1
|
Tao X, Tao R, Wang K, Wu L. Anti-inflammatory mechanism of Apolipoprotein A-I. Front Immunol 2024; 15:1417270. [PMID: 39040119 PMCID: PMC11260610 DOI: 10.3389/fimmu.2024.1417270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 06/19/2024] [Indexed: 07/24/2024] Open
Abstract
Apolipoprotein A-I(ApoA-I) is a member of blood apolipoproteins, it is the main component of High density lipoprotein(HDL). ApoA-I undergoes a series of complex processes from its generation to its composition as spherical HDL. It not only has a cholesterol reversal transport function, but also has a function in modulating the inflammatory response. ApoA-I exerts its anti-inflammatory effects mainly by regulating the functions of immune cells, such as monocytes/macrophages, dendritic cells, neutrophils, and T lymphocytes. It also modulates the function of vascular endothelial cells and adipocytes. Additionally, ApoA-I directly exerts anti-inflammatory effects against pathogenic microorganisms or their products. Intensive research on ApoA-I will hopefully lead to better diagnosis and treatment of inflammatory diseases.
Collapse
Affiliation(s)
| | | | - Kaiyang Wang
- Department of Emergency, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | | |
Collapse
|
2
|
Huelsboemer L, Knoedler L, Kochen A, Yu CT, Hosseini H, Hollmann KS, Choi AE, Stögner VA, Knoedler S, Hsia HC, Pomahac B, Kauke-Navarro M. Cellular therapeutics and immunotherapies in wound healing - on the pulse of time? Mil Med Res 2024; 11:23. [PMID: 38637905 PMCID: PMC11025282 DOI: 10.1186/s40779-024-00528-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 04/10/2024] [Indexed: 04/20/2024] Open
Abstract
Chronic, non-healing wounds represent a significant challenge for healthcare systems worldwide, often requiring significant human and financial resources. Chronic wounds arise from the complex interplay of underlying comorbidities, such as diabetes or vascular diseases, lifestyle factors, and genetic risk profiles which may predispose extremities to local ischemia. Injuries are further exacerbated by bacterial colonization and the formation of biofilms. Infection, consequently, perpetuates a chronic inflammatory microenvironment, preventing the progression and completion of normal wound healing. The current standard of care (SOC) for chronic wounds involves surgical debridement along with localized wound irrigation, which requires inpatient care under general anesthesia. This could be followed by, if necessary, defect coverage via a reconstructive ladder utilizing wound debridement along with skin graft, local, or free flap techniques once the wound conditions are stabilized and adequate blood supply is restored. To promote physiological wound healing, a variety of approaches have been subjected to translational research. Beyond conventional wound healing drugs and devices that currently supplement treatments, cellular and immunotherapies have emerged as promising therapeutics that can behave as tailored therapies with cell- or molecule-specific wound healing properties. However, in contrast to the clinical omnipresence of chronic wound healing disorders, there remains a shortage of studies condensing the current body of evidence on cellular therapies and immunotherapies for chronic wounds. This review provides a comprehensive exploration of current therapies, experimental approaches, and translational studies, offering insights into their efficacy and limitations. Ultimately, we hope this line of research may serve as an evidence-based foundation to guide further experimental and translational approaches and optimize patient care long-term.
Collapse
Affiliation(s)
- Lioba Huelsboemer
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Leonard Knoedler
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
- School of Medicine, University of Regensburg, 93040, Regensburg, Germany
| | - Alejandro Kochen
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
- Regenerative Wound Healing Center, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Catherine T Yu
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Helia Hosseini
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Katharina S Hollmann
- School of Medicine, University of Wuerzburg, 97070, Würzburg, Germany
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Ashley E Choi
- California University of Science and Medicine, Colton, CA, 92324, USA
| | - Viola A Stögner
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Samuel Knoedler
- School of Medicine, University of Regensburg, 93040, Regensburg, Germany
| | - Henry C Hsia
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
- Regenerative Wound Healing Center, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Bohdan Pomahac
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Martin Kauke-Navarro
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
3
|
Cornice J, Verzella D, Arboretto P, Vecchiotti D, Capece D, Zazzeroni F, Franzoso G. NF-κB: Governing Macrophages in Cancer. Genes (Basel) 2024; 15:197. [PMID: 38397187 PMCID: PMC10888451 DOI: 10.3390/genes15020197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 01/26/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are the major component of the tumor microenvironment (TME), where they sustain tumor progression and or-tumor immunity. Due to their plasticity, macrophages can exhibit anti- or pro-tumor functions through the expression of different gene sets leading to distinct macrophage phenotypes: M1-like or pro-inflammatory and M2-like or anti-inflammatory. NF-κB transcription factors are central regulators of TAMs in cancers, where they often drive macrophage polarization toward an M2-like phenotype. Therefore, the NF-κB pathway is an attractive therapeutic target for cancer immunotherapy in a wide range of human tumors. Hence, targeting NF-κB pathway in the myeloid compartment is a potential clinical strategy to overcome microenvironment-induced immunosuppression and increase anti-tumor immunity. In this review, we discuss the role of NF-κB as a key driver of macrophage functions in tumors as well as the principal strategies to overcome tumor immunosuppression by targeting the NF-κB pathway.
Collapse
Affiliation(s)
- Jessica Cornice
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK; (J.C.); (P.A.)
| | - Daniela Verzella
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Paola Arboretto
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK; (J.C.); (P.A.)
| | - Davide Vecchiotti
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Daria Capece
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Francesca Zazzeroni
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Guido Franzoso
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK; (J.C.); (P.A.)
| |
Collapse
|
4
|
Vicari P. The many faces of a macrophage. Morphologie 2023; 107:163-166. [PMID: 36207239 DOI: 10.1016/j.morpho.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/27/2022] [Accepted: 09/13/2022] [Indexed: 05/14/2023]
Affiliation(s)
- P Vicari
- Hematology Service at Hospital do Servidor Público Estado de São Paulo- IAMSPE, Rua Pedro de Toledo, 1800 13, Andar- central 04029-000, São Paulo, SP, Brazil.
| |
Collapse
|
5
|
Jia D, Cai J, Yao F, Zhu P, Xu X, Qi Y, Wang H. Effect of Bacillus Subtilis on Immune Function of Hd11 Chicken Macrophages. BRAZILIAN JOURNAL OF POULTRY SCIENCE 2023. [DOI: 10.1590/1806-9061-2022-1641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Affiliation(s)
- D Jia
- Jiangsu Lihua Animal Husbandry Co., Ltd, P.R.China
| | - J Cai
- Yangzhou University, P.R.China
| | - F Yao
- Yangzhou University, P.R.China
| | - P Zhu
- Jiangsu Lihua Animal Husbandry Co., Ltd, P.R.China; Yangzhou University, P.R.China
| | - X Xu
- Jiangsu Lihua Animal Husbandry Co., Ltd, P.R.China
| | - Y Qi
- Jiangsu Lihua Animal Husbandry Co., Ltd, P.R.China
| | - H Wang
- Yangzhou University, P.R.China
| |
Collapse
|
6
|
Singh A, Mayengbam SS, Yaduvanshi H, Wani MR, Bhat MK. Obesity Programs Macrophages to Support Cancer Progression. Cancer Res 2022; 82:4303-4312. [PMID: 36191083 DOI: 10.1158/0008-5472.can-22-1257] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 06/14/2022] [Accepted: 09/26/2022] [Indexed: 01/24/2023]
Abstract
Obesity induces multifactorial effects such as dyslipidemia, insulin resistance, and arterial hypertension that influence the progression of many diseases. Obesity is associated with an increased incidence of cancers, and multiple mechanisms link obesity with cancer initiation and progression. Macrophages participate in the homeostasis of adipose tissue and play an important role in cancer. Adipose tissue expansion in obesity alters the balance between pro- and anti-inflammatory macrophages, which is a primary cause of inflammation. Chronic low-grade inflammation driven by macrophages is also an important characteristic of cancer. Adipocytes secrete various adipokines, including adiponectin, leptin, IL6, and TNFα, that influence macrophage behavior and tumor progression. Furthermore, other metabolic effects of obesity, such as hyperlipidemia, hyperglycemia, and hypercholesterolemia, can also regulate macrophage functionality in cancer. This review summarizes how obesity influences macrophage-tumor cell interactions and the role of macrophages in the response to anticancer therapies under obese conditions.
Collapse
Affiliation(s)
- Abhijeet Singh
- Department of Biotechnology, National Centre for Cell Science, Government of India, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, Maharashtra, India
| | - Shyamananda Singh Mayengbam
- Department of Biotechnology, National Centre for Cell Science, Government of India, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, Maharashtra, India
| | - Himanshi Yaduvanshi
- Department of Biotechnology, National Centre for Cell Science, Government of India, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, Maharashtra, India
| | - Mohan R Wani
- Department of Biotechnology, National Centre for Cell Science, Government of India, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, Maharashtra, India
| | - Manoj Kumar Bhat
- Department of Biotechnology, National Centre for Cell Science, Government of India, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, Maharashtra, India
| |
Collapse
|
7
|
Yu L, Wang L, Hu G, Ren L, Qiu C, Li S, Zhou X, Chen S, Chen R. Reprogramming alternative macrophage polarization by GATM-mediated endogenous creatine synthesis: A potential target for HDM-induced asthma treatment. Front Immunol 2022; 13:937331. [PMID: 36177049 PMCID: PMC9513582 DOI: 10.3389/fimmu.2022.937331] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Cellular energy metabolism plays a crucial role in the regulation of macrophage polarization and in the execution of immune functions. A recent study showed that Slc6a8-mediated creatine uptake from exogenous supplementation modulates macrophage polarization, yet little is known about the role of the de novo creatine de novobiosynthesis pathway in macrophage polarization. Here, we observed that glycine amidinotransferase (GATM), the rate-limiting enzyme for creatine synthesis, was upregulated in alternative (M2) polarized macrophages, and was dependent on the transcriptional factor STAT6, whereas GATM expression was suppressed in the classical polarized (M1) macrophage. Next, we revealed that exogenous creatine supplementation enhanced IL-4-induced M2 polarization, confirming recent work. Furthermore, we revealed that genetic ablation of GATM did not affect expression of M1 marker genes (Nos2, IL1b, IL12b) or the production of nitric oxide in both peritoneal macrophages (PMs) and bone marrow-derived macrophages (BMDMs). By contrast, expression levels of M2 markers (Arg1, Mrc1, Ccl17 and Retnla) were lower following GATM deletion. Moreover, we found that deletion of GATM in resident alveolar macrophages (AMs) significantly blocked M2 polarization but with no obvious effect on the number of cells in knockout mice. Lastly, an upregulation of GATM was found in lung tissue and bronchoalveolar lavage fluid macrophages from HDM-induced asthmatic mice. Our study uncovers a previously uncharacterized role for the de novo creatine biosynthesis enzyme GATM in M2 macrophage polarization, which may be involved in the pathogenesis of related inflammatory diseases such as an T helper 2 (Th2)-associated allergic asthma.
Collapse
Affiliation(s)
- Li Yu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People’s Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Lingwei Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People’s Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Guang Hu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People’s Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Laibin Ren
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People’s Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chen Qiu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People’s Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Shun Li
- Department of Animal Model, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- *Correspondence: Rongchang Chen, ; Shanze Chen, ; Xiaohui Zhou, ; Shun Li,
| | - Xiaohui Zhou
- Department of Animal Model, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- *Correspondence: Rongchang Chen, ; Shanze Chen, ; Xiaohui Zhou, ; Shun Li,
| | - Shanze Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People’s Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen, China
- *Correspondence: Rongchang Chen, ; Shanze Chen, ; Xiaohui Zhou, ; Shun Li,
| | - Rongchang Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People’s Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen, China
- *Correspondence: Rongchang Chen, ; Shanze Chen, ; Xiaohui Zhou, ; Shun Li,
| |
Collapse
|
8
|
Yu L, Wang L, Hu G, Ren L, Qiu C, Li S, Zhou X, Chen S, Chen R. Reprogramming alternative macrophage polarization by GATM-mediated endogenous creatine synthesis: A potential target for HDM-induced asthma treatment. Front Immunol 2022; 13:937331. [PMID: 36177049 PMCID: PMC9513582 DOI: 10.3389/fimmu.2022.937331 10.3389/fimmu.2022.937331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Cellular energy metabolism plays a crucial role in the regulation of macrophage polarization and in the execution of immune functions. A recent study showed that Slc6a8-mediated creatine uptake from exogenous supplementation modulates macrophage polarization, yet little is known about the role of the de novo creatine de novobiosynthesis pathway in macrophage polarization. Here, we observed that glycine amidinotransferase (GATM), the rate-limiting enzyme for creatine synthesis, was upregulated in alternative (M2) polarized macrophages, and was dependent on the transcriptional factor STAT6, whereas GATM expression was suppressed in the classical polarized (M1) macrophage. Next, we revealed that exogenous creatine supplementation enhanced IL-4-induced M2 polarization, confirming recent work. Furthermore, we revealed that genetic ablation of GATM did not affect expression of M1 marker genes (Nos2, IL1b, IL12b) or the production of nitric oxide in both peritoneal macrophages (PMs) and bone marrow-derived macrophages (BMDMs). By contrast, expression levels of M2 markers (Arg1, Mrc1, Ccl17 and Retnla) were lower following GATM deletion. Moreover, we found that deletion of GATM in resident alveolar macrophages (AMs) significantly blocked M2 polarization but with no obvious effect on the number of cells in knockout mice. Lastly, an upregulation of GATM was found in lung tissue and bronchoalveolar lavage fluid macrophages from HDM-induced asthmatic mice. Our study uncovers a previously uncharacterized role for the de novo creatine biosynthesis enzyme GATM in M2 macrophage polarization, which may be involved in the pathogenesis of related inflammatory diseases such as an T helper 2 (Th2)-associated allergic asthma.
Collapse
Affiliation(s)
- Li Yu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People’s Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Lingwei Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People’s Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Guang Hu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People’s Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Laibin Ren
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People’s Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chen Qiu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People’s Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Shun Li
- Department of Animal Model, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China,*Correspondence: Rongchang Chen, ; Shanze Chen, ; Xiaohui Zhou, ; Shun Li,
| | - Xiaohui Zhou
- Department of Animal Model, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China,*Correspondence: Rongchang Chen, ; Shanze Chen, ; Xiaohui Zhou, ; Shun Li,
| | - Shanze Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People’s Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen, China,*Correspondence: Rongchang Chen, ; Shanze Chen, ; Xiaohui Zhou, ; Shun Li,
| | - Rongchang Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People’s Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen, China,*Correspondence: Rongchang Chen, ; Shanze Chen, ; Xiaohui Zhou, ; Shun Li,
| |
Collapse
|
9
|
Liposome encapsulated clodronate mediated elimination of pathogenic macrophages and microglia: A promising pharmacological regime to defuse cytokine storm in COVID-19. MEDICINE IN DRUG DISCOVERY 2022; 15:100136. [PMID: 35721801 PMCID: PMC9190184 DOI: 10.1016/j.medidd.2022.100136] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 12/14/2022] Open
Abstract
The emergence of new SARS-CoV-2 variants continues to pose an enormous public health concern. The SARS-CoV-2 infection disrupted host immune response accounting for cytokine storm has been linked to multiorgan failure and mortality in a significant portion of positive cases. Abruptly activated macrophages have been identified as the key pathogenic determinant of cytokine storm in COVID-19. Besides, reactive microglia have been known to discharge a surplus amount of proinflammatory factors leading to neuropathogenic events in the brains of SARS-CoV-2 infected individuals. Considering the fact, depletion of activated macrophages and microglia could be proposed to eradicate the life-threatening cytokine storm in COVID-19. Clodronate, a non-nitrogenous bisphosphonate drug has been identified as a potent macrophage and microglial depleting agent. While recent advancement in the field of liposome encapsulation technology offers the most promising biological tool for drug delivery, liposome encapsulated clodronate has been reported to effectively target and induce prominent phagocytic cell death in activated macrophages and microglia compared to free clodronate molecules. Thus, in this review article, we emphasize that depletion of activated macrophages and microglial cells by administration of liposome encapsulated clodronate can be a potential therapeutic strategy to diminish the pathogenic cytokine storm and alleviate multiorgan failure in COVID-19. Moreover, recently developed COVID-19 vaccines appear to render the chronic activation of macrophages accounting for immunological dysregulation in some cases. Therefore, the use of liposome encapsulated clodronate can also be extended to the clinical management of unforeseen immunogenic reactions resulting from activated macrophages associated adverse effects of COVID-19 vaccines.
Collapse
|
10
|
Cruz MS, Loureiro JP, Oliveira MJ, Macedo MF. The iNKT Cell-Macrophage Axis in Homeostasis and Disease. Int J Mol Sci 2022; 23:ijms23031640. [PMID: 35163561 PMCID: PMC8835952 DOI: 10.3390/ijms23031640] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 12/12/2022] Open
Abstract
Invariant natural killer T (iNKT) cells are CD1d-restricted, lipid-reactive T cells that exhibit preponderant immunomodulatory properties. The ultimate protective or deleterious functions displayed by iNKT cells in tissues are known to be partially shaped by the interactions they establish with other immune cells. In particular, the iNKT cell–macrophage crosstalk has gained growing interest over the past two decades. Accumulating evidence has highlighted that this immune axis plays central roles not only in maintaining homeostasis but also during the development of several pathologies. Hence, this review summarizes the reported features of the iNKT cell–macrophage axis in health and disease. We discuss the pathophysiological significance of this interplay and provide an overview of how both cells communicate with each other to regulate disease onset and progression in the context of infection, obesity, sterile inflammation, cancer and autoimmunity.
Collapse
Affiliation(s)
- Mariana S. Cruz
- Cell Activation and Gene Expression Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.S.C.); (J.P.L.)
- Department of Medical Sciences, University of Aveiro (UA), 3810-193 Aveiro, Portugal
| | - José Pedro Loureiro
- Cell Activation and Gene Expression Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.S.C.); (J.P.L.)
- Experimental Immunology Group, Department of Biomedicine (DBM), University of Basel and University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Maria J. Oliveira
- Tumour and Microenvironment Interactions Group, Instituto Nacional de Engenharia Biomédica (INEB), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal;
- Department of Molecular Biology, ICBAS-Institute of Biomedical Sciences Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Maria Fatima Macedo
- Cell Activation and Gene Expression Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.S.C.); (J.P.L.)
- Department of Medical Sciences, University of Aveiro (UA), 3810-193 Aveiro, Portugal
- Correspondence:
| |
Collapse
|
11
|
Jiang Y, Tao Z, Chen H, Xia S. Endoplasmic Reticulum Quality Control in Immune Cells. Front Cell Dev Biol 2021; 9:740653. [PMID: 34660599 PMCID: PMC8511527 DOI: 10.3389/fcell.2021.740653] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/07/2021] [Indexed: 12/18/2022] Open
Abstract
The endoplasmic reticulum quality control (ERQC) system, including endoplasmic reticulum-associated degradation (ERAD), the unfolded protein response (UPR), and autophagy, presides over cellular protein secretion and maintains proteostasis in mammalian cells. As part of the immune system, a variety of proteins are synthesized and assembled correctly for the development, activation, and differentiation of immune cells, such as dendritic cells (DCs), macrophages, myeloid-derived-suppressor cells (MDSCs), B lymphocytes, T lymphocytes, and natural killer (NK) cells. In this review, we emphasize the role of the ERQC in these immune cells, and also discuss how the imbalance of ER homeostasis affects the immune response, thereby suggesting new therapeutic targets for immunotherapy.
Collapse
Affiliation(s)
- Yalan Jiang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zehua Tao
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Hua Chen
- Department of Colorectal Surgery, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
12
|
Monocyte Differentiation into Destructive Macrophages on In Vitro Administration of Gingival Crevicular Fluid from Periodontitis Patients. J Pers Med 2021; 11:jpm11060555. [PMID: 34203667 PMCID: PMC8232302 DOI: 10.3390/jpm11060555] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/10/2021] [Accepted: 06/12/2021] [Indexed: 01/01/2023] Open
Abstract
Background: Periodontitis is an inflammatory condition of the tooth-supporting structures initiated and perpetuated by pathogenic bacteria present in the dental plaque biofilm. In periodontitis, immune cells infiltrate the periodontium to prevent bacterial insult. Macrophages derived from monocytes play an important role in antigen presentation to lymphocytes. However, they are also implicated in causing periodontal destruction and bystander damage to the host tissues. Objectives: The objective of the present study was to quantify the cytokine profile of gingival crevicular fluid (GCF) samples obtained from patients with periodontitis. The study further aimed to assess if GCF of periodontitis patients could convert CD14+ monocytes into macrophages of destructive phenotype in an in vitro setting. The secondary objectives of the study were to assess if macrophages that resulted from GCF treatment of monocytes could affect the synthetic properties, stemness, expression of extracellular matrix proteins, adhesion molecules expressed by gingival stem cells, gingival mesenchymal stromal cells, and osteoblasts. Methods: GCF, blood, and gingival tissue samples were obtained from periodontitis subjects and healthy individuals based on specific protocols. Cytokine profiles of the GCF samples were analyzed. CD14+ monocytes were isolated from whole blood, cultured, and treated with the GCF of periodontitis patients to observe if they differentiated into macrophages. Further, the macrophages were assessed for a phenotype by surface marker analysis and cytokine assays. These macrophages were co-cultured with gingival stem cells, epithelial, stromal cells, and osteoblasts to assess the effects of the macrophages on the synthetic activity of the cells. Results: The GCF samples of periodontitis patients had significantly higher levels of IFN gamma, M-CSF, and GM-CSF. Administration of the GCF samples to CD14+ monocytes resulted in their conversion to macrophages that tested positive for CD80, CD86, and CD206. These macrophages produced increased levels of IL-1β, TNF-α, and IL-6. Co-culture of the macrophages with gingival stem cells, epithelial cells, and stromal cells resulted in increased cytotoxicity and apoptotic rates to the gingival cells. A reduced expression of markers related to stemness, extracellular matrix, and adhesion namely OCT4, NANOG, KRT5, POSTN, COL3A1, CDH1, and CDH3 were seen. The macrophages profoundly affected the production of mineralized nodules by osteoblasts and significantly reduced the expression of COL1A1, OSX, and OCN genes. Conclusion: In periodontitis patients, blood-derived monocytes transform into macrophages of a destructive phenotype due to the characteristic cytokine environment of their GCF. Further, the macrophages affect the genotype and phenotype of the resident cells of the periodontium, aggravate periodontal destruction, as well as jeopardize periodontal healing and resolution of inflammation.
Collapse
|
13
|
Crawford CK, Kol A. The Mucosal Innate Immune Response to Cryptosporidium parvum, a Global One Health Issue. Front Cell Infect Microbiol 2021; 11:689401. [PMID: 34113580 PMCID: PMC8185216 DOI: 10.3389/fcimb.2021.689401] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/07/2021] [Indexed: 02/02/2023] Open
Abstract
Cryptosporidium parvum is an apicomplexan parasite that infects the intestinal epithelium of humans and livestock animals worldwide. Cryptosporidiosis is a leading cause of diarrheal-related deaths in young children and a major cause of economic loss in cattle operations. The disease is especially dangerous to infants and immunocompromised individuals, for which there is no effective treatment or vaccination. As human-to-human, animal-to-animal and animal-to-human transmission play a role in cryptosporidiosis disease ecology, a holistic 'One Health' approach is required for disease control. Upon infection, the host's innate immune response restricts parasite growth and initiates the adaptive immune response, which is necessary for parasite clearance and recovery. The innate immune response involves a complex communicative interplay between epithelial and specialized innate immune cells. Traditional models have been used to study innate immune responses to C. parvum but cannot fully recapitulate natural host-pathogen interactions. Recent shifts to human and bovine organoid cultures are enabling deeper understanding of host-specific innate immunity response to infection. This review examines recent advances and highlights research gaps in our understanding of the host-specific innate immune response to C. parvum. Furthermore, we discuss evolving research models used in the field and potential developments on the horizon.
Collapse
Affiliation(s)
- Charles K Crawford
- Department of Pathology, Microbiology, & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Amir Kol
- Department of Pathology, Microbiology, & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
14
|
Gao WJ, Liu JX, Liu MN, Yao YD, Liu ZQ, Liu L, He HH, Zhou H. Macrophage 3D migration: A potential therapeutic target for inflammation and deleterious progression in diseases. Pharmacol Res 2021; 167:105563. [PMID: 33746053 DOI: 10.1016/j.phrs.2021.105563] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 12/14/2022]
Abstract
Macrophages are heterogeneous cells that have different physiological functions, such as chemotaxis, phagocytosis, endocytosis, and secretion of various factors. All physiological functions of macrophages are integral to homeostasis, immune defense and tissue repair. However, in several diseases, macrophages are recruited from the blood towards inflammatory sites. This process is called macrophage migration, which promotes deleterious disease progression. Macrophage migration is a key player in many inflammatory diseases, autoimmune diseases and cancers because it contributes to the accumulation of proinflammatory factors, the destruction of tissues and the development of tumors. Therefore, macrophage migration is proposed to be a potential therapeutic target. Macrophages migrate between two-dimensional (2D) and three-dimensional (3D) environments, implying that distinct migratory features and mechanisms are involved. Compared with the 2D migration of macrophages, 3D migration involves more complex variations in cellular morphology and dynamics. The structure of the extracellular matrix, a key factor, is modified in diseases that influence macrophage 3D migration. Macrophage 3D migration relates to disease pathology. Research that focuses on macrophage 3D migration is an emerging field and was reviewed in this article to indicate the molecular and cellular mechanisms of macrophage migration in 3D environments and to provide potential targets for controlling disease progression associated with this migration.
Collapse
Affiliation(s)
- Wan-Jiao Gao
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, PR China
| | - Jian-Xin Liu
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua City, Hunan Province, PR China
| | - Meng-Nan Liu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, PR China; National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, Sichuan, PR China
| | - Yun-Da Yao
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, PR China
| | - Zhong-Qiu Liu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Liang Liu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, PR China; Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Huan-Huan He
- The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai City, Guangdong Province 519000, PR China
| | - Hua Zhou
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, PR China; Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangzhou University of Chinese Medicine, Guangzhou, PR China; Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai City, Guangdong Province 519000, PR China.
| |
Collapse
|
15
|
Chen Y, Shen J. Mucosal immunity and tRNA, tRF, and tiRNA. J Mol Med (Berl) 2020; 99:47-56. [PMID: 33200232 DOI: 10.1007/s00109-020-02008-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/15/2020] [Accepted: 11/06/2020] [Indexed: 12/13/2022]
Abstract
Mucosal immunity has crucial roles in human diseases such as respiratory tract infection, inflammatory bowel diseases (IBD), and colorectal cancer (CRC). Recent studies suggest that the mononuclear phagocyte system, cancer cells, bacteria, and viruses induce the mucosal immune reaction by various pathways, and can be major factors in the pathogenesis of these diseases. Transfer RNA (tRNA) and its fragments, including tRNA-derived RNA fragments (tRFs) and tRNA-derived stress-induced RNAs (tiRNAs), have emerged as a hot topic in recent years. They not only are verified as essential for transcription and translation but also play roles in cellular homeostasis and functions, such as cell metastasis, proliferation, and apoptosis. However, the specific relationship between their biological regulation and mucosal immunity remains unclear to date. In the present review, we carry out a comprehensive discussion on the specific roles of tRNA, tRFs, and tiRNAs relevant to mucosal immunity and related diseases.
Collapse
Affiliation(s)
- Yueying Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, 160# Pu Jian Ave, Shanghai, 200127, China
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160# Pu Jian Ave, Shanghai, 200127, China
- Shanghai Institute of Digestive Disease, 160# Pu Jian Ave, Shanghai, 200127, China
| | - Jun Shen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, 160# Pu Jian Ave, Shanghai, 200127, China.
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160# Pu Jian Ave, Shanghai, 200127, China.
- Shanghai Institute of Digestive Disease, 160# Pu Jian Ave, Shanghai, 200127, China.
| |
Collapse
|
16
|
Immunotherapy of gynecological cancers. Best Pract Res Clin Obstet Gynaecol 2019; 60:97-110. [PMID: 31003902 DOI: 10.1016/j.bpobgyn.2019.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/01/2019] [Indexed: 12/29/2022]
Abstract
Oncology treatments have evolved from intuitive, via empiric, to the present precision medicine, with the integration of molecular targeted therapies in our treatment arsenal. The use of the patients' powerful immune system has long been contemplated and recently led to the integration of immunotherapy to overturn the well-documented inhibitory effects of the tumor on the immune system and restore it to a state of activity against the cancer. Recent favorable results have shown the value and effectiveness of immunotherapy against gynecological cancers. In particular, the checkpoint inhibitors, targeting the programmed death-1 (PD-1) pathway, have shown durable clinical responses with manageable toxicity. Several phase II and III clinical trials testing the association of different regimen of chemotherapy and immunotherapy are ongoing in gynecological cancers, and important results are expected. In this chapter, we outline the main principles of immunotherapy for gynecological cancers and summarize the current strategies used in clinical trials.
Collapse
|
17
|
Ferko MA, Catelas I. Effects of metal ions on caspase-1 activation and interleukin-1β release in murine bone marrow-derived macrophages. PLoS One 2018; 13:e0199936. [PMID: 30138321 PMCID: PMC6107125 DOI: 10.1371/journal.pone.0199936] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/15/2018] [Indexed: 02/07/2023] Open
Abstract
Ions released from metal implants have been associated with adverse tissue reactions and are therefore a major concern. Studies with macrophages have shown that cobalt, chromium, and nickel ions can activate the NLRP3 inflammasome, a multiprotein complex responsible for the activation of caspase-1 (a proteolytic enzyme converting pro-interleukin [IL]-1β to mature IL-1β). However, the mechanism(s) of inflammasome activation by metal ions remain largely unknown. The objectives of the present study were to determine if, in macrophages: 1. caspase-1 activation and IL-1β release induced by metal ions are oxidative stress-dependent; and 2. IL-1β release induced by metal ions is NF-κB signaling pathway-dependent. Lipopolysaccharide (LPS)-primed murine bone marrow-derived macrophages (BMDM) were exposed to Co2+ (6-48 ppm), Cr3+ (100-500 ppm), or Ni2+ (12-96 ppm), in the presence or absence of a caspase-1 inhibitor (Z-WEHD-FMK), an antioxidant (L-ascorbic acid [L-AA]), or an NF-κB inhibitor (JSH-23). Culture supernatants were analyzed for caspase-1 by western blotting and/or IL-1β release by ELISA. Immunoblotting revealed the presence of caspase-1 (p20 subunit) in supernatants of BMDM incubated with Cr3+, but not with Ni2+ or Co2+. When L-AA (2 mM) was present with Cr3+, the caspase-1 p20 subunit was undetectable and IL-1β release decreased down to the level of the negative control, thereby demonstrating that caspase-1 activation and IL-1β release induced by Cr3+ was oxidative stress-dependent. ELISA demonstrated that Cr3+ induced the highest release of IL-1β, while Co2+ had no or limited effects. In the presence of Ni2+, the addition of L-AA (2 mM) also decreased IL-1β release, below the level of the negative control, suggesting that IL-1β release induced by Ni2+ was also oxidative stress-dependent. Finally, when present during both priming with LPS and activation with Cr3+, JSH-23 blocked IL-1β release, demonstrating NF-κB involvement. Overall, this study showed that while both Cr3+ and Ni2+ may be inducing inflammasome activation, Cr3+ is likely a more potent activator, acting through oxidative stress and the NF-κB signaling pathway.
Collapse
Affiliation(s)
| | - Isabelle Catelas
- Department of Mechanical Engineering, University of Ottawa, Ottawa, Ontario, Canada
- Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
18
|
Ni L, Fang P, Hu ZL, Zhou HY, Chen JG, Wang F, Jin Y. Identification and Function of Acid-sensing Ion Channels in RAW 264.7 Macrophage Cells. Curr Med Sci 2018; 38:436-442. [PMID: 30074209 DOI: 10.1007/s11596-018-1897-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/07/2017] [Indexed: 12/20/2022]
Abstract
Activation of acid-sensing ion channels (ASICs) plays an important role in neuroinflammation. Macrophage recruitment to the sites of inflammation is an essential step in host defense. ASIC1 and ASIC3 have been reported to mediate the endocytosis and maturation of bone marrow derived macrophages. However, the expression and inflammation-related functions of ASICs in RAW 264.7 cells, another common macrophage, are still elusive. In the present study, we first demonstrated the presence of ASIC1, ASIC2a and ASIC3 in RAW 264.7 macrophage cell line by using reverse transcriptase polymerase chain reaction (RT-PCR), Western blotting and immunofluorescence experiments. The non-specific ASICs inhibitor amiloride and specific homomeric ASICla blocker PcTxl reduced the production of iNOS and COX-2 by LPS-induced activating RAW 264.7 cells. Furthermore, not only amiloride but also PcTxl inhibited the migration and LPS-induced apoptosis of RAW 264.7 cells. Taken together, our findings suggest that ASICs promote the inflammatory response and apoptosis of RAW 264.7 cells, and ASICs may serve as a potential novel target for immunological disease therapy.
Collapse
Affiliation(s)
- Lan Ni
- Department of Pharmacology, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Peng Fang
- Department of Pharmacology, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhuang-Li Hu
- Department of Pharmacology, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hai-Yun Zhou
- Department of Pharmacology, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jian-Guo Chen
- Department of Pharmacology, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fang Wang
- Department of Pharmacology, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - You Jin
- Department of Pharmacology, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
19
|
Stichling N, Suomalainen M, Flatt JW, Schmid M, Pacesa M, Hemmi S, Jungraithmayr W, Maler MD, Freudenberg MA, Plückthun A, May T, Köster M, Fejer G, Greber UF. Lung macrophage scavenger receptor SR-A6 (MARCO) is an adenovirus type-specific virus entry receptor. PLoS Pathog 2018. [PMID: 29522575 PMCID: PMC5862501 DOI: 10.1371/journal.ppat.1006914] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Macrophages are a diverse group of phagocytic cells acting in host protection against stress, injury, and pathogens. Here, we show that the scavenger receptor SR-A6 is an entry receptor for human adenoviruses in murine alveolar macrophage-like MPI cells, and important for production of type I interferon. Scavenger receptors contribute to the clearance of endogenous proteins, lipoproteins and pathogens. Knockout of SR-A6 in MPI cells, anti-SR-A6 antibody or the soluble extracellular SR-A6 domain reduced adenovirus type-C5 (HAdV-C5) binding and transduction. Expression of murine SR-A6, and to a lower extent human SR-A6 boosted virion binding to human cells and transduction. Virion clustering by soluble SR-A6 and proximity localization with SR-A6 on MPI cells suggested direct adenovirus interaction with SR-A6. Deletion of the negatively charged hypervariable region 1 (HVR1) of hexon reduced HAdV-C5 binding and transduction, implying that the viral ligand for SR-A6 is hexon. SR-A6 facilitated macrophage entry of HAdV-B35 and HAdV-D26, two important vectors for transduction of hematopoietic cells and human vaccination. The study highlights the importance of scavenger receptors in innate immunity against human viruses. Macrophages are a diverse group of phagocytic cells acting in host protection against stress, injury, and pathogens. They phenotypically and functionally adapt to their local environment, for example, peritoneal macrophages are distinct from brain-resident microglia, from liver-resident Kupffer cells or lung macrophages in the lung. Airway macrophages are among the first cells to encounter human respiratory viruses, such as adenoviruses. They release pro-inflammatory cytokines, kill pathogens, present antigens, and restore tissues. Yet, interactions of viruses with lung macrophages are poorly understood, and it is unclear, how they lead to infection or virus clearance. Here we identified the murine scavenger receptor SR-A6 as a receptor for a subset of human adenoviruses on alveolar macrophage-like cells, so-called MPI cells. Scavenger receptors comprise a large family of trans-membrane proteins, and contribute to the clearance of endogenous proteins, lipoproteins and pathogens. In a series of robust experimentation, we show that adenoviruses use SR-A6 as an entry receptor for infection of MPI cells, and production of type I interferon. MPI cells are non-transformed, self-renewing macrophages derived from fetal murine liver, and closely resemble adult alveolar macrophages. The results demonstrate that SR-A6 binds virions on the surface of alveolar macrophage-like cells, and leads to infection.
Collapse
MESH Headings
- Adenoviridae Infections/immunology
- Adenoviridae Infections/metabolism
- Adenoviridae Infections/virology
- Adenoviruses, Human/immunology
- Animals
- Humans
- Immunity, Innate
- Lung/immunology
- Lung/metabolism
- Lung/virology
- Macrophages/immunology
- Macrophages/metabolism
- Macrophages/virology
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/metabolism
- Macrophages, Alveolar/virology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Protein Binding
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/physiology
- Virus Internalization
Collapse
Affiliation(s)
- Nicole Stichling
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- Molecular Life Sciences Graduate School, ETH and University of Zurich, Switzerland
| | - Maarit Suomalainen
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Justin W. Flatt
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Markus Schmid
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Martin Pacesa
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Silvio Hemmi
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Wolfgang Jungraithmayr
- University Hospital Zurich, Institute of Thorax Surgery, Zurich, Switzerland
- present address: Department of Thoracic Surgery, Medical University Brandenburg, Neuruppin, Germany
| | - Mareike D. Maler
- Max-Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Allergy Research Group, Department of Dermatology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Marina A. Freudenberg
- Max-Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, Albert-Ludwigs-Universität, Freiburg, Germany
- Department of Pneumology, Medical Center–University of Freiburg and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Tobias May
- Inscreenex GmbH, Inhoffenstr. Brunswick, Germany
| | - Mario Köster
- Helmholtz-Zentrum für Infektionsforschung GmbH, Braunschweig, Germany
| | - György Fejer
- School of Biomedical and Healthcare Sciences, Peninsula Schools of Medicine and Dentistry, University of Plymouth, Plymouth, United Kingdom
| | - Urs F. Greber
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
20
|
Rousseau M, Duchez AC, Lee CHC, Boilard E, Laffont B, Corduan A, Provost P. Platelet microparticles reprogram macrophage gene expression and function. Thromb Haemost 2017; 115:311-23. [DOI: 10.1160/th15-05-0389] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 07/22/2015] [Indexed: 12/20/2022]
Abstract
SummaryPlatelet microparticles (MPs) represent the most abundant MPs subtype in the circulation, and can mediate intercellular communication through delivery of bioactives molecules, such as cytokines, proteins, lipids and RNAs. Here, we show that platelet MPs can be internalised by primary human macrophages and deliver functional miR-126–3p. The increase in macrophage miR-126–3p levels was not prevented by actinomycin D, suggesting that it was not due to de novo gene transcription. Platelet MPs dose-dependently downregulated expression of four predicted mRNA targets of miR-126–3p, two of which were confirmed also at the protein level. The mRNA downregulatory effects of platelet MPs were abrogated by expression of a neutralising miR-126–3p sponge, implying the involvement of miR-126–3p. Transcriptome-wide, microarray analyses revealed that as many as 66 microRNAs and 653 additional RNAs were significantly and differentially expressed in macrophages upon exposure to platelet MPs. More specifically, platelet MPs induced an upregulation of 34 microRNAs and a concomitant downregulation of 367 RNAs, including mRNAs encoding for cytokines/chemokines CCL4, CSF1 and TNF. These changes were associated with reduced CCL4, CSF1 and TNF cytokine/chemokine release by macrophages, and accompanied by a marked increase in their phagocytic capacity. These findings demonstrate that platelet MPs can modify the transcriptome of macrophages, and reprogram their function towards a phagocytic phenotype.Supplementary Material to this article is available online at www.thrombosis-online.com.
Collapse
|
21
|
Thomson S, Hamilton CA, Hope JC, Katzer F, Mabbott NA, Morrison LJ, Innes EA. Bovine cryptosporidiosis: impact, host-parasite interaction and control strategies. Vet Res 2017; 48:42. [PMID: 28800747 PMCID: PMC5553596 DOI: 10.1186/s13567-017-0447-0] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 08/01/2017] [Indexed: 01/08/2023] Open
Abstract
Gastrointestinal disease caused by the apicomplexan parasite Cryptosporidium parvum is one of the most important diseases of young ruminant livestock, particularly neonatal calves. Infected animals may suffer from profuse watery diarrhoea, dehydration and in severe cases death can occur. At present, effective therapeutic and preventative measures are not available and a better understanding of the host-pathogen interactions is required. Cryptosporidium parvum is also an important zoonotic pathogen causing severe disease in people, with young children being particularly vulnerable. Our knowledge of the immune responses induced by Cryptosporidium parasites in clinically relevant hosts is very limited. This review discusses the impact of bovine cryptosporidiosis and describes how a thorough understanding of the host-pathogen interactions may help to identify novel prevention and control strategies.
Collapse
Affiliation(s)
- Sarah Thomson
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Edinburgh, EH26 0PZ, Scotland, UK
| | - Carly A Hamilton
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Jayne C Hope
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Frank Katzer
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Edinburgh, EH26 0PZ, Scotland, UK
| | - Neil A Mabbott
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Liam J Morrison
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Elisabeth A Innes
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Edinburgh, EH26 0PZ, Scotland, UK.
| |
Collapse
|
22
|
Goo YH, Son SH, Yechoor VK, Paul A. Transcriptional Profiling of Foam Cells Reveals Induction of Guanylate-Binding Proteins Following Western Diet Acceleration of Atherosclerosis in the Absence of Global Changes in Inflammation. J Am Heart Assoc 2016; 5:e002663. [PMID: 27091181 PMCID: PMC4859273 DOI: 10.1161/jaha.115.002663] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Background Foam cells are central to two major pathogenic processes in atherogenesis: cholesterol buildup in arteries and inflammation. The main underlying cause of cholesterol deposition in arteries is hypercholesterolemia. This study aimed to assess, in vivo, whether elevated plasma cholesterol also alters the inflammatory balance of foam cells. Methods and Results Apolipoprotein E–deficient mice were fed regular mouse chow through the study or were switched to a Western‐type diet (WD) 2 or 14 weeks before death. Consecutive sections of the aortic sinus were used for lesion quantification or to isolate RNA from foam cells by laser‐capture microdissection (LCM) for microarray and quantitative polymerase chain reaction analyses. WD feeding for 2 or 14 weeks significantly increased plasma cholesterol, but the size of atherosclerotic lesions increased only in the 14‐week WD group. Expression of more genes was affected in foam cells of mice under prolonged hypercholesterolemia than in mice fed WD for 2 weeks. However, most transcripts coding for inflammatory mediators remained unchanged in both WD groups. Among the main players in inflammatory or immune responses, chemokine (C‐X‐C motif) ligand 13 was induced in foam cells of mice under WD for 2 weeks. The interferon‐inducible GTPases, guanylate‐binding proteins (GBP)3 and GBP6, were induced in the 14‐week WD group, and other GBP family members were moderately increased. Conclusions Our results indicate that acceleration of atherosclerosis by hypercholesterolemia is not linked to global changes in the inflammatory balance of foam cells. However, induction of GBPs uncovers a novel family of immune modulators with a potential role in atherogenesis.
Collapse
Affiliation(s)
- Young-Hwa Goo
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY
| | - Se-Hee Son
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY
| | - Vijay K Yechoor
- Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Antoni Paul
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY
| |
Collapse
|
23
|
Mice Lacking Endoglin in Macrophages Show an Impaired Immune Response. PLoS Genet 2016; 12:e1005935. [PMID: 27010826 PMCID: PMC4806930 DOI: 10.1371/journal.pgen.1005935] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 02/24/2016] [Indexed: 12/26/2022] Open
Abstract
Endoglin is an auxiliary receptor for members of the TGF-β superfamily and plays an important role in the homeostasis of the vessel wall. Mutations in endoglin gene (ENG) or in the closely related TGF-β receptor type I ACVRL1/ALK1 are responsible for a rare dominant vascular dysplasia, the Hereditary Hemorrhagic Telangiectasia (HHT), or Rendu-Osler-Weber syndrome. Endoglin is also expressed in human macrophages, but its role in macrophage function remains unknown. In this work, we show that endoglin expression is triggered during the monocyte-macrophage differentiation process, both in vitro and during the in vivo differentiation of blood monocytes recruited to foci of inflammation in wild-type C57BL/6 mice. To analyze the role of endoglin in macrophages in vivo, an endoglin myeloid lineage specific knock-out mouse line (Engfl/flLysMCre) was generated. These mice show a predisposition to develop spontaneous infections by opportunistic bacteria. Engfl/flLysMCre mice also display increased survival following LPS-induced peritonitis, suggesting a delayed immune response. Phagocytic activity is impaired in peritoneal macrophages, altering one of the main functions of macrophages which contributes to the initiation of the immune response. We also observed altered expression of TGF-β1 target genes in endoglin deficient peritoneal macrophages. Overall, the altered immune activity of endoglin deficient macrophages could help to explain the higher rate of infectious diseases seen in HHT1 patients. Endoglin is a transmembrane protein and an auxiliary receptor for TGF-β with an important role in the homeostasis of the vessel wall. However, endoglin was originally identified as a human cell surface antigen expressed in a pre-B leukemic cell line. Mutations in ENG are responsible for the Hereditary Hemorrhagic Telangiectasia type 1 (HHT1) or Rendu-Osler-Weber syndrome. HHT is a rare disease, with a prevalence of 1/5,000 to 1/8,000. It is an autosomal dominant disorder characterized by a multisystemic vascular dysplasia, recurrent hemorrhages and arteriovenous malformations in internal organs. Interestingly, endoglin expression is also triggered during the monocyte-macrophage differentiation process. In our laboratory, we described that up-regulation of endoglin during in vitro differentiation of blood monocytes is age-dependent and impaired in monocytes from HHT patients, suggesting a role of endoglin in macrophages. In the present work, we first analyzed endoglin expression during differentiation of peripheral blood monocytes to macrophages under in vitro and in vivo conditions. Next, to investigate endoglin’s role in macrophage function in vivo, a myeloid-lineage specific endoglin knock-out mouse line was generated (Engfl/flLysMCre). Endoglin deficiency in macrophages predisposed animals to spontaneous infections and led to delayed endotoxin-induced mortality. Phagocytic activity by peritoneal macrophages was reduced in the absence of endoglin and altered expression of TGF-β target genes was consistent with an altered balance of TGF-β signaling. The results show a novel role for endoglin in mouse macrophages, which if analogous in human macrophages, may explain, at least in part, the increased infection rates seen in HHT patients.
Collapse
|
24
|
Sokolova EV, Karetin Y, Davydova VN, Byankina AO, Kalitnik AA, Bogdanovich LN, Yermak IM. Carrageenans effect on neutrophils alone and in combination with LPS in vitro. J Biomed Mater Res A 2016; 104:1603-9. [PMID: 26915063 DOI: 10.1002/jbm.a.35693] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 02/16/2016] [Accepted: 02/18/2016] [Indexed: 01/14/2023]
Abstract
Influence of sulfated red algal polysaccharides (κ-, λ-, and κ/β-carrageenans) and degraded derivative of κ/β-carrageenan on neutrophils/monocytes activation alone and in combination with lipopolysaccharide was investigated by means of determination of reactive oxygen species production, latex microparticles engulfment, total and extracellular myeloperoxidase induction and the analysis of silhouette and contour two-dimensional images of flattened cells. Carrageenans alone can activate neutrophils with much less potency than lipopolysaccharide (LPS) and the sulfation degree of carrageenans stipulates high activity in this role. On the other hand, carrageenans especially with low contents of sulfate groups are able to interfere with LPS in vitro resulting in reducing inter- and intracellular activation of neutrophils killing mechanisms. Further research is necessary to relate these findings to actions on the whole animal or human in vivo. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1603-1609, 2016.
Collapse
Affiliation(s)
- E V Sokolova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku, Vladivostok, 690022, Russia
| | - Y Karetin
- A.V. Zhirmunsky Institute of Marine Biology, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok, 690022, Russia.,Department of Cell Biology and Genetics School of Natural Sciences, Far Eastern Federal University, Vladivostok, 690022, Russia
| | - V N Davydova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku, Vladivostok, 690022, Russia
| | - A O Byankina
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku, Vladivostok, 690022, Russia
| | - A A Kalitnik
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku, Vladivostok, 690022, Russia
| | - L N Bogdanovich
- Medical Association of the Far East Branch of the Russian Academy of Sciences, St. Kirova, 95, Vladivostok, 690022, Russia
| | - I M Yermak
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku, Vladivostok, 690022, Russia
| |
Collapse
|
25
|
Salvador A, Sandgren KJ, Liang F, Thompson EA, Koup RA, Pedraz JL, Hernandez RM, Loré K, Igartua M. Design and evaluation of surface and adjuvant modified PLGA microspheres for uptake by dendritic cells to improve vaccine responses. Int J Pharm 2015; 496:371-81. [DOI: 10.1016/j.ijpharm.2015.10.037] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/06/2015] [Accepted: 10/09/2015] [Indexed: 10/22/2022]
|
26
|
Graczyk D, White RJ, Ryan KM. Involvement of RNA Polymerase III in Immune Responses. Mol Cell Biol 2015; 35:1848-59. [PMID: 25776554 PMCID: PMC4405649 DOI: 10.1128/mcb.00990-14] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 08/11/2014] [Accepted: 03/06/2015] [Indexed: 12/12/2022] Open
Abstract
Inflammation in the tumor microenvironment has many tumor-promoting effects. In particular, tumor-associated macrophages (TAMs) produce many cytokines which can support tumor growth by promoting survival of malignant cells, angiogenesis, and metastasis. Enhanced cytokine production by TAMs is tightly coupled with protein synthesis. In turn, translation of proteins depends on tRNAs, short abundant transcripts that are made by RNA polymerase III (Pol III). Here, we connect these facts by showing that stimulation of mouse macrophages with lipopolysaccharides (LPS) from the bacterial cell wall causes transcriptional upregulation of tRNA genes. The transcription factor NF-κB is a key transcription factor mediating inflammatory signals, and we report that LPS treatment causes an increased association of the NF-κB subunit p65 with tRNA genes. In addition, we show that p65 can directly associate with the Pol III transcription factor TFIIIB and that overexpression of p65 induces Pol III-dependent transcription. As a consequence of these effects, we show that inhibition of Pol III activity in macrophages restrains cytokine secretion and suppresses phagocytosis, two key functional characteristics of these cells. These findings therefore identify a radical new function for Pol III in the regulation of macrophage function which may be important for the immune responses associated with both normal and malignant cells.
Collapse
Affiliation(s)
- Damian Graczyk
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Robert J White
- Department of Biology, University of York, York, United Kingdom
| | - Kevin M Ryan
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| |
Collapse
|
27
|
Hansen JF, Bendtzen K, Boas M, Frederiksen H, Nielsen CH, Rasmussen ÅK, Feldt-Rasmussen U. Influence of phthalates on cytokine production in monocytes and macrophages: a systematic review of experimental trials. PLoS One 2015; 10:e0120083. [PMID: 25811352 PMCID: PMC4374770 DOI: 10.1371/journal.pone.0120083] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 01/19/2015] [Indexed: 12/13/2022] Open
Abstract
Background Phthalates are a group of endocrine disrupting chemicals suspected to influence the immune system. The aim of this systematic review is to summarise the present knowledge on the influence of phthalates on monocyte and macrophage production and secretion of cytokines, an influence which could affect both pro- and anti-inflammatory abilities of these cells. Strategy and Results A systematic search was performed in Medline, Embase and Toxline in June 2013, last updated 3rd of August 2014. Criteria used to select studies were described and published beforehand online on Prospero (http://www.crd.york.ac.uk/NIHR_PROSPERO, registration number CRD42013004236). In vivo, ex vivo and in vitro studies investigating the influence of phthalates on cytokine mRNA expression and cytokine secretion in animals and humans were included. A total of 11 reports, containing 12 studies, were found eligible for inclusion. In these, a total of four different phthalate diesters, six primary metabolites (phthalate monoesters) and seven different cytokines were investigated. Though all studies varied greatly in study design and species sources, four out of five studies that investigated di-2-ethylhexyl phthalate found an increased tumour necrosis factor-α secretion/production from monocytes or macrophages. A summary of cytokine measurements was not possible since few studies were comparable in study design and due to insufficient reporting of raw data for most of the included studies. Conclusion Results from this review have suggested that at least one phthalate (di-2-ethylhexyl phthalate) has the ability to enhance tumour necrosis factor-α production/secretion from monocytes/macrophages in vitro, but also observed ex vivo. Influence of other phthalates on other cytokines has only been investigated in few studies. Thus, in vitro studies on primary human monocytes/macrophages as well as more in vivo studies are needed to confirm or dispute these findings.
Collapse
Affiliation(s)
- Juliana Frohnert Hansen
- Department of Medical Endocrinology, PE 2132, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Bendtzen
- Institute for Inflammation Research, section 7521, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Malene Boas
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Hanne Frederiksen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Claus H. Nielsen
- Institute for Inflammation Research, section 7521, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Åse Krogh Rasmussen
- Department of Medical Endocrinology, PE 2132, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ulla Feldt-Rasmussen
- Department of Medical Endocrinology, PE 2132, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
28
|
Induction of long-term immunity against respiratory syncytial virus glycoprotein by an osmotic polymeric nanocarrier. Acta Biomater 2014; 10:4606-4617. [PMID: 25110285 DOI: 10.1016/j.actbio.2014.07.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 07/26/2014] [Accepted: 07/31/2014] [Indexed: 11/22/2022]
Abstract
Respiratory syncytial virus (RSV) is one of the most common causes of viral deaths in infants worldwide, yet no effective vaccines are available. Here, we report an osmotically active polysaccharide-based polysorbitol transporter (PST) prepared from sorbitol diacrylate and low-molecular-weight polyethylenimine (PEI) showing a potent, yet safe, adjuvant activity and acting as an effective delivery tool for RSV glycoprotein (RGp) antigen. PST showed no toxicity in vitro or in vivo, unlike PEI and the well-known experimental mucosal adjuvant cholera toxin (CT). PST formed nano-sized complexes with RGp by simple mixing, without affecting antigenic stability. The complexes exhibited negative surface charges that made them highly efficient in the selective activation of phagocytic cells and enhancement of phagocytic uptake. This resulted in an improved cytokine production and in the significant augmentation of RGp-specific antibody production, which persisted for over 200 days. Interestingly, PST/RGp enhanced phagocytic uptake owing to the osmotic property of PST and its negative zeta potential, suggesting that PST could selectively stimulate phagocytic cells, thereby facilitating a long-lived antigen-specific immune response, which was presumably further enhanced by the polysaccharide properties of PST.
Collapse
|
29
|
Gómez-González NE, García-García E, Montero J, García-Alcázar A, Meseguer J, García-Ayala A, Mulero V. Isolation of mast cells from the peritoneal exudate of the teleost fish gilthead sea bream (Sparus aurata L.). FISH & SHELLFISH IMMUNOLOGY 2014; 40:225-232. [PMID: 25047357 DOI: 10.1016/j.fsi.2014.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 07/02/2014] [Accepted: 07/07/2014] [Indexed: 06/03/2023]
Abstract
Inflammation is the first response of animals to infection or tissue damage. Sparus aurata (Perciformes) was the first fish species shown to possess histamine-containing mast cells at mucosal tissues. We report a separation protocol for obtaining highly enriched (over 95% purity) preparations of fish mast cells in high numbers (5-20 million mast cells per fish). The peritoneal exudate of S. aurata is composed of lymphocytes, acidophilic granulocytes, macrophages and mast cells. We separated the lymphocyte fraction through discontinuous density gradient centrifugation. The remaining cells were cultivated overnight in RPMI-1640 culture medium containing 5% fetal calf serum, which allowed macrophages to adhere to the cell culture flasks. Finally, acidophilic granulocytes were separated from the mast cells though a Magnetic-Activated Cell Separation (MACS) protocol, using a monoclonal antibody against these cells. The purity of mast cells-enriched fractions was analyzed by flow cytometry and by transmission electron microscopy. The functionality of purified mast cells was confirmed by the detection of histamine release by ELISA after stimulation with compound 48/80 and the induction of the pro-inflammatory cytokines IL-1β and IL-8 following stimulation with bacterial DNA. This fish mast cells separation protocol is a stepping stone for further studies addressing the evolution of vertebrate inflammatory mechanisms.
Collapse
Affiliation(s)
- Nuria Esther Gómez-González
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, IMIB-Arrixaca, Campus Universitario de Espinardo, 30100 Murcia, Spain
| | - Erick García-García
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, IMIB-Arrixaca, Campus Universitario de Espinardo, 30100 Murcia, Spain
| | - Jana Montero
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, IMIB-Arrixaca, Campus Universitario de Espinardo, 30100 Murcia, Spain
| | - Alicia García-Alcázar
- Instituto Español de Oceanografía, Planta de Cultivos Marinos, Ctra. de la Azohía s/n, 30860 Puerto de Mazarrón, Murcia, Spain
| | - José Meseguer
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, IMIB-Arrixaca, Campus Universitario de Espinardo, 30100 Murcia, Spain
| | - Alfonsa García-Ayala
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, IMIB-Arrixaca, Campus Universitario de Espinardo, 30100 Murcia, Spain
| | - Victoriano Mulero
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, IMIB-Arrixaca, Campus Universitario de Espinardo, 30100 Murcia, Spain.
| |
Collapse
|
30
|
Zheng PP, van der Weiden M, Kros JM. Fast tracking of co-localization of multiple markers by using the nanozoomer slide scanner and NDPViewer. J Cell Physiol 2014; 229:967-73. [PMID: 24374845 DOI: 10.1002/jcp.24538] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 12/12/2013] [Indexed: 12/20/2022]
Abstract
The detection of co-localization of immunohistochemical markers in tissues or cells requires the application of the confocal laser scanning microscope (CLSM) to multiple immunofluorescence (MIF) stainings. CLSM is operationally sophisticated but requires time-consuming procedures of imaging and reconstruction, and a professional operator is required for manipulation of the microscopic system. Therefore, this technique is less suitable for the examination of many samples in a short time. Moreover, the technique only allows imaging of selected areas of a sample at one time and is not practical for fast panoramic mapping and tracking of whole tissue sections. Here we show a powerful high-throughput and operationally simple histological approach using the Hamamatsu NDP slide scanner (Hamamatsu Nanozoomer 2.0HT) and its viewing platform (NDP.Viewer). The approach not only enables fast mapping and tracking of overlapping spots or regions stained with multiple markers, but also offers panoramic screening of whole tissue sections with fully electronic manipulation for the visualization and analysis of any individual regions.
Collapse
Affiliation(s)
- Ping-Pin Zheng
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | |
Collapse
|
31
|
Perry JA, Koteva K, Verschoor CP, Wang W, Bowdish DME, Wright GD. A macrophage-stimulating compound from a screen of microbial natural products. J Antibiot (Tokyo) 2014; 68:40-6. [DOI: 10.1038/ja.2014.83] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 05/20/2014] [Accepted: 05/28/2014] [Indexed: 12/21/2022]
|
32
|
Italiani P, Mazza EMC, Lucchesi D, Cifola I, Gemelli C, Grande A, Battaglia C, Bicciato S, Boraschi D. Transcriptomic profiling of the development of the inflammatory response in human monocytes in vitro. PLoS One 2014; 9:e87680. [PMID: 24498352 PMCID: PMC3912012 DOI: 10.1371/journal.pone.0087680] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 12/29/2013] [Indexed: 12/31/2022] Open
Abstract
Monocytes/macrophages are key players in all phases of physiological and pathological inflammation. To understanding the regulation of macrophage functional differentiation during inflammation, we designed an in vitro model that recapitulates the different phases of the reaction (recruitment, initiation, development, and resolution), based on human primary blood monocytes exposed to sequential changes in microenvironmental conditions. All reaction phases were profiled by transcriptomic microarray analysis. Distinct clusters of genes were identified that are differentially regulated through the different phases of inflammation. The gene sets defined by GSEA analysis revealed that the inflammatory phase was enriched in inflammatory pathways, while the resolution phase comprised pathways related to metabolism and gene rearrangement. By comparing gene clusters differentially expressed in monocytes vs. M1 and vs. M2 macrophages extracted from an in-house created meta-database, it was shown that cells in the model resemble M1 during the inflammatory phase and M2 during resolution. The validation of inflammatory and transcriptional factors by qPCR and ELISA confirmed the transcriptomic profiles in the different phases of inflammation. The accurate description of the development of the human inflammatory reaction provided by this in vitro kinetic model can help in identifying regulatory mechanisms in physiological conditions and during pathological derangements.
Collapse
Affiliation(s)
- Paola Italiani
- Institute of Biomedical Technologies, National Research Council, Pisa/Segrate, Italy
| | - Emilia M. C. Mazza
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Davide Lucchesi
- Institute of Biomedical Technologies, National Research Council, Pisa/Segrate, Italy
| | - Ingrid Cifola
- Institute of Biomedical Technologies, National Research Council, Pisa/Segrate, Italy
| | - Claudia Gemelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alexis Grande
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Battaglia
- Institute of Biomedical Technologies, National Research Council, Pisa/Segrate, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Segrate, Italy
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Diana Boraschi
- Institute of Biomedical Technologies, National Research Council, Pisa/Segrate, Italy
- Institute of Protein Biochemistry, National Research Council, Naples, Italy
| |
Collapse
|
33
|
Nich C, Takakubo Y, Pajarinen J, Ainola M, Salem A, Sillat T, Rao AJ, Raska M, Tamaki Y, Takagi M, Konttinen YT, Goodman SB, Gallo J. Macrophages-Key cells in the response to wear debris from joint replacements. J Biomed Mater Res A 2013; 101:3033-45. [PMID: 23568608 PMCID: PMC3775910 DOI: 10.1002/jbm.a.34599] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 11/16/2012] [Accepted: 01/12/2013] [Indexed: 12/14/2022]
Abstract
The generation of wear debris is an inevitable result of normal usage of joint replacements. Wear debris particles stimulate local and systemic biological reactions resulting in chronic inflammation, periprosthetic bone destruction, and eventually, implant loosening, and revision surgery. The latter may be indicated in up to 15% patients in the decade following the arthroplasty using conventional polyethylene. Macrophages play multiple roles in both inflammation and in maintaining tissue homeostasis. As sentinels of the innate immune system, they are central to the initiation of this inflammatory cascade, characterized by the release of proinflammatory and pro-osteoclastic factors. Similar to the response to pathogens, wear particles elicit a macrophage response, based on the unique properties of the cells belonging to this lineage, including sensing, chemotaxis, phagocytosis, and adaptive stimulation. The biological processes involved are complex, redundant, both local and systemic, and highly adaptive. Cells of the monocyte/macrophage lineage are implicated in this phenomenon, ultimately resulting in differentiation and activation of bone resorbing osteoclasts. Simultaneously, other distinct macrophage populations inhibit inflammation and protect the bone-implant interface from osteolysis. Here, the current knowledge about the physiology of monocyte/macrophage lineage cells is reviewed. In addition, the pattern and consequences of their interaction with wear debris and the recent developments in this field are presented.
Collapse
Affiliation(s)
- Christophe Nich
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California; Laboratoire de Biomécanique et Biomatériaux Ostéo-Articulaires-UMR CNRS 7052, Faculté de Médecine-Université Paris 7, Paris, France; Department of Orthopaedic Surgery, European Teaching Hospital, Assistance Publique-Hôpitaux de Paris-Université Paris 5, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Acedo SC, Gambero S, Cunha FGP, Lorand-Metze I, Gambero A. Participation of leptin in the determination of the macrophage phenotype: an additional role in adipocyte and macrophage crosstalk. In Vitro Cell Dev Biol Anim 2013; 49:473-8. [PMID: 23708919 DOI: 10.1007/s11626-013-9629-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 04/28/2013] [Indexed: 11/25/2022]
Abstract
Macrophages develop into specialized cell types with special functional properties, depending on locally produced stimuli. Adipose tissue macrophages present particular characteristics, such as the M2 cell phenotype, and produce cytokines and chemokines usually produced by M1 cells. Our aim was to study the role of leptin, which is an adipokine produced and released by adipocytes, in the induction of these characteristics in macrophages found in the adipose tissue. Human CD14(+) cells were obtained and maintained in culture with IFN-γ (classical M1 phenotype), IL-4 (alternative M2 phenotype) or leptin for 5 d. Surface marker expression was then analyzed by cytometry. In addition, the release of tumor necrosis factor alpha (TNF-α), interleukin (IL)-6, IL-1β, IL-10, IL-1ra, MCP-1, MIP-1α, and RANTES was quantified by ELISA after an LPS stimulus, in the culture supernatant. Macrophages exposed to leptin in culture expressed surface markers that were more similar to the M2 phenotype, but they were able to produce TNF-α, IL-6, IL-1β, IL-1ra, IL-10, MCP-1, and MIP-1α, as observed for M1 cells. Results suggest that leptin strongly contributes to the phenotype observed in macrophages found in adipose tissue.
Collapse
Affiliation(s)
- Simone Coghetto Acedo
- Clinical Pharmacology and Gastroenterology Unit, São Francisco University Medical School, Av. São Francisco de Assis 218, 12916-900, Bragança Paulista, SP, Brazil
| | | | | | | | | |
Collapse
|
35
|
Chen X, Wang Y, Li Q, Tsai S, Thomas A, Shizuru JA, Cao TM. Pathways analysis of differential gene expression induced by engrafting doses of total body irradiation for allogeneic bone marrow transplantation in mice. Immunogenetics 2013; 65:597-607. [PMID: 23703256 DOI: 10.1007/s00251-013-0710-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Accepted: 05/04/2013] [Indexed: 01/13/2023]
Abstract
A major challenge in allogeneic bone marrow (BM) transplantation is overcoming engraftment resistance to avoid the clinical problem of graft rejection. Identifying gene pathways that regulate BM engraftment may reveal molecular targets for overcoming engraftment barriers. Previously, we developed a mouse model of BM transplantation that utilizes recipient conditioning with non-myeloablative total body irradiation (TBI). We defined TBI doses that lead to graft rejection, that conversely are permissive for engraftment, and mouse strain variation with regards to the permissive TBI dose. We now report gene expression analysis, using Agilent Mouse 8x60K microarrays, in spleens of mice conditioned with varied TBI doses for correlation to the expected engraftment phenotype. The spleens of mice given engrafting doses of TBI, compared with non-engrafting TBI doses, demonstrated substantially broader gene expression changes, significant at the multiple testing-corrected P <0.05 level and with fold change ≥2. Functional analysis revealed significant enrichment for a down-regulated canonical pathway involving B-cell development. Genes enriched in this pathway suggest that suppressing donor antigen processing and presentation may be pivotal effects conferred by TBI to enable engraftment. Regardless of TBI dose and recipient mouse strain, pervasive genomic changes related to inflammation was observed and reflected by significant enrichment for canonical pathways and association with upstream regulators. These gene expression changes suggest that macrophage and complement pathways may be targeted to overcome engraftment barriers. These exploratory results highlight gene pathways that may be important in mediating BM engraftment resistance.
Collapse
Affiliation(s)
- Xinjian Chen
- Department of Pathology, University of Utah, Salt Lake City, UT 84132, USA
| | | | | | | | | | | | | |
Collapse
|