1
|
Bessoles S, Chiron A, Sarrabayrouse G, De La Grange P, Abina AM, Hacein-Bey-Abina S. Erythropoietin induces tumour progression and CD39 expression on immune cells in a preclinical model of triple-negative breast cancer. Immunology 2024; 173:360-380. [PMID: 38953295 DOI: 10.1111/imm.13832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/14/2024] [Indexed: 07/04/2024] Open
Abstract
The adverse effects observed in some cancer patients treated with erythropoiesis-stimulating agents such as erythropoietin (EPO) might be due to the latter's well-known immunosuppressive functions. Here, we used a mouse model of syngeneic triple-negative breast cancer to explore EPO's immunomodulatory role in a tumour setting. Our results showed that EPO treatment promotes tumour growth, exacerbates the 'immune desert', and results in a 'cold tumour'. EPO treatment changed the immune cell distribution in peripheral blood, secondary lymphoid organs, and the tumour microenvironment (TME). Our in-depth analysis showed that EPO mainly impacts CD4 T cells by accelerating their activation in the spleen and thus their subsequent exhaustion in the TME. This process is accompanied by a general elevation of CD39 expression by several immune cells (notably CD4 T cells in the tumour and spleen), which promotes an immunosuppressive TME. Lastly, we identified a highly immunosuppressive CD39+ regulatory T cell population (ICOS+, CTLA4+, Ki67+) as a potential biomarker of the risk of EPO-induced tumour progression. EPO displays pleiotropic immunosuppressive functions and enhances mammary tumour progression in mice.
Collapse
Affiliation(s)
- Stéphanie Bessoles
- Université Paris Cité, CNRS, INSERM, UTCBS, Unité des Technologies Chimiques et Biologiques pour la Santé, Paris, France
| | - Andrada Chiron
- Université Paris Cité, CNRS, INSERM, UTCBS, Unité des Technologies Chimiques et Biologiques pour la Santé, Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| | - Guillaume Sarrabayrouse
- Université Paris Cité, CNRS, INSERM, UTCBS, Unité des Technologies Chimiques et Biologiques pour la Santé, Paris, France
| | | | - Amine M Abina
- Université Paris Cité, CNRS, INSERM, UTCBS, Unité des Technologies Chimiques et Biologiques pour la Santé, Paris, France
| | - Salima Hacein-Bey-Abina
- Université Paris Cité, CNRS, INSERM, UTCBS, Unité des Technologies Chimiques et Biologiques pour la Santé, Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| |
Collapse
|
2
|
Wu L, Zhang Y, Zhao F, Sheng J, Gu N. Magnetic Characterization of Human Intrinsically Magnetic Monocytes Through a Novel Optical Tracking-Based Magnetic Sensor. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307306. [PMID: 38312110 DOI: 10.1002/smll.202307306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/17/2024] [Indexed: 02/06/2024]
Abstract
Intrinsically magnetic cells naturally occur within organisms and are believed to be linked to iron metabolism and certain cellular functions while the functional significance of this magnetism is largely unexplored. To better understand this property, an approach named Optical Tracking-based Magnetic Sensor (OTMS) has been developed. This multi-target tracking system is designed to measure the magnetic moment of individual cells. The OTMS generates a tunable magnetic field and induces movement in magnetic cells that are subsequently analyzed through a learning-based tracking-by-detection system. The magnetic moment of numerous cells can be calculated simultaneously, thereby providing a quantitative tool to assess cellular magnetic properties within populations. Upon deploying the OTMS, a stable population of magnetic cells in human peripheral monocytes is discovered. Further application in the analysis of clinical blood samples reveals an intriguing pattern: the proportion of magnetic monocytes differs significantly between systemic lupus erythematosus (SLE) patients and healthy volunteers. This variation is positively correlated with disease activity, a trend not observed in patients with rheumatoid arthritis (RA). The study, therefore, presents a new frontier in the investigation of the magnetic characteristics of naturally occurring magnetic cells, opening the door to potential diagnostic and therapeutic applications that leverage cellular magnetism.
Collapse
Affiliation(s)
- Linyuan Wu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Engineering, Southeast University, Nanjing, 210009, China
| | - Yuxin Zhang
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Engineering, Southeast University, Nanjing, 210009, China
| | - Fengfeng Zhao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jingyi Sheng
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Engineering, Southeast University, Nanjing, 210009, China
| | - Ning Gu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Engineering, Southeast University, Nanjing, 210009, China
- Medical School, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
3
|
Lafuente-Gómez N, de Lázaro I, Dhanjani M, García-Soriano D, Sobral MC, Salas G, Mooney DJ, Somoza Á. Multifunctional magnetic nanoparticles elicit anti-tumor immunity in a mouse melanoma model. Mater Today Bio 2023; 23:100817. [PMID: 37822453 PMCID: PMC10562177 DOI: 10.1016/j.mtbio.2023.100817] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/13/2023] Open
Abstract
Immunotherapy has emerged as a promising strategy to eradicate cancer cells. Particularly, the development of cancer vaccines to induce a potent and sustained antigen-specific T cell response has become a center of attention. Herein, we describe a novel immunotherapy based on magnetic nanoparticles (MNP) covalently modified with the OVA254-267 antigen and a CpG oligonucleotide via disulfide bonds. The MNP-CpG-COVA significantly enhances dendritic cell activation and CD8+ T cell antitumoral response against B16-OVA melanoma cells in vitro. Notably, the immune response induced by the covalently modified MNP is more potent and sustained over time than that triggered by the free components, highlighting the advantage of nanoformulations in immunotherapies. What is more, the nanoparticles are stable in the blood after in vivo administration and induce potent levels of systemic tumor-specific effector CD8 + T cells. Overall, our findings highlight the potential of covalently functionalized MNP to induce robust immune responses against mouse melanoma.
Collapse
Affiliation(s)
- Nuria Lafuente-Gómez
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Immunology Service, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa, Madrid, 28006, Spain
| | - Irene de Lázaro
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Department of Biomedical Engineering, NYU Tandon School of Engineering, New York University, New York, NY, 10010, USA
- NYU Cardiovascular Research Center, Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York University, New York, NY, 10010, USA
| | - Mónica Dhanjani
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
| | - David García-Soriano
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
| | - Miguel C. Sobral
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Gorka Salas
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
- Unidad de Nanobiotecnología Asociada al Centro Nacional de Biotecnología (CNB-CSIC), Madrid, 28049, Spain
| | - David J. Mooney
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid, 28049, Spain
- Unidad de Nanobiotecnología Asociada al Centro Nacional de Biotecnología (CNB-CSIC), Madrid, 28049, Spain
| |
Collapse
|
4
|
Kajiyama S, Nagatake T, Ishikawa S, Hosomi K, Shimada Y, Matsui Y, Kunisawa J. Lentinula Edodes Mycelia extract regulates the function of antigen-presenting cells to activate immune cells and prevent tumor-induced deterioration of immune function. BMC Complement Med Ther 2023; 23:281. [PMID: 37553633 PMCID: PMC10408224 DOI: 10.1186/s12906-023-04106-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 07/27/2023] [Indexed: 08/10/2023] Open
Abstract
Immune cell activation is essential for cancer rejection; however, the tumor microenvironment leads to deterioration of immune function, which enables cancer cells to survive and proliferate. We previously reported that oral ingestion of Lentinula Edodes Mycelia (L.E.M.) extract enhances the tumor antigen-specific T-cell response and exerts an antitumor effect in a tumor-bearing mouse model. In this study, we focused on antigen-presenting cells (APCs) located upstream of the immune system, induced a T-cell response, then examined the impact of L.E.M. extract on the APCs. L.E.M. extract enhanced the expression of MHC-I, MHC-II, CD86, CD80, and CD40 in bone marrow-derived dendritic cells (DCs) and strongly induced the production of IL-12. L.E.M.-stimulated DCs enhanced IFN-γ production from CD8+ T cells and induced their differentiation into effector cells. Furthermore, L.E.M. extract promoted IL-12 production and suppressed the production of IL-10 and TGF-β by transforming bone marrow-derived macrophages into M1-like macrophages. Furthermore, in a B16F10 melanoma inoculation model, DCs in the spleen were decreased and their activation was suppressed by the presence of cancer; however, ingestion of L.E.M. extract prevented this functional deterioration of DCs. In the spleen of cancer-bearing mice, the number of CD11b- F4/80+ macrophages in a hypoactivated state was also increased, whereas L.E.M. extract suppressed the increase of such macrophages. These findings suggest that L.E.M. extract may exhibit an antitumor immune response by regulating the function of APCs to induce cytotoxic T lymphocytes, as well as by suppressing the decline in antigen-presenting cell activity caused by the presence of cancer.
Collapse
Affiliation(s)
- Shota Kajiyama
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki-city, Osaka, 567-0085, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Central R & D Laboratory, Kobayashi Pharmaceutical Co., Ltd, Ibaragi, Osaka, Japan
| | - Takahiro Nagatake
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki-city, Osaka, 567-0085, Japan
- Laboratory of Functional Anatomy, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa, Japan
| | - Satoru Ishikawa
- Central R & D Laboratory, Kobayashi Pharmaceutical Co., Ltd, Ibaragi, Osaka, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki-city, Osaka, 567-0085, Japan
| | - Yuki Shimada
- Central R & D Laboratory, Kobayashi Pharmaceutical Co., Ltd, Ibaragi, Osaka, Japan
| | - Yasunori Matsui
- Central R & D Laboratory, Kobayashi Pharmaceutical Co., Ltd, Ibaragi, Osaka, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki-city, Osaka, 567-0085, Japan.
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.
- Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Kobe, Japan.
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Graduate School of Medicine, Graduate School of Dentistry, Graduate School of Science, Osaka University, Suita, Japan.
| |
Collapse
|
5
|
Slusarczyk P, Mandal PK, Zurawska G, Niklewicz M, Chouhan K, Mahadeva R, Jończy A, Macias M, Szybinska A, Cybulska-Lubak M, Krawczyk O, Herman S, Mikula M, Serwa R, Lenartowicz M, Pokrzywa W, Mleczko-Sanecka K. Impaired iron recycling from erythrocytes is an early hallmark of aging. eLife 2023; 12:79196. [PMID: 36719185 PMCID: PMC9931393 DOI: 10.7554/elife.79196] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 01/30/2023] [Indexed: 02/01/2023] Open
Abstract
Aging affects iron homeostasis, as evidenced by tissue iron loading and anemia in the elderly. Iron needs in mammals are met primarily by iron recycling from senescent red blood cells (RBCs), a task chiefly accomplished by splenic red pulp macrophages (RPMs) via erythrophagocytosis. Given that RPMs continuously process iron, their cellular functions might be susceptible to age-dependent decline, a possibility that has been unexplored to date. Here, we found that 10- to 11-month-old female mice exhibit iron loading in RPMs, largely attributable to a drop in iron exporter ferroportin, which diminishes their erythrophagocytosis capacity and lysosomal activity. Furthermore, we identified a loss of RPMs during aging, underlain by the combination of proteotoxic stress and iron-dependent cell death resembling ferroptosis. These impairments lead to the retention of senescent hemolytic RBCs in the spleen, and the formation of undegradable iron- and heme-rich extracellular protein aggregates, likely derived from ferroptotic RPMs. We further found that feeding mice an iron-reduced diet alleviates iron accumulation in RPMs, enhances their ability to clear erythrocytes, and reduces damage. Consequently, this diet ameliorates hemolysis of splenic RBCs and reduces the burden of protein aggregates, mildly increasing serum iron availability in aging mice. Taken together, we identified RPM collapse as an early hallmark of aging and demonstrated that dietary iron reduction improves iron turnover efficacy.
Collapse
Affiliation(s)
- Patryk Slusarczyk
- International Institute of Molecular and Cell Biology in WarsawWarsawPoland
| | | | - Gabriela Zurawska
- International Institute of Molecular and Cell Biology in WarsawWarsawPoland
| | - Marta Niklewicz
- International Institute of Molecular and Cell Biology in WarsawWarsawPoland
| | - Komal Chouhan
- International Institute of Molecular and Cell Biology in WarsawWarsawPoland
| | | | - Aneta Jończy
- International Institute of Molecular and Cell Biology in WarsawWarsawPoland
| | - Matylda Macias
- International Institute of Molecular and Cell Biology in WarsawWarsawPoland
| | | | | | - Olga Krawczyk
- Maria Sklodowska-Curie National Research Institute of OncologyWarsawPoland
| | - Sylwia Herman
- Laboratory of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian UniversityCracowPoland
| | - Michal Mikula
- Maria Sklodowska-Curie National Research Institute of OncologyWarsawPoland
| | - Remigiusz Serwa
- IMol Polish Academy of SciencesWarsawPoland
- ReMedy International Research Agenda Unit, IMol Polish Academy of SciencesWarsawPoland
| | - Małgorzata Lenartowicz
- Laboratory of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian UniversityCracowPoland
| | - Wojciech Pokrzywa
- International Institute of Molecular and Cell Biology in WarsawWarsawPoland
| | | |
Collapse
|
6
|
Long-Term Clearance and Biodistribution of Magnetic Nanoparticles Assessed by AC Biosusceptometry. MATERIALS 2022; 15:ma15062121. [PMID: 35329574 PMCID: PMC8948936 DOI: 10.3390/ma15062121] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/08/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022]
Abstract
Once administered in an organism, the physiological parameters of magnetic nanoparticles (MNPs) must be addressed, as well as their possible interactions and retention and elimination profiles. Alternating current biosusceptometry (ACB) is a biomagnetic detection system used to detect and quantify MNPs. The aims of this study were to evaluate the biodistribution and clearance of MNPs profiles through long-time in vivo analysis and determine the elimination time carried out by the association between the ACB system and MnFe2O4 nanoparticles. The liver, lung, spleen, kidneys, and heart and a blood sample were collected for biodistribution analysis and, for elimination analysis, and over 60 days. During the period analyzed, the animal’s feces were also collectedd. It was possible to notice a higher uptake by the liver and the spleen due to their characteristics of retention and uptake. In 60 days, we observed an absence of MNPs in the spleen and a significant decay in the liver. We also determined the MNPs’ half-life through the liver and the spleen elimination. The data indicated a concentration decay profile over the 60 days, which suggests that, in addition to elimination via feces, there is an endogenous mechanism of metabolization or possible agglomeration of MNPs, resulting in loss of ACB signal intensity.
Collapse
|
7
|
Gas K, Sawicki M. In Situ Compensation Method for Precise Integral SQUID Magnetometry of Miniscule Biological, Chemical, and Powder Specimens Requiring the Use of Capsules. MATERIALS 2022; 15:ma15020495. [PMID: 35057212 PMCID: PMC8780521 DOI: 10.3390/ma15020495] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 01/25/2023]
Abstract
Steadily growing interest in magnetic characterization of organic compounds for therapeutic purposes or of other irregularly shaped specimens calls for refinements of experimental methodology to satisfy experimental challenges. Encapsulation in capsules remains the method of choice, but its applicability in precise magnetometry is limited. This is particularly true for minute specimens in the single milligram range as they are outweighed by the capsules and are subject to large alignment errors. We present here a completely new experimental methodology that permits 30-fold in situ reduction of the signal of capsules by substantially restoring the symmetry of the sample holder that is otherwise broken by the presence of the capsule. In practical terms it means that the standard 30 mg capsule is seen by the magnetometer as approximately a 1 mg object, effectively opening the window for precise magnetometry of single milligram specimens. The method is shown to work down to 1.8 K and in the whole range of the magnetic fields. The method is demonstrated and validated using the reciprocal space option of MPMS-SQUID magnetometers; however, it can be easily incorporated in any magnetometer that can accommodate straw sample holders (i.e., the VSM-SQUID). Importantly, the improved sensitivity is accomplished relying only on the standard accessories and data reduction method provided by the SQUID manufacturer, eliminating the need for elaborate raw data manipulations.
Collapse
|
8
|
Splenic red pulp macrophages provide a niche for CML stem cells and induce therapy resistance. Leukemia 2022; 36:2634-2646. [PMID: 36163264 PMCID: PMC7613762 DOI: 10.1038/s41375-022-01682-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 08/03/2022] [Accepted: 08/09/2022] [Indexed: 11/10/2022]
Abstract
Disease progression and relapse of chronic myeloid leukemia (CML) are caused by therapy resistant leukemia stem cells (LSCs), and cure relies on their eradication. The microenvironment in the bone marrow (BM) is known to contribute to LSC maintenance and resistance. Although leukemic infiltration of the spleen is a hallmark of CML, it is unknown whether spleen cells form a niche that maintains LSCs. Here, we demonstrate that LSCs preferentially accumulate in the spleen and contribute to disease progression. Spleen LSCs were located in the red pulp close to red pulp macrophages (RPM) in CML patients and in a murine CML model. Pharmacologic and genetic depletion of RPM reduced LSCs and decreased their cell cycling activity in the spleen. Gene expression analysis revealed enriched stemness and decreased myeloid lineage differentiation in spleen leukemic stem and progenitor cells (LSPCs). These results demonstrate that splenic RPM form a niche that maintains CML LSCs in a quiescent state, resulting in disease progression and resistance to therapy.
Collapse
|
9
|
Thomann S, Weiler SME, Wei T, Sticht C, De La Torre C, Tóth M, Rose F, Tang Y, Ritz T, Ball C, Glimm H, Ryschich E, Schirmacher P, Breuhahn K. YAP-induced Ccl2 expression is associated with a switch in hepatic macrophage identity and vascular remodelling in liver cancer. Liver Int 2021; 41:3011-3023. [PMID: 34459091 DOI: 10.1111/liv.15048] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 07/11/2021] [Accepted: 08/25/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIM The development of hepatocellular carcinoma (HCC) is associated with the formation of communication networks leading to the recruitment of disease-modifying macrophages. However, how oncogenes in tumour cells control paracrine communication is not fully understood. METHODS Transgenic mice with liver-specific expression of the constitutively active yes-associated protein (YAPS127A ) or an orthotopic implantation model served as tumour models. FACS-sorted F4/80+ /CD11bdim /CD146- /retinoid- macrophages from healthy and tumour-bearing livers were used for transcriptomic profiling. Expression data of 242 human HCCs and a tissue microarray consisting of 91 HCCs and seven liver tissues were analyzed. RESULTS Screening of primary tumour cells expressing YAPS127A identified CC chemokine ligand 2 (Ccl2) as a macrophage chemoattractant, whose expression was regulated in a YAP/TEA domain family member 4 (TEAD4)-dependent manner. Ccl2 expression was associated with a loss of Kupffer cells (KCs) and an increase in immature macrophages (Mɸimm ) in hepatocarcinogenesis. Recruited Mɸimm were characterized by a lack of functional polarization (M0 signature) and high expression of the Ccl2 receptors C-C motif chemokine receptor 2 (Ccr2), C-X3-C motif chemokine receptor 1 (Cx3cr1) and pro-angiogenic platelet-derived growth factors (Pdgfa/Pdgfb). Mɸimm formed cellular clusters in the perivascular space, which correlated with vascular morphometric changes indicative for angiogenesis. In human HCCs, the M0 signature served as an identifier for poor clinical outcome and CCL2 correlated with YAP expression and vascular network formation. CONCLUSIONS In conclusion, YAP/TEAD4-regulated Ccl2 associates with perivascular recruitment of unpolarized Mɸimm and may contribute to a proangiogenic microenvironment in liver cancer.
Collapse
Affiliation(s)
- Stefan Thomann
- Institute of Pathology, University Hospital Heidelberg, Germany.,Institute of Systems Immunology, University of Würzburg, Germany
| | | | - Teng Wei
- Institute of Pathology, University Hospital Heidelberg, Germany.,Cytotherapy Laboratory, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, Guangdong, China
| | - Carsten Sticht
- Next Generation Sequencing Core Facility, Medical Faculty Mannheim, Germany
| | | | - Marcell Tóth
- Institute of Pathology, University Hospital Heidelberg, Germany
| | - Fabian Rose
- Institute of Pathology, University Hospital Heidelberg, Germany
| | - Yingyue Tang
- Institute of Pathology, University Hospital Heidelberg, Germany
| | - Thomas Ritz
- Institute of Pathology, University Hospital Heidelberg, Germany
| | - Claudia Ball
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Hanno Glimm
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Translational Functional Cancer Genomics, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Personalized Oncology, University Hospital Carl Gustav Carus TU Dresden, Dresden, Germany.,German Consortium for Translational Cancer Research (DKTK), Dresden, Germany
| | - Eduard Ryschich
- Department of Surgery, University Hospital Heidelberg, Germany
| | | | - Kai Breuhahn
- Institute of Pathology, University Hospital Heidelberg, Germany
| |
Collapse
|
10
|
Shim YA, Weliwitigoda A, Campbell T, Dosanjh M, Johnson P. Splenic erythroid progenitors decrease TNF-α production by macrophages and reduce systemic inflammation in a mouse model of T cell-induced colitis. Eur J Immunol 2020; 51:567-579. [PMID: 33180325 DOI: 10.1002/eji.202048687] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 09/13/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022]
Abstract
In inflammatory bowel disease (IBD), inflammation can occur beyond the intestine and spread systemically causing complications such as arthritis, cachexia, and anemia. Here, we determine the impact of CD45, a pan-leukocyte marker and tyrosine phosphatase, on IBD. Using a mouse model of T cell transfer colitis, CD25- CD45RBhigh CD4+ T cells were transferred into Rag1-deficient mice (RAGKO) and CD45-deficient RAGKO mice (CD45RAGKO). Weight loss and systemic wasting syndrome were delayed in CD45RAGKO mice compared to RAGKO mice, despite equivalent inflammation in the colon. CD45RAGKO mice had reduced serum levels of TNF-α, and reduced TNF-α production by splenic myeloid cells. CD45RAGKO mice also had increased numbers of erythroid progenitors in the spleen, which had previously been shown to be immunosuppressive. Adoptive transfer of these erythroid progenitors into RAGKO mice reduced their weight loss and TNF-α expression by splenic red pulp macrophages. In vitro, erythroid cells suppressed TNF-α expression in red pulp macrophages in a phagocytosis-dependent manner. These findings show a novel role for erythroid progenitors in suppressing the pro-inflammatory function of splenic macrophages and cachexia associated with IBD.
Collapse
Affiliation(s)
- Yaein Amy Shim
- Department of Microbiology and Immunology, University of British Columbia, British Columbia, Canada
| | - Asanga Weliwitigoda
- Department of Microbiology and Immunology, University of British Columbia, British Columbia, Canada.,Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Teresa Campbell
- Department of Microbiology and Immunology, University of British Columbia, British Columbia, Canada.,Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Manisha Dosanjh
- Department of Microbiology and Immunology, University of British Columbia, British Columbia, Canada
| | - Pauline Johnson
- Department of Microbiology and Immunology, University of British Columbia, British Columbia, Canada
| |
Collapse
|
11
|
Newman L, Jasim DA, Prestat E, Lozano N, de Lazaro I, Nam Y, Assas BM, Pennock J, Haigh SJ, Bussy C, Kostarelos K. Splenic Capture and In Vivo Intracellular Biodegradation of Biological-Grade Graphene Oxide Sheets. ACS NANO 2020; 14:10168-10186. [PMID: 32658456 PMCID: PMC7458483 DOI: 10.1021/acsnano.0c03438] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/13/2020] [Indexed: 05/20/2023]
Abstract
Carbon nanomaterials, including 2D graphene-based materials, have shown promising applicability to drug delivery, tissue engineering, diagnostics, and various other biomedical areas. However, to exploit the benefits of these materials in some of the areas mentioned, it is necessary to understand their possible toxicological implications and long-term fate in vivo. We previously demonstrated that following intravenous administration, 2D graphene oxide (GO) nanosheets were largely excreted via the kidneys; however, a small but significant portion of the material was sequestered in the spleen. Herein, we interrogate the potential consequences of this accumulation and the fate of the spleen-residing GO over a period of nine months. We show that our thoroughly characterized GO materials are not associated with any detectable pathological consequences in the spleen. Using confocal Raman mapping of tissue sections, we determine the sub-organ biodistribution of GO at various time points after administration. The cells largely responsible for taking up the material are confirmed using immunohistochemistry coupled with Raman spectroscopy, and transmission electron microscopy (TEM). This combination of techniques identified cells of the splenic marginal zone as the main site of GO bioaccumulation. In addition, through analyses using both bright-field TEM coupled with electron diffraction and Raman spectroscopy, we reveal direct evidence of in vivo intracellular biodegradation of GO sheets with ultrastructural precision. This work offers critical information about biological processing and degradation of thin GO sheets by normal mammalian tissue, indicating that further development and exploitation of GO in biomedicine would be possible.
Collapse
Affiliation(s)
- Leon Newman
- Nanomedicine
Lab, National Graphene Institute and Faculty of Biology, Medicine
& Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Dhifaf A. Jasim
- Nanomedicine
Lab, National Graphene Institute and Faculty of Biology, Medicine
& Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Eric Prestat
- Department
of Materials, School of Natural Sciences, The University of Manchester, Manchester, M13 9PL, United Kingdom
| | - Neus Lozano
- Nanomedicine
Lab, National Graphene Institute and Faculty of Biology, Medicine
& Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), Barcelona, 08193, Spain
| | - Irene de Lazaro
- Nanomedicine
Lab, National Graphene Institute and Faculty of Biology, Medicine
& Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Yein Nam
- Nanomedicine
Lab, National Graphene Institute and Faculty of Biology, Medicine
& Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Bakri M. Assas
- Lydia
Becker Institute of Immunology and Inflammation, and Division of Infection,
Immunity and Respiratory Medicine, School of Biological Sciences,
Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Department
of Immunology, Faculty of Applied Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Joanne Pennock
- Lydia
Becker Institute of Immunology and Inflammation, and Division of Infection,
Immunity and Respiratory Medicine, School of Biological Sciences,
Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Sarah J. Haigh
- Department
of Materials, School of Natural Sciences, The University of Manchester, Manchester, M13 9PL, United Kingdom
| | - Cyrill Bussy
- Nanomedicine
Lab, National Graphene Institute and Faculty of Biology, Medicine
& Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Kostas Kostarelos
- Nanomedicine
Lab, National Graphene Institute and Faculty of Biology, Medicine
& Health, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), Barcelona, 08193, Spain
| |
Collapse
|
12
|
Humphries J, Pizzi D, Sonderegger SE, Fletcher NL, Houston ZH, Bell CA, Kempe K, Thurecht KJ. Hyperbranched Poly(2-oxazoline)s and Poly(ethylene glycol): A Structure–Activity Comparison of Biodistribution. Biomacromolecules 2020; 21:3318-3331. [DOI: 10.1021/acs.biomac.0c00765] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- James Humphries
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - David Pizzi
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology; Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Stefan E. Sonderegger
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Nicholas L. Fletcher
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Zachary H. Houston
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Craig A. Bell
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Kristian Kempe
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology; Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Kristofer J. Thurecht
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
13
|
Nagala M, Crocker PR. Towards understanding the cell surface phenotype, metabolic properties and immune functions of resident macrophages of the peritoneal cavity and splenic red pulp using high resolution quantitative proteomics. Wellcome Open Res 2020. [DOI: 10.12688/wellcomeopenres.16061.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background:Resident macrophages (Mϕs) are distributed throughout the body and are important for maintaining tissue homeostasis and for defence against infections. Tissue Mϕs are highly adapted to their microenvironment and thought to mediate tissue-specific functions involving metabolism and immune defence that are not fully elucidated. Methods:We have used high resolution quantitative proteomics to gain insights into the functions of two types of resident tissue Mϕs: peritoneal cavity Mϕs and splenic red pulp Mϕs. The cellular expression levels of many proteins were validated by flow cytometry and were consistently in agreement with the proteomics data.Results:Peritoneal and splenic red pulp macrophages displayed major differences in cell surface phenotype reflecting their adaptation to different tissue microenvironments and tissue-specific functions. Peritoneal Mϕs were shown to be enriched in a number of key enzymes and metabolic pathways normally associated with the liver, such as metabolism of fructose, detoxification, nitrogen homeostasis and the urea cycle. Supporting these observations, we show that peritoneal Mϕs are able to utilise glutamine and glutamate which are rich in peritoneum for urea generation. In comparison, splenic red pulp Mϕs were enriched in proteins important for adaptive immunity such as antigen presenting MHC molecules, in addition to proteins required for erythrocyte homeostasis and iron turnover. We also show that these tissue Mϕs may utilise carbon and nitrogen substrates for different metabolic fates to support distinct tissue-specific roles.Conclusions:This study provides new insights into the functions of tissue Mϕs in immunity and homeostasis. The comprehensive proteomics data sets are a valuable resource for biologists and immunologists.
Collapse
|
14
|
Enders M, Franken L, Philipp MS, Kessler N, Baumgart AK, Eichler M, Wiertz EJH, Garbi N, Kurts C. Splenic Red Pulp Macrophages Cross-Prime Early Effector CTL That Provide Rapid Defense against Viral Infections. THE JOURNAL OF IMMUNOLOGY 2019; 204:87-100. [PMID: 31776205 DOI: 10.4049/jimmunol.1900021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 10/29/2019] [Indexed: 12/13/2022]
Abstract
Cross-presentation allows dendritic cells (DCs) to present peptides derived from endocytosed Ags on MHC class I molecules, which is important for activating CTL against viral infections and tumors. Type 1 classical DCs (cDC1), which depend on the transcription factor Batf3, are considered the main cross-presenting cells. In this study, we report that soluble Ags are efficiently cross-presented also by transcription factor SpiC-dependent red pulp macrophages (RPM) of the spleen. In contrast to cDC1, RPM used the mannose receptor for Ag uptake and employed the proteasome- and TAP-dependent cytosolic cross-presentation pathway, previously shown to be used in vitro by bone marrow-derived DCs. In an in vivo vaccination model, both cDC1 and RPM cross-primed CTL efficiently but with distinct kinetics. Within a few days, RPM induced very early effector CTL of a distinct phenotype (Ly6A/E+ Ly6C(+) KLRG1- CD127- CX3CR1- Grz-B+). In an adenoviral infection model, such CTL contained the early viral spread, whereas cDC1 induced short-lived effector CTL that eventually cleared the virus. RPM-induced early effector CTL also contributed to the endogenous antiviral response but not to CTL memory generation. In conclusion, RPM can contribute to antiviral immunity by generating a rapid CTL defense force that contains the virus until cDC1-induced CTL are available to eliminate it. This function can be harnessed for improving vaccination strategies aimed at inducing CTL.
Collapse
Affiliation(s)
- Marika Enders
- Institut für Experimentelle Immunologie, Rheinische Friedrich-Wilhelms-Universität, 53127 Bonn, Germany; and
| | - Lars Franken
- Institut für Experimentelle Immunologie, Rheinische Friedrich-Wilhelms-Universität, 53127 Bonn, Germany; and
| | - Marie-Sophie Philipp
- Institut für Experimentelle Immunologie, Rheinische Friedrich-Wilhelms-Universität, 53127 Bonn, Germany; and
| | - Nina Kessler
- Institut für Experimentelle Immunologie, Rheinische Friedrich-Wilhelms-Universität, 53127 Bonn, Germany; and
| | - Ann-Kathrin Baumgart
- Institut für Experimentelle Immunologie, Rheinische Friedrich-Wilhelms-Universität, 53127 Bonn, Germany; and
| | - Melanie Eichler
- Institut für Experimentelle Immunologie, Rheinische Friedrich-Wilhelms-Universität, 53127 Bonn, Germany; and
| | - Emmanuel J H Wiertz
- Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Natalio Garbi
- Institut für Experimentelle Immunologie, Rheinische Friedrich-Wilhelms-Universität, 53127 Bonn, Germany; and
| | - Christian Kurts
- Institut für Experimentelle Immunologie, Rheinische Friedrich-Wilhelms-Universität, 53127 Bonn, Germany; and
| |
Collapse
|
15
|
Pek RH, Yuan X, Rietzschel N, Zhang J, Jackson L, Nishibori E, Ribeiro A, Simmons W, Jagadeesh J, Sugimoto H, Alam MZ, Garrett L, Haldar M, Ralle M, Phillips JD, Bodine DM, Hamza I. Hemozoin produced by mammals confers heme tolerance. eLife 2019; 8:e49503. [PMID: 31571584 PMCID: PMC6773446 DOI: 10.7554/elife.49503] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/24/2019] [Indexed: 12/28/2022] Open
Abstract
Free heme is cytotoxic as exemplified by hemolytic diseases and genetic deficiencies in heme recycling and detoxifying pathways. Thus, intracellular accumulation of heme has not been observed in mammalian cells to date. Here we show that mice deficient for the heme transporter SLC48A1 (also known as HRG1) accumulate over ten-fold excess heme in reticuloendothelial macrophage lysosomes that are 10 to 100 times larger than normal. Macrophages tolerate these high concentrations of heme by crystallizing them into hemozoin, which heretofore has only been found in blood-feeding organisms. SLC48A1 deficiency results in impaired erythroid maturation and an inability to systemically respond to iron deficiency. Complete heme tolerance requires a fully-operational heme degradation pathway as haplo insufficiency of HMOX1 combined with SLC48A1 inactivation causes perinatal lethality demonstrating synthetic lethal interactions between heme transport and degradation. Our studies establish the formation of hemozoin by mammals as a previously unsuspected heme tolerance pathway.
Collapse
Affiliation(s)
- Rini H Pek
- Department of Animal and Avian SciencesUniversity of MarylandCollege ParkUnited States
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkUnited States
| | - Xiaojing Yuan
- Department of Animal and Avian SciencesUniversity of MarylandCollege ParkUnited States
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkUnited States
| | - Nicole Rietzschel
- Department of Animal and Avian SciencesUniversity of MarylandCollege ParkUnited States
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkUnited States
| | - Jianbing Zhang
- Department of Animal and Avian SciencesUniversity of MarylandCollege ParkUnited States
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkUnited States
| | - Laurie Jackson
- Department of MedicineUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - Eiji Nishibori
- Faculty of Pure and Applied SciencesUniversity of TsukubaTsukubaJapan
- Tsukuba Research Center for Energy Materials ScienceUniversity of TsukabaTsukabaJapan
| | - Ana Ribeiro
- Department of Animal and Avian SciencesUniversity of MarylandCollege ParkUnited States
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkUnited States
| | - William Simmons
- Genetics and Molecular Biology BranchNational Human Genome Research Institute, National Institutes of HealthBethesdaUnited States
| | - Jaya Jagadeesh
- Genetics and Molecular Biology BranchNational Human Genome Research Institute, National Institutes of HealthBethesdaUnited States
| | | | - Md Zahidul Alam
- Department of Pathology and Laboratory MedicinePerelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Lisa Garrett
- NHGRI Embryonic Stem Cell and Transgenic Mouse CoreNational Human Genome Research Institute, National Institutes of HealthBethesdaUnited States
| | - Malay Haldar
- Department of Pathology and Laboratory MedicinePerelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Martina Ralle
- Department of Molecular and Medical GeneticsOregon Health and Science UniversityPortlandUnited States
| | - John D Phillips
- Department of MedicineUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - David M Bodine
- Genetics and Molecular Biology BranchNational Human Genome Research Institute, National Institutes of HealthBethesdaUnited States
| | - Iqbal Hamza
- Department of Animal and Avian SciencesUniversity of MarylandCollege ParkUnited States
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkUnited States
| |
Collapse
|
16
|
Indomethacin reduces rates of aortic dissection and rupture of the abdominal aorta by inhibiting monocyte/macrophage accumulation in a murine model. Sci Rep 2019; 9:10751. [PMID: 31341173 PMCID: PMC6656736 DOI: 10.1038/s41598-019-46673-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 07/01/2019] [Indexed: 12/16/2022] Open
Abstract
Aortic dissection is a life-threatening condition, which is characterised by separation of the constituent layers of the aortic wall. We have recently shown that monocyte/macrophage infiltration into the aortic wall is a pathogenic mechanism of the condition. In the present study, we investigated whether the anti-inflammatory agent, indomethacin, could inhibit monocyte/macrophage accumulation in the aortic wall and ensuing dissection. Indomethacin was administered (from 3 days prior with daily oral administration) to mice in which aortic dissection was induced using beta-aminopropionitrile (BAPN) and angiotensin II (Ang II) infusion (2 weeks). Indomethacin prevented death from abdominal aortic dissection and decreased incidence of aortic dissection by as high as 40%. Histological and flow cytometry analyses showed that indomethacin administration resulted in inhibition of monocyte transendothelial migration and monocyte/macrophage accumulation in the aortic wall. These results indicate that indomethacin administration reduces rate of onset of aortic dissection in a murine model of the condition.
Collapse
|
17
|
Sulaiman A, McGarry S, El-Sahli S, Li L, Chambers J, Phan A, Côté M, Cron GO, Alain T, Le Y, Lee SH, Liu S, Figeys D, Gadde S, Wang L. Co-targeting Bulk Tumor and CSCs in Clinically Translatable TNBC Patient-Derived Xenografts via Combination Nanotherapy. Mol Cancer Ther 2019; 18:1755-1764. [DOI: 10.1158/1535-7163.mct-18-0873] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 01/18/2019] [Accepted: 07/08/2019] [Indexed: 11/16/2022]
|
18
|
McLeod JS, Church JT, Coughlin MA, Carr B, Poling C, Sarosi E, Perkins EM, Quinones MC, Hala P, Rabah R, Freiheit E, Rojas-Pena A, Bartlett RH, Mychaliska GB. Splenic development and injury in premature lambs supported by the artificial placenta. J Pediatr Surg 2019; 54:1147-1152. [PMID: 30902457 PMCID: PMC6545267 DOI: 10.1016/j.jpedsurg.2019.02.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 02/21/2019] [Indexed: 11/30/2022]
Abstract
INTRODUCTION The purpose of this study is to evaluate splenic effects during artificial placenta (AP) support. METHODS AP lambs (118-121 d, n = 14) were delivered and placed on the AP support for a goal of 10-14 days. Cannulation used right jugular drainage and umbilical vein reinfusion. Early (ETC; 115-120 d; n = 7) and late (LTC; 125-131 d; n = 7) tissue controls were delivered and immediately sacrificed. Spleens were formalin fixed, H&E stained, and graded for injury, response to inflammation, and extramedullary hematopoiesis (EMH). CD68 and CD163 stains were used to assess for macrophage activation and density. Clinical variables were correlated with splenic scores. Groups were compared using Fisher's Exact Test and descriptive statistics. p < 0.05 indicated significance. RESULTS Mean survival for AP lambs was 12 ± 5 d. There was no necrosis found in any of the groups. Vascular congestion and sinusoidal histiocytosis did not significantly differ between AP and control groups (p = 0.72; p = 0.311). There were significantly more pigmented macrophages (p = 0.008), CD163 (p = <0.001), and CD68 (p = <0.001) stained cells in the AP group. ETC and LTC demonstrated more EMH than AP spleens (p = <0.001). CONCLUSIONS During AP support, spleens appear to develop normally and exhibit an appropriate inflammatory response. After initiation of AP support, EMH transitions away from the spleen. STUDY TYPE Research Paper/Therapeutic Potential. LEVEL OF EVIDENCE N/A.
Collapse
Affiliation(s)
- Jennifer S. McLeod
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, Michigan
| | - Joseph T. Church
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, Michigan
| | - Megan A. Coughlin
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, Michigan
| | - Benjamin Carr
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, Michigan
| | - Clinton Poling
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, Michigan
| | - Ellery Sarosi
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, Michigan
| | | | - Matias Caceres Quinones
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, Michigan
| | - Pavel Hala
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, Michigan
| | - Raja Rabah
- Department of Statistics, Michigan Medicine, Ann Arbor, Michigan
| | | | - Alvaro Rojas-Pena
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, Michigan
| | - Robert H Bartlett
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, Michigan
| | - George B. Mychaliska
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, Michigan,Fetal Diagnosis and Treatment Center, C.S. Mott Children’s Hospital, Michigan Medicine, Ann Arbor, Michigan
| |
Collapse
|
19
|
In vivo evaluation of thiol-functionalized superparamagnetic iron oxide nanoparticles. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 99:171-179. [DOI: 10.1016/j.msec.2019.01.118] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 01/22/2019] [Accepted: 01/23/2019] [Indexed: 11/21/2022]
|
20
|
Efremova MV, Naumenko VA, Spasova M, Garanina AS, Abakumov MA, Blokhina AD, Melnikov PA, Prelovskaya AO, Heidelmann M, Li ZA, Ma Z, Shchetinin IV, Golovin YI, Kireev II, Savchenko AG, Chekhonin VP, Klyachko NL, Farle M, Majouga AG, Wiedwald U. Magnetite-Gold nanohybrids as ideal all-in-one platforms for theranostics. Sci Rep 2018; 8:11295. [PMID: 30050080 PMCID: PMC6062557 DOI: 10.1038/s41598-018-29618-w] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 07/16/2018] [Indexed: 12/21/2022] Open
Abstract
High-quality, 25 nm octahedral-shaped Fe3O4 magnetite nanocrystals are epitaxially grown on 9 nm Au seed nanoparticles using a modified wet-chemical synthesis. These Fe3O4-Au Janus nanoparticles exhibit bulk-like magnetic properties. Due to their high magnetization and octahedral shape, the hybrids show superior in vitro and in vivo T2 relaxivity for magnetic resonance imaging as compared to other types of Fe3O4-Au hybrids and commercial contrast agents. The nanoparticles provide two functional surfaces for theranostic applications. For the first time, Fe3O4-Au hybrids are conjugated with two fluorescent dyes or the combination of drug and dye allowing the simultaneous tracking of the nanoparticle vehicle and the drug cargo in vitro and in vivo. The delivery to tumors and payload release are demonstrated in real time by intravital microscopy. Replacing the dyes by cell-specific molecules and drugs makes the Fe3O4-Au hybrids a unique all-in-one platform for theranostics.
Collapse
Affiliation(s)
- Maria V Efremova
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation
| | - Victor A Naumenko
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation
| | - Marina Spasova
- Faculty of Physics and Center for Nanointegration Duisburg-Essen, University of Duisburg-Essen, Duisburg, 47057, Germany
| | - Anastasiia S Garanina
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation
| | - Maxim A Abakumov
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation
- Department of Medical Nanobiotechnology, Russian National Research Medical University, Moscow, 117997, Russian Federation
| | - Anastasia D Blokhina
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Pavel A Melnikov
- Department of Fundamental and Applied Neurobiology, Serbsky National Medical Research Center for Psychiatry and Narcology, Ministry of Health and Social Development of the Russian Federation, Moscow, 119034, Russian Federation
| | | | - Markus Heidelmann
- ICAN - Interdisciplinary Center for Analytics on the Nanoscale and Center for Nanointegration Duisburg-Essen, University of Duisburg-Essen, Duisburg, 47057, Germany
| | - Zi-An Li
- Faculty of Physics and Center for Nanointegration Duisburg-Essen, University of Duisburg-Essen, Duisburg, 47057, Germany
| | - Zheng Ma
- Faculty of Physics and Center for Nanointegration Duisburg-Essen, University of Duisburg-Essen, Duisburg, 47057, Germany
| | - Igor V Shchetinin
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation
| | - Yuri I Golovin
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
- Derzhavin Tambov State University, Nanocenter, Tambov, 392000, Russian Federation
| | - Igor I Kireev
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Alexander G Savchenko
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation
| | - Vladimir P Chekhonin
- Department of Medical Nanobiotechnology, Russian National Research Medical University, Moscow, 117997, Russian Federation
- Department of Fundamental and Applied Neurobiology, Serbsky National Medical Research Center for Psychiatry and Narcology, Ministry of Health and Social Development of the Russian Federation, Moscow, 119034, Russian Federation
| | - Natalia L Klyachko
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation
| | - Michael Farle
- Faculty of Physics and Center for Nanointegration Duisburg-Essen, University of Duisburg-Essen, Duisburg, 47057, Germany
| | - Alexander G Majouga
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russian Federation.
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation.
- D. Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russian Federation.
| | - Ulf Wiedwald
- National University of Science and Technology «MISIS», Moscow, 119049, Russian Federation.
- Faculty of Physics and Center for Nanointegration Duisburg-Essen, University of Duisburg-Essen, Duisburg, 47057, Germany.
| |
Collapse
|
21
|
Sant S, Grzelak L, Wang Z, Pizzolla A, Koutsakos M, Crowe J, Loudovaris T, Mannering SI, Westall GP, Wakim LM, Rossjohn J, Gras S, Richards M, Xu J, Thomas PG, Loh L, Nguyen THO, Kedzierska K. Single-Cell Approach to Influenza-Specific CD8 + T Cell Receptor Repertoires Across Different Age Groups, Tissues, and Following Influenza Virus Infection. Front Immunol 2018; 9:1453. [PMID: 29997621 PMCID: PMC6030351 DOI: 10.3389/fimmu.2018.01453] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/12/2018] [Indexed: 11/13/2022] Open
Abstract
CD8+ T cells recognizing antigenic peptides derived from conserved internal viral proteins confer broad protection against distinct influenza viruses. As memory CD8+ T cells change throughout the human lifetime and across tissue compartments, we investigated how T cell receptor (TCR) composition and diversity relate to memory CD8+ T cells across anatomical sites and immunological phases of human life. We used ex vivo peptide-HLA tetramer magnetic enrichment, single-cell multiplex RT-PCR for both the TCR-alpha (TCRα) and TCR-beta (TCRβ) chains, and new TCRdist and grouping of lymphocyte interactions by paratope hotspots (GLIPH) algorithms to compare TCRs directed against the most prominent human influenza epitope, HLA-A*02:01-M158–66 (A2+M158). We dissected memory TCR repertoires directed toward A2+M158 CD8+ T cells within human tissues and compared them to human peripheral blood of young and elderly adults. Furthermore, we compared these memory CD8+ T cell repertoires to A2+M158 CD8+ TCRs during acute influenza disease in patients hospitalized with avian A/H7N9 virus. Our study provides the first ex vivo comparative analysis of paired antigen-specific TCR-α/β clonotypes across different tissues and peripheral blood across different age groups. We show that human A2+M158 CD8+ T cells can be readily detected in human lungs, spleens, and lymph nodes, and that tissue A2+M158 TCRαβ repertoires reflect A2+M158 TCRαβ clonotypes derived from peripheral blood in healthy adults and influenza-infected patients. A2+M158 TCRαβ repertoires displayed distinct features only in elderly adults, with large private TCRαβ clonotypes replacing the prominent and public TRBV19/TRAV27 TCRs. Our study provides novel findings on influenza-specific TCRαβ repertoires within human tissues, raises the question of how we can prevent the loss of optimal TCRαβ signatures with aging, and provides important insights into the rational design of T cell-mediated vaccines and immunotherapies.
Collapse
Affiliation(s)
- Sneha Sant
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Ludivine Grzelak
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,École Normale Supérieure Paris-Saclay, Université Paris-Saclay, Cachan, France
| | - Zhongfang Wang
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Angela Pizzolla
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Marios Koutsakos
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Jane Crowe
- Deepdene Surgery, Deepdene, VIC, Australia
| | - Thomas Loudovaris
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Stuart I Mannering
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Glen P Westall
- Lung Transplant Unit, Alfred Hospital, Melbourne, VIC, Australia
| | - Linda M Wakim
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, Australia.,School of Medicine, Institute of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Stephanie Gras
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, Australia
| | - Michael Richards
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Jianqing Xu
- Shanghai Public Health Clinical Center, Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Paul G Thomas
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, United States
| | - Liyen Loh
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
22
|
Li Y, Lou C, Wang W. STIM1 deficiency protects the liver from ischemia/reperfusion injury in mice. Biochem Biophys Res Commun 2018; 496:422-428. [PMID: 29305862 DOI: 10.1016/j.bbrc.2018.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 01/02/2018] [Indexed: 02/07/2023]
Abstract
Hepatic ischemia reperfusion (I/R) injury is unavoidable in various clinical conditions. Despite considerable investigation, the underlying molecular mechanism revealing liver I/R injury remains elusive. Stromal interaction molecule 1 (STIM1) plays essential role in regulating the induction of cellular responses to a number of stress conditions, including temperature changes, elevated ROS, and hypoxia. Here, to explore if STIM1 is involved in hepatic injury, wild type (WT) and STIM1-knockout (STIM1-/-) mice were subjected to I/R. Our results indicated that the WT mice with hepatic I/R injury showed higher STIM1 expressions from gene and protein levels in liver tissue samples. Similar results were observed in hypoxia-exposed cells in vitro. Significantly, STIM1-/- attenuated hepatic injury compared to the WT mice after I/R, as evidenced by the improved pathological alterations in liver sections. WT mice subjected to liver I/R showed higher serum alanine aminotransferase (ALT) and aminotransferase (AST) levels, as well as pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α), interleukin (IL)-6 and IL-1β, which were significantly reduced by STIM1-/-. In addition, STIM1-/- also decreased the liver mRNA levels of pro-inflammatory cytokines in mice after I/R injury. Furthermore, significantly decreased oxidative stress was found in STIM1-/- mice after I/R injury compared to the WT group of mice, evidenced by the enhanced superoxide dismutase (SOD) activity and the reduced malondialdehyde (MDA) and reactive oxygen species (ROS) levels in liver tissue samples. Moreover, STIM1-/- mice with hepatic I/R injury displayed the down-regulated nuclear factor of activated T cell (NFAT1), Orai1 and cleaved Caspase-3 levels in liver, contributing to apoptosis suppression. The results above were confirmed in hypoxia-treated cells lacking of STIM1 expression. Together, the findings suggested that STIM1-deletion protects the liver from I/R injury in mice through inhibiting inflammation, oxidative stress and apoptosis. STIM1 could be considered as a potential therapeutic target to ameliorate I/R injury.
Collapse
Affiliation(s)
- Yanyang Li
- Department of Pediatrics, Huaihe Hospital, Henan University, Kaifeng 475000, China.
| | - Chunyan Lou
- Department of Pediatrics, Huaihe Hospital, Henan University, Kaifeng 475000, China
| | - Weiying Wang
- Department of Pediatrics, Huaihe Hospital, Henan University, Kaifeng 475000, China
| |
Collapse
|
23
|
Hirayama D, Iida T, Nakase H. The Phagocytic Function of Macrophage-Enforcing Innate Immunity and Tissue Homeostasis. Int J Mol Sci 2017; 19:E92. [PMID: 29286292 PMCID: PMC5796042 DOI: 10.3390/ijms19010092] [Citation(s) in RCA: 488] [Impact Index Per Article: 69.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 12/19/2017] [Accepted: 12/27/2017] [Indexed: 12/20/2022] Open
Abstract
Macrophages are effector cells of the innate immune system that phagocytose bacteria and secrete both pro-inflammatory and antimicrobial mediators. In addition, macrophages play an important role in eliminating diseased and damaged cells through their programmed cell death. Generally, macrophages ingest and degrade dead cells, debris, tumor cells, and foreign materials. They promote homeostasis by responding to internal and external changes within the body, not only as phagocytes, but also through trophic, regulatory, and repair functions. Recent studies demonstrated that macrophages differentiate from hematopoietic stem cell-derived monocytes and embryonic yolk sac macrophages. The latter mainly give rise to tissue macrophages. Macrophages exist in all vertebrate tissues and have dual functions in host protection and tissue injury, which are maintained at a fine balance. Tissue macrophages have heterogeneous phenotypes in different tissue environments. In this review, we focused on the phagocytic function of macrophage-enforcing innate immunity and tissue homeostasis for a better understanding of the role of tissue macrophages in several pathological conditions.
Collapse
Affiliation(s)
- Daisuke Hirayama
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Minami 1-jo Nishi 16-chome, Chuo-ku, Sapporo, Hokkaido 060-8543, Japan.
| | - Tomoya Iida
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Minami 1-jo Nishi 16-chome, Chuo-ku, Sapporo, Hokkaido 060-8543, Japan.
| | - Hiroshi Nakase
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Minami 1-jo Nishi 16-chome, Chuo-ku, Sapporo, Hokkaido 060-8543, Japan.
| |
Collapse
|
24
|
Costa da Silva M, Breckwoldt MO, Vinchi F, Correia MP, Stojanovic A, Thielmann CM, Meister M, Muley T, Warth A, Platten M, Hentze MW, Cerwenka A, Muckenthaler MU. Iron Induces Anti-tumor Activity in Tumor-Associated Macrophages. Front Immunol 2017; 8:1479. [PMID: 29167669 PMCID: PMC5682327 DOI: 10.3389/fimmu.2017.01479] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/23/2017] [Indexed: 12/13/2022] Open
Abstract
Tumor-associated macrophages (TAMs) frequently help to sustain tumor growth and mediate immune suppression in the tumor microenvironment (TME). Here, we identified a subset of iron-loaded, pro-inflammatory TAMs localized in hemorrhagic areas of the TME. The occurrence of iron-loaded TAMs (iTAMs) correlated with reduced tumor size in patients with non-small cell lung cancer. Ex vivo experiments established that TAMs exposed to hemolytic red blood cells (RBCs) were converted into pro-inflammatory macrophages capable of directly killing tumor cells. This anti-tumor effect could also be elicited via iron oxide nanoparticles. When tested in vivo, tumors injected with such iron oxide nanoparticles led to significantly smaller tumor sizes compared to controls. These results identify hemolytic RBCs and iron as novel players in the TME that repolarize TAMs to exert direct anti-tumor effector function. Thus, the delivery of iron to TAMs emerges as a simple adjuvant therapeutic strategy to promote anti-cancer immune responses.
Collapse
Affiliation(s)
- Milene Costa da Silva
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), Heidelberg University, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.,Graduate Program in Areas of Basic and Applied Biology (GABBA), Abel Salazar Biomedical Sciences Institute (ICBAS), University of Porto, Porto, Portugal.,Innate Immunity Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Michael O Breckwoldt
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany.,German Cancer Consortium, Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Francesca Vinchi
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), Heidelberg University, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Margareta P Correia
- Innate Immunity Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ana Stojanovic
- Innate Immunity Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carl Maximilian Thielmann
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), Heidelberg University, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Michael Meister
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany.,Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Muley
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany.,Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Arne Warth
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany.,Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Michael Platten
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany.,German Cancer Consortium, Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias W Hentze
- Molecular Medicine Partnership Unit (MMPU), Heidelberg University, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Adelheid Cerwenka
- Innate Immunity Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Immunbiochemistry, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Martina U Muckenthaler
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), Heidelberg University, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
25
|
Mowat AM, Scott CL, Bain CC. Barrier-tissue macrophages: functional adaptation to environmental challenges. Nat Med 2017; 23:1258-1270. [PMID: 29117177 DOI: 10.1038/nm.4430] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 09/29/2017] [Indexed: 12/12/2022]
Abstract
Macrophages are found throughout the body, where they have crucial roles in tissue development, homeostasis and remodeling, as well as being sentinels of the innate immune system that can contribute to protective immunity and inflammation. Barrier tissues, such as the intestine, lung, skin and liver, are exposed constantly to the outside world, which places special demands on resident cell populations such as macrophages. Here we review the mounting evidence that although macrophages in different barrier tissues may be derived from distinct progenitors, their highly specific properties are shaped by the local environment, which allows them to adapt precisely to the needs of their anatomical niche. We discuss the properties of macrophages in steady-state barrier tissues, outline the factors that shape their differentiation and behavior and describe how macrophages change during protective immunity and inflammation.
Collapse
Affiliation(s)
- Allan McI Mowat
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow, UK
| | - Charlotte L Scott
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow, UK
- Laboratory of Myeloid Cell Ontogeny and Functional Specialization, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Calum C Bain
- The University of Edinburgh/MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
26
|
Audemard-Verger A, Rivière M, Durand A, Peranzoni E, Guichard V, Hamon P, Bonilla N, Guilbert T, Boissonnas A, Auffray C, Eberl G, Lucas B, Martin B. Macrophages Induce Long-Term Trapping of γδ T Cells with Innate-like Properties within Secondary Lymphoid Organs in the Steady State. THE JOURNAL OF IMMUNOLOGY 2017; 199:1998-2007. [PMID: 28779024 DOI: 10.4049/jimmunol.1700430] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 07/10/2017] [Indexed: 01/06/2023]
Abstract
So far, peripheral T cells have mostly been described to circulate between blood, secondary lymphoid organs (SLOs), and lymph in the steady state. This nomadic existence would allow them to accomplish their surveying task for both foreign Ags and survival signals. Although it is now well established that γδ T cells can be rapidly recruited to inflammatory sites or in certain tumor microenvironments, the trafficking properties of peripheral γδ T cells have been poorly studied in the steady state. In the present study, we highlight the existence of resident γδ T cells in the SLOs of specific pathogen-free mice. Indeed, using several experimental approaches such as the injection of integrin-neutralizing Abs that inhibit the entry of circulating lymphocytes into lymph nodes and long-term parabiosis experiments, we have found that, contrary to Ly-6C-/+CD44lo and Ly-6C+CD44hi γδ T cells, a significant proportion of Ly-6C-CD44hi γδ T cells are trapped for long periods of time within lymph nodes and the spleen in the steady state. Specific in vivo cell depletion strategies have allowed us to demonstrate that macrophages are the main actors involved in this long-term retention of Ly-6C-CD44hi γδ T cells in SLOs.
Collapse
Affiliation(s)
| | - Matthieu Rivière
- Institut Cochin, CNRS UMR8104, INSERM U1016, Paris Descartes Université, 75014 Paris, France
| | - Aurélie Durand
- Institut Cochin, CNRS UMR8104, INSERM U1016, Paris Descartes Université, 75014 Paris, France
| | - Elisa Peranzoni
- Institut Cochin, CNRS UMR8104, INSERM U1016, Paris Descartes Université, 75014 Paris, France
| | - Vincent Guichard
- Institut Cochin, CNRS UMR8104, INSERM U1016, Paris Descartes Université, 75014 Paris, France.,Paris Diderot Université, 75013 Paris, France
| | - Pauline Hamon
- Université Paris 6, INSERM U1135, CNRS ERL8255, Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Universités, Université Pierre et Marie Curie, 75013 Paris, France
| | - Nelly Bonilla
- Institut Cochin, CNRS UMR8104, INSERM U1016, Paris Descartes Université, 75014 Paris, France
| | - Thomas Guilbert
- Institut Cochin, CNRS UMR8104, INSERM U1016, Paris Descartes Université, 75014 Paris, France
| | - Alexandre Boissonnas
- Université Paris 6, INSERM U1135, CNRS ERL8255, Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Universités, Université Pierre et Marie Curie, 75013 Paris, France
| | - Cédric Auffray
- Institut Cochin, CNRS UMR8104, INSERM U1016, Paris Descartes Université, 75014 Paris, France
| | - Gérard Eberl
- Unité Microenvironment and Immunity, Institut Pasteur, 75724 Paris, France; and.,INSERM U1224, 75724 Paris, France
| | - Bruno Lucas
- Institut Cochin, CNRS UMR8104, INSERM U1016, Paris Descartes Université, 75014 Paris, France
| | - Bruno Martin
- Institut Cochin, CNRS UMR8104, INSERM U1016, Paris Descartes Université, 75014 Paris, France;
| |
Collapse
|
27
|
Khmelinskii I, Makarov V. Temperature dependence of the spin relaxation time of Fe 3 O 4 and hemozoin superparamagnetic nanocrystals. Chem Phys 2017. [DOI: 10.1016/j.chemphys.2017.06.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
28
|
Microfluidic sorting of intrinsically magnetic cells under visual control. Sci Rep 2017; 7:6942. [PMID: 28761104 PMCID: PMC5537260 DOI: 10.1038/s41598-017-06946-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/19/2017] [Indexed: 12/04/2022] Open
Abstract
Magnetic cell sorting provides a valuable complementary mechanism to fluorescent techniques, especially if its parameters can be fine-tuned. In addition, there has recently been growing interest in studying naturally occurring magnetic cells and genetic engineering of cells to render them magnetic in order to control molecular processes via magnetic fields. For such approaches, contamination-free magnetic separation is an essential capability. We here present a robust and tunable microfluidic sorting system in which magnetic gradients of up to 1700 T/m can be applied to cells flowing through a sorting channel by reversible magnetization of ferrofluids. Visual control of the sorting process allowed us to optimize sorting efficiencies for a large range of sizes and magnetic moments of cells. Using automated quantification based on imaging of fluorescent markers, we showed that macrophages containing phagocytosed magnetic nanoparticles, with cellular magnetic dipole moments on the order of 10 fAm2, could be sorted with an efficiency of 90 ± 1%. Furthermore, we successfully sorted intrinsically magnetic magnetotactic bacteria with magnetic moments of 0.1 fAm2. In distinction to column-based magnetic sorting devices, microfluidic systems can prevent sample contact with superparamagnetic material. This ensures contamination-free separation of naturally occurring or bioengineered magnetic cells and is essential for downstream characterization of their properties.
Collapse
|
29
|
Kumar A, Bezbradica JS, Stanic AK, Joyce S. Characterization and Functional Analysis of Mouse Semi-invariant Natural T Cells. ACTA ACUST UNITED AC 2017; 117:14.13.1-14.13.55. [PMID: 28369682 DOI: 10.1002/cpim.22] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Semi-invariant natural killer T (iNKT) cells are CD1d-restricted innate-like lymphocytes that recognize lipid agonists. Activated iNKT cells have immunoregulatory properties. Human and mouse iNKT cell functions elicited by different glycolipid agonists are highly conserved, making the mouse an excellent animal model for understanding iNKT cell biology in vivo. This unit describes basic methods for the characterization and quantification (see Basic Protocol 1) and functional analysis of mouse iNKT cells in vivo or in vitro. This unit also contains protocols that describe enrichment and purification of iNKT cells, generation of CD1d tetramer, and lipid antigen loading onto cell-bound and soluble CD1d for activation of NKT cell hybridomas. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Amrendra Kumar
- Veterans Administration, Tennessee Valley Healthcare System, Nashville, Tennessee.,Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | - Sebastian Joyce
- Veterans Administration, Tennessee Valley Healthcare System, Nashville, Tennessee.,Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
30
|
Nairz M, Theurl I, Swirski FK, Weiss G. "Pumping iron"-how macrophages handle iron at the systemic, microenvironmental, and cellular levels. Pflugers Arch 2017; 469:397-418. [PMID: 28251312 PMCID: PMC5362662 DOI: 10.1007/s00424-017-1944-8] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/25/2017] [Accepted: 01/29/2017] [Indexed: 12/12/2022]
Abstract
Macrophages reside in virtually every organ. First arising during embryogenesis, macrophages replenish themselves in the adult through a combination of self-renewal and influx of bone marrow-derived monocytes. As large phagocytic cells, macrophages participate in innate immunity while contributing to tissue-specific homeostatic functions. Among the key metabolic tasks are senescent red blood cell recycling, free heme detoxification, and provision of iron for de novo hemoglobin synthesis. While this systemic mechanism involves the shuttling of iron between spleen, liver, and bone marrow through the concerted function of defined macrophage populations, similar circuits appear to exist within the microenvironment of other organs. The high turnover of iron is the prerequisite for continuous erythropoiesis and tissue integrity but challenges macrophages’ ability to maintain cellular iron homeostasis and immune function. This review provides a brief overview of systemic, microenvironmental, and cellular aspects of macrophage iron handling with a focus on exciting and unresolved questions in the field.
Collapse
Affiliation(s)
- Manfred Nairz
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Anichstr. 35, 6020, Innsbruck, Austria. .,Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA. .,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Igor Theurl
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Anichstr. 35, 6020, Innsbruck, Austria
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Guenter Weiss
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Anichstr. 35, 6020, Innsbruck, Austria.
| |
Collapse
|
31
|
Loevenich K, Ueffing K, Abel S, Hose M, Matuschewski K, Westendorf AM, Buer J, Hansen W. DC-Derived IL-10 Modulates Pro-inflammatory Cytokine Production and Promotes Induction of CD4 +IL-10 + Regulatory T Cells during Plasmodium yoelii Infection. Front Immunol 2017; 8:152. [PMID: 28293237 PMCID: PMC5328999 DOI: 10.3389/fimmu.2017.00152] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/30/2017] [Indexed: 02/02/2023] Open
Abstract
The cytokine IL-10 plays a crucial role during malaria infection by counteracting the pro-inflammatory immune response. We and others demonstrated that Plasmodium yoelii infection results in enhanced IL-10 production in CD4+ T cells accompanied by the induction of an immunosuppressive phenotype. However, it is unclear whether this is a direct effect caused by the parasite or an indirect consequence due to T cell activation by IL-10-producing antigen-presenting cells. Here, we demonstrate that CD11c+CD11b+CD8− dendritic cells (DCs) produce elevated levels of IL-10 after P. yoelii infection of BALB/c mice. DC-specific ablation of IL-10 in P. yoelii-infected IL-10flox/flox/CD11c-cre mice resulted in increased IFN-γ and TNF-α production with no effect on MHC-II, CD80, or CD86 expression in CD11c+ DCs. Accordingly, DC-specific ablation of IL-10 exacerbated systemic IFN-γ and IL-12 production without altering P. yoelii blood stage progression. Strikingly, DC-specific inactivation of IL-10 in P. yoelii-infected mice interfered with the induction of IL-10-producing CD4+ T cells while raising the frequency of IFN-γ-secreting CD4+ T cells. These results suggest that P. yoelii infection promotes IL-10 production in DCs, which in turn dampens secretion of pro-inflammatory cytokines and supports the induction of CD4+IL-10+ T cells.
Collapse
Affiliation(s)
- Katharina Loevenich
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen , Essen , Germany
| | - Kristina Ueffing
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen , Essen , Germany
| | - Simone Abel
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen , Essen , Germany
| | - Matthias Hose
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen , Essen , Germany
| | - Kai Matuschewski
- Institute of Biology, Humboldt University, Berlin, Germany; Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen , Essen , Germany
| | - Jan Buer
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen , Essen , Germany
| | - Wiebke Hansen
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen , Essen , Germany
| |
Collapse
|
32
|
Abstract
In light of an enhanced awareness of ethical questions and ever increasing costs when working with animals in biomedical research, there is a dedicated and sometimes fierce debate concerning the (lack of) reproducibility of animal models and their relevance for human inflammatory diseases. Despite evident advancements in searching for alternatives, that is, replacing, reducing, and refining animal experiments-the three R's of Russel and Burch (1959)-understanding the complex interactions of the cells of the immune system, the nervous system and the affected tissue/organ during inflammation critically relies on in vivo models. Consequently, scientific advancement and ultimately novel therapeutic interventions depend on improving the reproducibility of animal inflammation models. As a prelude to the remaining hands-on protocols described in this volume, here, we summarize potential pitfalls of preclinical animal research and provide resources and background reading on how to avoid them.
Collapse
|
33
|
Mannose receptor induces T-cell tolerance via inhibition of CD45 and up-regulation of CTLA-4. Proc Natl Acad Sci U S A 2016; 113:10649-54. [PMID: 27601670 DOI: 10.1073/pnas.1605885113] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The mannose receptor (MR) is an endocytic receptor involved in serum homeostasis and antigen presentation. Here, we identify the MR as a direct regulator of CD8(+) T-cell activity. We demonstrate that MR expression on dendritic cells (DCs) impaired T-cell cytotoxicity in vitro and in vivo. This regulatory effect of the MR was mediated by a direct interaction with CD45 on the T cell, inhibiting its phosphatase activity, which resulted in up-regulation of cytotoxic T-lymphocyte-associated Protein 4 (CTLA-4) and the induction of T-cell tolerance. Inhibition of CD45 prevented expression of B-cell lymphoma 6 (Bcl-6), a transcriptional inhibitor that directly bound the CTLA-4 promoter and regulated its activity. These data demonstrate that endocytic receptors expressed on DCs contribute to the regulation of T-cell functionality.
Collapse
|
34
|
Inyushin M, Kucheryavih Y, Kucheryavih L, Rojas L, Khmelinskii I, Makarov V. Superparamagnetic Properties of Hemozoin. Sci Rep 2016; 6:26212. [PMID: 27188748 PMCID: PMC4870585 DOI: 10.1038/srep26212] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 04/27/2016] [Indexed: 11/09/2022] Open
Abstract
We report that hemozoin nanocrystals demonstrate superparamagnetic properties, with direct measurements of the synthetic hemozoin magnetization. The results show that the magnetic permeability constant varies from μ = 4585 (at -20 °C) to 3843 (+20 °C), with the values corresponding to a superparamagnetic system. Similar results were obtained from the analysis of the diffusion separation of natural hemozoin nanocrystals in the magnetic field gradient, with μ = 6783 exceeding the value obtained in direct measurements by the factor of 1.8. This difference is interpreted in terms of structural differences between the synthetic and natural hemozoin. The ab initio analysis of the hemozoin elementary cell showed that the Fe(3+) ion is in the high-spin state (S = 5/2), while the exchange interaction between Fe(3+) electron-spin states was much stronger than kBT at room temperature. Thus, the spin dynamics of the neighboring Fe(3+) ions are strongly correlated, lending support to the superparamagnetism.
Collapse
Affiliation(s)
- M. Inyushin
- Universidad Central del Caribe, Bayamón, PR 00960-6032, USA
| | | | - L. Kucheryavih
- Universidad Central del Caribe, Bayamón, PR 00960-6032, USA
| | - L. Rojas
- Universidad Central del Caribe, Bayamón, PR 00960-6032, USA
| | - I. Khmelinskii
- University of the Algarve, FCT, DQB and CIQA, 8005-139, Faro, Portugal
| | - V. Makarov
- University of Puerto Rico, Rio Piedras Campus, PO Box 23343, San Juan, PR 00931-3343, USA
| |
Collapse
|
35
|
Franken L, Schiwon M, Kurts C. Macrophages: sentinels and regulators of the immune system. Cell Microbiol 2016; 18:475-87. [PMID: 26880038 DOI: 10.1111/cmi.12580] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 02/03/2016] [Accepted: 02/11/2016] [Indexed: 12/15/2022]
Abstract
The important role of macrophages in host defense against a variety of pathogens has long been recognized and has been documented and reviewed in numerous publications. Recently, it has become clear that tissue macrophages are not entirely derived from monocytes, as has been assumed for a long time, but rather show an ontogenetic dichotomy in most tissues: while part of the tissue macrophages are derived from monocytes, a major subset is prenatally seeded from the yolk sac. The latter subset shows a remarkable longevity and is maintained by self-renewal in the adult animal. This paradigm shift poses interesting questions: are these two macrophage subsets functionally equivalent cells that are recruited into the tissue at different development stages, or are both macrophage subsets discrete cell types with distinct functions, which have to exist side by side? Is the functional specialization that can be observed in most macrophages due to their lineage or due to their anatomical niche? This review will give an overview about what we know of macrophage ontogeny and will discuss the influence of the macrophage lineage and location on their functional specialization.
Collapse
Affiliation(s)
- Lars Franken
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms-University, Sigmund-Freud-Str. 25, Bonn, 53105, Germany
| | - Marzena Schiwon
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms-University, Sigmund-Freud-Str. 25, Bonn, 53105, Germany
| | - Christian Kurts
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms-University, Sigmund-Freud-Str. 25, Bonn, 53105, Germany
| |
Collapse
|
36
|
Kurts C, Gottschalk C, Bedoui S, Heinzel S, Godfrey D, Enders A. German Society for Immunology and Australasian Society for Immunology joint Workshop 3(rd) -4(th) December 2015 - Meeting report. Eur J Immunol 2016; 46:265-8. [PMID: 26840197 DOI: 10.1002/eji.201670024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The German Society for Immunology (DGfI) and the Australasian Society for Immunology (ASI) hosted the first DGfI-ASI joint workshop from December 3-4, 2015 in Canberra, Australia. A delegation of 15 distinguished German immunologists discussed the workshop topic "immune regulation in infections and immune mediated diseases" with the aim to establish new German-Australasian collaborations, discuss new concepts in the field of immune regulation and build a scientific network to create more utilizable resources for excellent (trans-border) immunological research. The workshop was associated with the 45(th) Annual Scientific Meeting of the ASI held from Nov 29-Dec 3, 2015, opening up even more opportunities for finding new collaboration partners. A return meeting will be linked to the annual DGfI meeting that will take place in 2017 in Erlangen.
Collapse
Affiliation(s)
- Christian Kurts
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Catherine Gottschalk
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Sammy Bedoui
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Susanne Heinzel
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Dale Godfrey
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Anselm Enders
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| |
Collapse
|
37
|
WISP1 mediates hepatic warm ischemia reperfusion injury via TLR4 signaling in mice. Sci Rep 2016; 6:20141. [PMID: 26821752 PMCID: PMC4731767 DOI: 10.1038/srep20141] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 12/30/2015] [Indexed: 12/22/2022] Open
Abstract
Wnt-induced secreted protein-1 (WISP1) is an extracellular matrix protein that has been reported in cancer researches. Our previous studies on WISP1 implied it could be a harmful mediator in septic mice. However, its role in liver ischemia reperfusion (I/R) injury is unknown. This study investigated the effects of WISP1 on liver I/R damage. Male C57BL/6 wild-type mice were used to undergo 60 min segmental (70%) ischemia. WISP1 expression was measured after indicated time points of reperfusion. Anti-WISP1 antibody was injected intraperitoneally to mice. Toll-like receptor 4 (TLR4) knockout mice and TIR-domain-containing adaptor inducing interferon-β (TRIF) knockout mice were adopted in this study. WISP1 was significantly enhanced after 6 h of reperfusion when compared with sham treated mice and significantly decreased either by TLR4 knockout mice or TRIF knockout mice. Anti-WISP1 antibody significantly decreased serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), pathological changes and pro-inflammatory cytokine levels in the mice following I/R. Furthermore, significantly increased serum transaminase levels were found in C57 wild-type mice treated with recombinant WISP1 protein, but not found in TLR4 knockout or TRIF knockout mice subjected to liver I/R. Taken together, WISP1 might contribute to hepatic ischemia reperfusion injury in mice and possibly depends on TLR4/TRIF signaling.
Collapse
|