. MNGC formation was partly inhibited by the presence of neutralizing TNF-alpha antibody in the responses induced by IL-4 and M-CSF. Autocrinal TNF-alpha production and modulation of cadhelin expression to regulate cell adhesion might be involved in this inhibitory action of thalidomide. Our results support thalidomides clinical efficacy in the treatment of chronic granulomatous disorders (granulomatosis).
Collapse
Affiliation(s)
- K Yasui
- Department of Pediatrics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Varin A, Gordon S. Alternative activation of macrophages: immune function and cellular biology. Immunobiology 2009; 214:630-41. [PMID: 19264378 DOI: 10.1016/j.imbio.2008.11.009] [Citation(s) in RCA: 273] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Accepted: 11/14/2008] [Indexed: 12/19/2022]
Abstract
Macrophages are the first line of defense of the organism against pathogens and, in response to the microenvironment, become differentially activated. In the presence of IL-4 and IL-13, cytokines that are produced in a Th-2 type response, particularly during allergy and parasitic infections, macrophages become differentially activated. Alternative activated macrophages play an important role in the protection of the host by decreasing inflammation and promoting tissues repair. However, alternative activation of macrophages also downregulates host protection against selected pathogens. This defect is associated with an altered receptor expression pattern and extensive modulation of intracellular membrane trafficking. This review shows how alternative activation of macrophages induces extensive cellular remodelling of phagocytic, endocytic, signaling and secretory pathways which play an important, but unclear role in the pathogenesis of different disease.
Collapse
Affiliation(s)
- Audrey Varin
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | | |
Collapse
|
49
|
do Nascimento MPP, de Campos Soares AMV, Dias-Melicio LA, Parise-Fortes MR, Martins RAR, Nakaira ET, Peraçoli MTS. Fungicidal activity of human monocyte-derived multinucleated giant cells induced in vitro by Paracoccidioides brasiliensis antigen. Mycopathologia 2008; 166:25-33. [DOI: 10.1007/s11046-007-9051-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Accepted: 08/14/2007] [Indexed: 10/22/2022]
|
50
|
Nuss KM, von Rechenberg B. Biocompatibility issues with modern implants in bone - a review for clinical orthopedics. Open Orthop J 2008; 2:66-78. [PMID: 19506701 PMCID: PMC2687115 DOI: 10.2174/1874325000802010066] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2008] [Revised: 03/13/2008] [Accepted: 04/07/2008] [Indexed: 11/22/2022] Open
Abstract
Skeletal defects may result from traumatic, infectious, congenital or neoplastic processes and are considered to be a challenge for reconstructive surgery. Although the autologous bone graft is still the "gold standard", there is continuing demand for bone substitutes because of associated disadvantages, such as limited supply and potential donor side morbidity [1]. This is not only true for indications in orthopedic and craniomaxillofacial surgeries, but also in repairing endodontic defects and in dental implantology.Before clinical use all new bone substitute materials have to be validated for their osseoconductive and - depending on the composition of the material also -inductive ability, as well as for their long-term biocompatibility in bone. Serving this purpose various bone healing models to test osteocompatibility and inflammatory potential of a novel material on one hand and, on the other hand, non-healing osseous defects to assess the healing potential of a bone substitute material have been developed. Sometimes the use of more than one implantation site can be helpful to provide a wide range of information about a new material [2].Important markers for biocompatibility and inflammatory responses are the cell types appearing after the implantation of foreign material. There, especially the role of foreign body giant cells (FBGC) is discussed controversial in the pertinent literature, such that it is not clear whether their presence marks an incompatibility of the biomaterial, or whether it belongs to a normal degradation behavior of modern, resorbable biomaterials.This publication is highlighting the different views currently existing about the function of FBGC that appear in response to biomaterials at the implantation sites. A short overview of the general classes of biomaterials, where FBGC may appear as cellular response, is added for clarity, but may not be complete.
Collapse
Affiliation(s)
| | - Brigitte von Rechenberg
- Address correspondence to this author at the The Musculoskeletal Research Unit (MSRU), Equine Department, Vetsuisse Faculty ZH, University of Zürich, Winterthurerstr. 260, 8057 Zürich, Switzerland; Tel: +41-44-635 8410; Fax: +41-44-635 8917; E-mail:
| |
Collapse
|