1
|
Usman S, Mondal AC. Menopause triggers microglia-associated neuroinflammation in Parkinson's disease. Brain Res 2025; 1859:149649. [PMID: 40250746 DOI: 10.1016/j.brainres.2025.149649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/30/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Microglia, the immune cells of brain, can drive neurodegenerative diseases like Parkinson's disease (PD). The resting microglia can polarize into two extremes, either proinflammatory M1 or anti-inflammatory M2 phenotype under a specific microenvironment. Different transcriptional factors and the release of various cytokines characterize these states. The released proinflammatory markers from M1 microglia lead to neuroinflammation that ultimately causes irreversible loss of dopaminergic neurons in PD patients, on the contrary, the M2 microglia possess neuroprotective activity. PD is caused by aggregation and misfolding of α-synuclein in the affected dopaminergic neurons. The misfolded α-synuclein is cytotoxic and can propagate like a prion from one cell to the other, acting like a template, that can initiate the conversion of normal proteins into abnormal conformation. The extracellular α-synuclein can interact and polarize the microglia into the M1 phenotype resulting in inflammation, thereby driving the progression of PD. The progression of neuroinflammation-mediated neurodegeneration in PD is seen higher in menopausal women; likely due to the low circulating estrogen levels. Estrogen hormones possess neuroprotective activity, and one of the ways is that they can polarize the microglia into M2 phenotypes and reduce α-synuclein-mediated microglial activation. A detailed understanding of the signaling mechanisms underlying microglial polarization between M1 and M2 phenotypes is crucial for identifying druggable targets to reduce PD symptoms, including in menopausal women.
Collapse
Affiliation(s)
- Sehar Usman
- Cellular and Molecular Neurobiology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Amal Chandra Mondal
- Cellular and Molecular Neurobiology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
2
|
Di Francesco AM, Pasciuto G, Verrecchia E, Sicignano LL, Gerardino L, Massaro MG, Urbani A, Manna R. Sarcoidosis and Cancer: The Role of the Granulomatous Reaction as a Double-Edged Sword. J Clin Med 2024; 13:5232. [PMID: 39274446 PMCID: PMC11396756 DOI: 10.3390/jcm13175232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Background/Objectives: The relationship between sarcoidosis and the occurrence of neoplasia deserves to be investigated, but this relation has been observed in different and heterogeneous populations, leading to conflicting data. To clarify the causal relationship between these two diseases, different risk factors (e.g., smoking), concurrent comorbidities, corticosteroid therapy, and metastasis development-as an expression of cancer aggressiveness-were investigated. Methods: In a retrospective study on 287 sarcoidosis outpatients at the Pneumological Department of the Gemelli Foundation (Rome, Italy) between 2000 and 2024, the diagnosis of cancer was recorded in 36 subjects (12.5%). Results: The reciprocal timeline of the diseases showed three different scenarios: (1) cancer preceding sarcoidosis or sarcoid-like reactions (63.8%); (2) cancer arising after sarcoidosis diagnosis (8.3%); and (3) sarcoidosis accompanying the onset of malignancy (27.8%). Only two subjects with sarcoidosis and cancer showed metastasis, and one of them was affected by lymphoma. Conclusions: These data suggest that granulomatous inflammation due to sarcoidosis may assume an ambivalent role as a "double-edged sword", according to the M1/M2 macrophage polarization model: it represents a protective shield, preventing the formation of metastasis through the induction of immune surveillance against cancer while, on the other hand, it can be a risk factor for carcinogenesis due to the persistence of a chronic active inflammatory status. Low-dose steroid treatment was administered in only 31.6% of the cancer-sarcoidosis subjects for less than six months to control inflammation activity, with no promotive effect on carcinogenesis observed.
Collapse
Affiliation(s)
- Angela Maria Di Francesco
- Periodic Fever and Rare Diseases Research Centre, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Giuliana Pasciuto
- Complex Pneumology Operational Unit, A. Gemelli Policlinic Foundation IRCCS, 00168 Rome, Italy
| | - Elena Verrecchia
- Department of Aging, Orthopaedic and Rheumatological Sciences, A. Gemelli Policlinic Foundation IRCCS, 00168 Rome, Italy
| | - Ludovico Luca Sicignano
- Department of Aging, Orthopaedic and Rheumatological Sciences, A. Gemelli Policlinic Foundation IRCCS, 00168 Rome, Italy
| | - Laura Gerardino
- Department of Aging, Orthopaedic and Rheumatological Sciences, A. Gemelli Policlinic Foundation IRCCS, 00168 Rome, Italy
| | - Maria Grazia Massaro
- Periodic Fever and Rare Diseases Research Centre, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Andrea Urbani
- Department of Chemistry, Biochemistry and Molecular Biology, A. Gemelli Policlinic Foundation IRCCS, 00168 Rome, Italy
| | - Raffaele Manna
- Periodic Fever and Rare Diseases Research Centre, Catholic University of Sacred Heart, 00168 Rome, Italy
| |
Collapse
|
3
|
Ashrafizadeh M, Aref AR, Sethi G, Ertas YN, Wang L. Natural product/diet-based regulation of macrophage polarization: Implications in treatment of inflammatory-related diseases and cancer. J Nutr Biochem 2024; 130:109647. [PMID: 38604457 DOI: 10.1016/j.jnutbio.2024.109647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
Macrophages are phagocytic cells with important physiological functions, including the digestion of cellular debris, foreign substances, and microbes, as well as tissue development and homeostasis. The tumor microenvironment (TME) shapes the aggressiveness of cancer, and the biological and cellular interactions in this complicated space can determine carcinogenesis. TME can determine the progression, biological behavior, and therapy resistance of human cancers. The macrophages are among the most abundant cells in the TME, and their functions and secretions can determine tumor progression. The education of macrophages to M2 polarization can accelerate cancer progression, and therefore, the re-education and reprogramming of these cells is promising. Moreover, macrophages can cause inflammation in aggravating pathological events, including cardiovascular diseases, diabetes, and neurological disorders. The natural products are pleiotropic and broad-spectrum functional compounds that have been deployed as ideal alternatives to conventional drugs in the treatment of cancer. The biological and cellular interactions in the TME can be regulated by natural products, and for this purpose, they enhance the M1 polarization of macrophages, and in addition to inhibiting proliferation and invasion, they impair the chemoresistance. Moreover, since macrophages and changes in the molecular pathways in these cells can cause inflammation, the natural products impair the pro-inflammatory function of macrophages to prevent the pathogenesis and progression of diseases. Even a reduction in macrophage-mediated inflammation can prevent organ fibrosis. Therefore, natural product-mediated macrophage targeting can alleviate both cancerous and non-cancerous diseases.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA; Department of Translational Sciences, Xsphera Biosciences Inc., Boston, Massachusetts, USA
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Türkiye; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Türkiye.
| | - Lu Wang
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
| |
Collapse
|
4
|
Li H, Li B, Zheng Y. Role of microglia/macrophage polarisation in intraocular diseases (Review). Int J Mol Med 2024; 53:45. [PMID: 38551157 PMCID: PMC10998719 DOI: 10.3892/ijmm.2024.5369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/05/2024] [Indexed: 04/02/2024] Open
Abstract
Macrophages form a crucial component of the innate immune system, and their activation is indispensable for various aspects of immune and inflammatory processes, tissue repair, and maintenance of the balance of the body's state. Macrophages are found in all ocular tissues, spanning from the front surface, including the cornea, to the posterior pole, represented by the choroid/sclera. The neural retina is also populated by specialised resident macrophages called microglia. The plasticity of microglia/macrophages allows them to adopt different activation states in response to changes in the tissue microenvironment. When exposed to various factors, microglia/macrophages polarise into distinct phenotypes, each exhibiting unique characteristics and roles. Furthermore, extensive research has indicated a close association between microglia/macrophage polarisation and the development and reversal of various intraocular diseases. The present article provides a review of the recent findings on the association between microglia/macrophage polarisation and ocular pathological processes (including autoimmune uveitis, optic neuritis, sympathetic ophthalmia, retinitis pigmentosa, glaucoma, proliferative vitreoretinopathy, subretinal fibrosis, uveal melanoma, ischaemic optic neuropathy, retinopathy of prematurity and choroidal neovascularization). The paradoxical role of microglia/macrophage polarisation in retinopathy of prematurity is also discussed. Several studies have shown that microglia/macrophages are involved in the pathology of ocular diseases. However, it is required to further explore the relevant mechanisms and regulatory processes. The relationship between the functional diversity displayed by microglia/macrophage polarisation and intraocular diseases may provide a new direction for the treatment of intraocular diseases.
Collapse
Affiliation(s)
- Haoran Li
- School of Opthalmology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Biao Li
- School of Opthalmology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Yanlin Zheng
- School of Opthalmology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
5
|
Ctortecka C, Clark NM, Boyle B, Seth A, Mani DR, Udeshi ND, Carr SA. Automated single-cell proteomics providing sufficient proteome depth to study complex biology beyond cell type classifications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.20.576369. [PMID: 38328197 PMCID: PMC10849471 DOI: 10.1101/2024.01.20.576369] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Mass spectrometry (MS)-based single-cell proteomics (SCP) has gained massive attention as a viable complement to other single cell approaches. The rapid technological and computational advances in the field have pushed the boundaries of sensitivity and throughput. However, reproducible quantification of thousands of proteins within a single cell at reasonable proteome depth to characterize biological phenomena remains a challenge. To address some of those limitations we present a combination of fully automated single cell sample preparation utilizing a dedicated chip within the picolitre dispensing robot, the cellenONE. The proteoCHIP EVO 96 can be directly interfaced with the Evosep One chromatographic system for in-line desalting and highly reproducible separation with a throughput of 80 samples per day. This, in combination with the Bruker timsTOF MS instruments, demonstrates double the identifications without manual sample handling. Moreover, relative to standard high-performance liquid chromatography, the Evosep One separation provides further 2-fold improvement in protein identifications. The implementation of the newest generation timsTOF Ultra with our proteoCHIP EVO 96-based sample preparation workflow reproducibly identifies up to 4,000 proteins per single HEK-293T without a carrier or match-between runs. Our current SCP depth spans over 4 orders of magnitude and identifies over 50 biologically relevant ubiquitin ligases. We complement our highly reproducible single-cell proteomics workflow to profile hundreds of lipopolysaccharide (LPS)-perturbed THP-1 cells and identified key regulatory proteins involved in interleukin and interferon signaling. This study demonstrates that the proteoCHIP EVO 96-based SCP sample preparation with the timsTOF Ultra provides sufficient proteome depth to study complex biology beyond cell-type classifications.
Collapse
Affiliation(s)
- Claudia Ctortecka
- Broad Institute of MIT and Harvard, 415 Main Street, 02142 Cambridge, MA, USA
| | - Natalie M. Clark
- Broad Institute of MIT and Harvard, 415 Main Street, 02142 Cambridge, MA, USA
| | - Brian Boyle
- Broad Institute of MIT and Harvard, 415 Main Street, 02142 Cambridge, MA, USA
| | - Anjali Seth
- Cellenion SASU, 60F avenue Rockefeller, 69008 Lyon, France
| | - D. R. Mani
- Broad Institute of MIT and Harvard, 415 Main Street, 02142 Cambridge, MA, USA
| | - Namrata D. Udeshi
- Broad Institute of MIT and Harvard, 415 Main Street, 02142 Cambridge, MA, USA
| | - Steven A. Carr
- Broad Institute of MIT and Harvard, 415 Main Street, 02142 Cambridge, MA, USA
| |
Collapse
|
6
|
Barroso M, Monaghan MG, Niesner R, Dmitriev RI. Probing organoid metabolism using fluorescence lifetime imaging microscopy (FLIM): The next frontier of drug discovery and disease understanding. Adv Drug Deliv Rev 2023; 201:115081. [PMID: 37647987 PMCID: PMC10543546 DOI: 10.1016/j.addr.2023.115081] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/20/2023] [Accepted: 08/24/2023] [Indexed: 09/01/2023]
Abstract
Organoid models have been used to address important questions in developmental and cancer biology, tissue repair, advanced modelling of disease and therapies, among other bioengineering applications. Such 3D microenvironmental models can investigate the regulation of cell metabolism, and provide key insights into the mechanisms at the basis of cell growth, differentiation, communication, interactions with the environment and cell death. Their accessibility and complexity, based on 3D spatial and temporal heterogeneity, make organoids suitable for the application of novel, dynamic imaging microscopy methods, such as fluorescence lifetime imaging microscopy (FLIM) and related decay time-assessing readouts. Several biomarkers and assays have been proposed to study cell metabolism by FLIM in various organoid models. Herein, we present an expert-opinion discussion on the principles of FLIM and PLIM, instrumentation and data collection and analysis protocols, and general and emerging biosensor-based approaches, to highlight the pioneering work being performed in this field.
Collapse
Affiliation(s)
- Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Michael G Monaghan
- Department of Mechanical, Manufacturing and Biomedical Engineering, Trinity College Dublin, Dublin 02, Ireland
| | - Raluca Niesner
- Dynamic and Functional In Vivo Imaging, Freie Universität Berlin and Biophysical Analytics, German Rheumatism Research Center, Berlin, Germany
| | - Ruslan I Dmitriev
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, C. Heymanslaan 10, 9000 Ghent, Belgium; Ghent Light Microscopy Core, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
7
|
Inhibition of Chitinase-3-like-1 expression by K284 ameliorates lipopolysaccharide-induced acute liver injury through down regulation of CXCL3. Int Immunopharmacol 2023. [DOI: 10.1016/j.intimp.2023.109877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
|
8
|
Li S, Wang C, Tang YD, Qin L, Chen T, Wang S, Bai Y, Cai X, Wang S. Interaction between Porcine Alveolar Macrophage-Tang Cells and Streptococcus suis Strains of Different Virulence: Phagocytosis and Apoptosis. Microorganisms 2023; 11:microorganisms11010160. [PMID: 36677452 PMCID: PMC9863715 DOI: 10.3390/microorganisms11010160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/21/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Streptococcus suis is an important swine bacterial pathogen that activates macrophages to secrete inflammatory cytokines. Primary porcine alveolar macrophages (PAMs) are inconvenient to obtain, but it is unknown whether immortalized PAM-Tang cells can replace them as a better cell model for the study of the interaction between S. suis and macrophages. In this study, the phagocytic integrity, polarization, and pro-inflammatory cytokine secretion of PAM-Tang cells were confirmed by live-cell imaging, electron microscopy, confocal microscopy, and ELISA. Interestingly, the S. suis serotype 9 avirulent strain W7119 induced higher levels of adhesion and pro-inflammatory cytokines in PAM-Tang cells than the S. suis serotype 2 virulent strain 700794. Prolonged incubation with S. suis caused more cytotoxic cell damage, and the virulent strain induced higher levels of cytotoxicity to PAM-Tang cells. The virulent strain also induced higher levels of apoptosis in PAM-Tang cells, as shown by terminal deoxynucleotidyl transferase (TdT)-mediated dUTP-biotin nick end labeling (TUNEL) assay. In addition, it is the first report of virulent and avirulent S. suis inducing PAM-Tang polarization towards pro-inflammatory M1 macrophages and p53- and caspase-dependent apoptosis in PAMs. Taken together, this study contributes to a better understand of interactions between macrophages and S. suis isolates of different virulence, and confirms that PAM-Tang cells provide a long-term, renewable resource for investigating macrophage infections with bacteria.
Collapse
Affiliation(s)
- Siqi Li
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Chunsheng Wang
- College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Yan-Dong Tang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Lei Qin
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Tianfeng Chen
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Shanghui Wang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Yuanzhe Bai
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Xuehui Cai
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
- Correspondence: (X.C.); (S.W.)
| | - Shujie Wang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
- Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin 150069, China
- Correspondence: (X.C.); (S.W.)
| |
Collapse
|
9
|
Synovial Fluid in Knee Osteoarthritis Extends Proinflammatory Niche for Macrophage Polarization. Cells 2022; 11:cells11244115. [PMID: 36552878 PMCID: PMC9776803 DOI: 10.3390/cells11244115] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Macrophage polarization is a steering factor of osteoarthritis (OA) progression. Synovial fluid (SF) obtained from OA patients with different Kellgren-Lawrence grades (KL grades) holds several proinflammatory factors and was hypothesized to induce macrophage differentiation and polarization by providing the needed microenvironment. U937 cells and peripheral-blood-mononuclear-cell-derived monocytes (PBMC-derived CD14+ cells) were induced with SFs of progressive KL grades for 48 h, and the status of the differentiated cells was evaluated by cell surface markers representing M1 and M2 macrophage phenotypes. Functional viability assessment of the differentiated cells was performed by cytokine estimation. The fraction of macrophages and their phenotypes were estimated by immunophenotyping of SF-isolated cells of different KL grades. A grade-wise proteome analysis of SFs was performed in search of the factors which are influential in macrophage differentiation and polarization. In the assay on U937 cells, induction with SF of KL grade III and IV showed a significant increase in M1 type (CD86+). The percentage of M2 phenotype (CD163+) was significantly higher after the induction with SF of KL grade II. A Significantly higher M1/M2 ratio was estimated in the cells induced with KL grade III and IV. The cell differentiation pattern in the assay on PBMC-derived CD14+ cells showed a grade-wise decline in both M1 (CD11C+, CD86+) and M2 phenotype (CD163+). Cytokine estimation specific to M1 (TNF-α, IL-6, IL-1β, IFN-γ) and M2 (IL-4 and IL-10) macrophages corelated with the differentiation pattern in the U937 cell assay, while it did not reveal any significant changes in the PBMC-derived CD14+ cells assay. SF cells' immunophenotyping showed the highest percentage of CD14+ macrophages in KL grade II; CD86+ and CD163+ cells were minimal in all KL grades' SFs. The proteome analysis revealed significantly expressed MIF, CAPG/MCP, osteopontin, and RAS-related RAB proteins in KL grade III and IV samples, which are linked with macrophages' movement, polarization, and migration-behavior. In conclusion, this study demonstrated that SF in OA joints acts as a niche and facilitates M1 phenotype polarization by providing a proinflammatory microenvironment.
Collapse
|
10
|
Loss of selenoprotein W in murine macrophages alters the hierarchy of selenoprotein expression, redox tone, and mitochondrial functions during inflammation. Redox Biol 2022; 59:102571. [PMID: 36516721 PMCID: PMC9762199 DOI: 10.1016/j.redox.2022.102571] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022] Open
Abstract
Macrophages play a pivotal role in mediating inflammation and subsequent resolution of inflammation. The availability of selenium as a micronutrient and the subsequent biosynthesis of selenoproteins, containing the 21st amino acid selenocysteine (Sec), are important for the physiological functions of macrophages. Selenoproteins regulate the redox tone in macrophages during inflammation, the early onset of which involves oxidative burst of reactive oxygen and nitrogen species. SELENOW is a highly expressed selenoprotein in bone marrow-derived macrophages (BMDMs). Beyond its described general role as a thiol and peroxide reductase and as an interacting partner for 14-3-3 proteins, its cellular functions, particularly in macrophages, remain largely unknown. In this study, we utilized Selenow knock-out (KO) murine bone marrow-derived macrophages (BMDMs) to address the role of SELENOW in inflammation following stimulation with bacterial endotoxin lipopolysaccharide (LPS). RNAseq-based temporal analyses of expression of selenoproteins and the Sec incorporation machinery genes suggested no major differences in the selenium utilization pathway in the Selenow KO BMDMs compared to their wild-type counterparts. However, selective enrichment of oxidative stress-related selenoproteins and increased ROS in Selenow-/- BMDMs indicated anomalies in redox homeostasis associated with hierarchical expression of selenoproteins. Selenow-/- BMDMs also exhibited reduced expression of arginase-1, a key enzyme associated with anti-inflammatory (M2) phenotype necessary to resolve inflammation, along with a significant decrease in efferocytosis of neutrophils that triggers pathways of resolution. Parallel targeted metabolomics analysis also confirmed an impairment in arginine metabolism in Selenow-/- BMDMs. Furthermore, Selenow-/- BMDMs lacked the ability to enhance characteristic glycolytic metabolism during inflammation. Instead, these macrophages atypically relied on oxidative phosphorylation for energy production when glucose was used as an energy source. These findings suggest that SELENOW expression in macrophages may have important implications on cellular redox processes and bioenergetics during inflammation and its resolution.
Collapse
|
11
|
Fu SP, Chen SY, Pang QM, Zhang M, Wu XC, Wan X, Wan WH, Ao J, Zhang T. Advances in the research of the role of macrophage/microglia polarization-mediated inflammatory response in spinal cord injury. Front Immunol 2022; 13:1014013. [PMID: 36532022 PMCID: PMC9751019 DOI: 10.3389/fimmu.2022.1014013] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/18/2022] [Indexed: 12/04/2022] Open
Abstract
It is often difficult to regain neurological function following spinal cord injury (SCI). Neuroinflammation is thought to be responsible for this failure. Regulating the inflammatory response post-SCI may contribute to the recovery of neurological function. Over the past few decades, studies have found that macrophages/microglia are one of the primary effector cells in the inflammatory response following SCI. Growing evidence has documented that macrophages/microglia are plastic cells that can polarize in response to microenvironmental signals into M1 and M2 macrophages/microglia. M1 produces pro-inflammatory cytokines to induce inflammation and worsen tissue damage, while M2 has anti-inflammatory activities in wound healing and tissue regeneration. Recent studies have indicated that the transition from the M1 to the M2 phenotype of macrophage/microglia supports the regression of inflammation and tissue repair. Here, we will review the role of the inflammatory response and macrophages/microglia in SCI and repair. In addition, we will discuss potential molecular mechanisms that induce macrophage/microglia polarization, with emphasis on neuroprotective therapies that modulate macrophage/microglia polarization, which will provide new insights into therapeutic strategies for SCI.
Collapse
Affiliation(s)
- Sheng-Ping Fu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Si-Yu Chen
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qi-Ming Pang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Meng Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiang-Chong Wu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xue Wan
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Wei-Hong Wan
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jun Ao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China,The Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,*Correspondence: Tao Zhang,
| |
Collapse
|
12
|
Litta F, Papait A, Lucchetti D, Farigu S, Parello A, Tenore CR, Campennì P, Silini AR, Giustiniani MC, Parolini O, Sgambato A, Ratto C. The pathogenesis of cryptoglandular anal fistula: New insight into the immunological profile. Colorectal Dis 2022; 24:1567-1575. [PMID: 35916639 PMCID: PMC10087133 DOI: 10.1111/codi.16290] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/08/2022] [Accepted: 07/27/2022] [Indexed: 01/07/2023]
Abstract
AIM The aetiology of cryptoglandular anal fistula (AF) is poorly understood. Evidence suggests that persistence and/or recurrence of the disease is more related to inflammatory than infectious factors. The aim of this study was to investigate the immune profile of cryptoglandular AF and to perform a histopathological characterization. METHOD Fistulectomy was performed in all patients; healthy ischioanal fat from the same patients was used as a control. Samples were evaluated by the Luminex xMAP system for the detection of 27 analytes. AF tissues were analysed using immunofluorescence. Staining was performed using primary antibodies to identify M1 inflammatory and M2 anti-inflammatory macrophages. Selective staining of total T lymphocytes and different T lymphocyte subsets was performed. RESULTS Twenty patients with AF underwent a fistulectomy. Specific cytokine pathways differentiated AF from healthy tissue: pro-inflammatory cytokines interleukin (IL)-1β, IL-4, IL-8 and IL-17 and the anti-inflammatory cytokine IL-10 were overexpressed in AF compared with controls. Chemokines involved in macrophage recruitment (CCL2, CCL3, CCL4) were higher in AF than in healthy fatty tissue. Moreover, we showed that Tc17 cells characterize AF patients, thus confirming the enzyme-linked immunosorbent assay data. Furthermore, elevated infiltration of CD68+ myeloid cells and a reduction of the M1/M2 ratio characterize AF patients. CONCLUSION A combination of inflammatory cytokines, chemokines and growth factors reside in the wound microenvironment of AF patients. For the first time an important prevalence of Tc17 cells and a reduction in the M1/M2 ratio was observed, thus suggesting new insights into the immunological characterization of AF patients.
Collapse
Affiliation(s)
- Francesco Litta
- Proctology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Andrea Papait
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS, Rome, Italy
| | - Donatella Lucchetti
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Serafina Farigu
- Centro di Ricerca E. Menni, Fondazione Poliambulanza, Brescia, Italy
| | - Angelo Parello
- Proctology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Claudio Ricciardi Tenore
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Paola Campennì
- Proctology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | | | - Ornella Parolini
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS, Rome, Italy
| | - Alessandro Sgambato
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Carlo Ratto
- Proctology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
13
|
Pantazi P, Clements T, Venø M, Abrahams VM, Holder B. Distinct non-coding RNA cargo of extracellular vesicles from M1 and M2 human primary macrophages. J Extracell Vesicles 2022; 11:e12293. [PMID: 36544271 PMCID: PMC9772496 DOI: 10.1002/jev2.12293] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 11/13/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Macrophages are important antigen presenting cells which can release extracellular vesicles (EVs) carrying functional cargo including non-coding RNAs. Macrophages can be broadly classified into M1 'classical' and M2 'alternatively-activated' macrophages. M1 macrophages have been linked with inflammation-associated pathologies, whereas a switch towards an M2 phenotype indicates resolution of inflammation and tissue regeneration. Here, we provide the first comprehensive analysis of the small RNA cargo of EVs from human M1 and M2 primary macrophages. Using small RNA sequencing, we identified several types of small non-coding RNAs in M1 and M2 macrophage EVs including miRNAs, isomiRs, tRNA fragments, piRNA, snRNA, snoRNA and Y-RNA fragments. Distinct differences were observed between M1 and M2 EVs, with higher relative abundance of miRNAs, and lower abundance of tRNA fragments in M1 compared to M2 EVs. MicroRNA-target enrichment analysis identified several gene targets involved in gene expression and inflammatory signalling pathways. EVs were also enriched in tRNA fragments, primarily originating from the 5' end or the internal region of the full length tRNAs, many of which were differentially abundant in M1 and M2 EVs. Similarly, several other small non-coding RNAs, namely snRNAs, snoRNAs and Y-RNA fragments, were differentially enriched in M1 and M2 EVs; we discuss their putative roles in macrophage EVs. In conclusion, we show that M1 and M2 macrophages release EVs with distinct RNA cargo, which has the potential to contribute to the unique effect of these cell subsets on their microenvironment.
Collapse
Affiliation(s)
- Paschalia Pantazi
- Institute of Reproductive and Developmental BiologyDepartment of Metabolism, Digestion, and ReproductionImperial College LondonLondonUK
| | - Toby Clements
- Institute of Reproductive and Developmental BiologyDepartment of Metabolism, Digestion, and ReproductionImperial College LondonLondonUK
| | | | - Vikki M. Abrahams
- Department of ObstetricsGynecology and Reproductive SciencesYale School of MedicineNew HavenConnecticutUSA
| | - Beth Holder
- Institute of Reproductive and Developmental BiologyDepartment of Metabolism, Digestion, and ReproductionImperial College LondonLondonUK
| |
Collapse
|
14
|
Zhang G, Luo W, Yang W, Li S, Li D, Zeng Y, Li Y. The importance of the
IL
‐1 family of cytokines in nanoimmunosafety and nanotoxicology. WIRES NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1850. [DOI: 10.1002/wnan.1850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 08/03/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022]
Affiliation(s)
- Guofang Zhang
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Wenhe Luo
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Wenjie Yang
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Su Li
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Dongjie Li
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Yanqiao Zeng
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Yang Li
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| |
Collapse
|
15
|
Greco MF, Rizzuto AS, Zarà M, Cafora M, Favero C, Solazzo G, Giusti I, Adorni MP, Zimetti F, Dolo V, Banfi C, Ferri N, Sirtori CR, Corsini A, Barbieri SS, Pistocchi A, Bollati V, Macchi C, Ruscica M. PCSK9 Confers Inflammatory Properties to Extracellular Vesicles Released by Vascular Smooth Muscle Cells. Int J Mol Sci 2022; 23:13065. [PMID: 36361853 PMCID: PMC9655172 DOI: 10.3390/ijms232113065] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/21/2022] [Accepted: 10/23/2022] [Indexed: 10/20/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) are key participants in both early- and late-stage atherosclerosis and influence neighbouring cells possibly by means of bioactive molecules, some of which are packed into extracellular vesicles (EVs). Proprotein convertase subtilisin/kexin type 9 (PCSK9) is expressed and secreted by VSMCs. This study aimed to unravel the role of PCSK9 on VSMCs-derived EVs in terms of content and functionality. EVs were isolated from human VSMCs overexpressing human PCSK9 (VSMCPCSK9-EVs) and tested on endothelial cells, monocytes, macrophages and in a model of zebrafish embryos. Compared to EVs released from wild-type VSMCs, VSMCPCSK9-EVs caused a rise in the expression of adhesion molecules in endothelial cells and of pro-inflammatory cytokines in monocytes. These acquired an increased migratory capacity, a reduced oxidative phosphorylation and secreted proteins involved in immune response and immune effector processes. Concerning macrophages, VSMCPCSK9-EVs enhanced inflammatory milieu and uptake of oxidized low-density lipoproteins, whereas the migratory capacity was reduced. When injected into zebrafish embryos, VSMCPCSK9-EVs favoured the recruitment of macrophages toward the site of injection. The results of the present study provide evidence that PCSK9 plays an inflammatory role by means of EVs, at least by those derived from smooth muscle cells of vascular origin.
Collapse
Affiliation(s)
- Maria Francesca Greco
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Alessandra Stefania Rizzuto
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Marta Zarà
- Centro Cardiologico Monzino, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), 20133 Milan, Italy
| | - Marco Cafora
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20133 Milan, Italy
| | - Chiara Favero
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20133 Milan, Italy
| | - Giulia Solazzo
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20133 Milan, Italy
| | - Ilaria Giusti
- Department of Life, Health and Environmental Sciences, Università degli Studi dell'Aquila, 67100 L'Aquila, Italy
| | - Maria Pia Adorni
- Unit of Neuroscience, Department of Medicine and Surgery, Università degli Studi di Parma, 43124 Parma, Italy
| | - Francesca Zimetti
- Department of Food and Drug, Università degli Studi di Parma, 43124 Parma, Italy
| | - Vincenza Dolo
- Department of Life, Health and Environmental Sciences, Università degli Studi dell'Aquila, 67100 L'Aquila, Italy
| | - Cristina Banfi
- Centro Cardiologico Monzino, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), 20133 Milan, Italy
| | - Nicola Ferri
- Department of Medicine, Università degli Studi di Padova, 35100 Padua, Italy
| | - Cesare R Sirtori
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Silvia Stella Barbieri
- Centro Cardiologico Monzino, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), 20133 Milan, Italy
| | - Anna Pistocchi
- Department of Medical Biotechnology and Translational, Università degli Studi di Milano, L.I.T.A., 20133 Milan, Italy
| | - Valentina Bollati
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20133 Milan, Italy
| | - Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
16
|
Khoza LJ, Kumar P, Dube A, Demana PH, Choonara YE. Insights into Innovative Therapeutics for Drug-Resistant Tuberculosis: Host-Directed Therapy and Autophagy Inducing Modified Nanoparticles. Int J Pharm 2022; 622:121893. [PMID: 35680110 PMCID: PMC9169426 DOI: 10.1016/j.ijpharm.2022.121893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 10/25/2022]
|
17
|
Tsai H, Wang Y, Liao C, Su C, Huang C, Chiu M, Yeh Y. Safety and the probiotic potential of
Bifidobacterium animalis
CP‐9. J Food Sci 2022; 87:2211-2228. [DOI: 10.1111/1750-3841.16129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/30/2022] [Accepted: 02/28/2022] [Indexed: 12/01/2022]
Affiliation(s)
- Hui‐Yun Tsai
- Department of Nutrition and Health Science Fooyin University Kaohsiung Taiwan
- Aging and Disease Prevention Research Center Fooyin University Kaohsiung Taiwan
| | - Ya‐Chin Wang
- Department of Medical Laboratory Sciences and Biotechnology Fooyin University Kaohsiung Taiwan
| | - Chorng‐An Liao
- Aging and Disease Prevention Research Center Fooyin University Kaohsiung Taiwan
- Biomed Analysis Center Fooyin University Hospital Pingtung Taiwan
| | - Chia‐Yan Su
- Aging and Disease Prevention Research Center Fooyin University Kaohsiung Taiwan
- School of Pharmacy Kaohsiung Medical University Kaohsiung Taiwan
| | - Cheng‐Hsieh Huang
- Aging and Disease Prevention Research Center Fooyin University Kaohsiung Taiwan
- Ph. D. Program in Environmental and Occupational Medicine College of Medicine, Kaohsiung Medical University and National Health Research Institutes Kaohsiung Taiwan
| | - Min‐Hsi Chiu
- Aging and Disease Prevention Research Center Fooyin University Kaohsiung Taiwan
- Department of Medical Laboratory Sciences and Biotechnology Fooyin University Kaohsiung Taiwan
- Biomed Analysis Center Fooyin University Hospital Pingtung Taiwan
| | - Yao‐Tsung Yeh
- Aging and Disease Prevention Research Center Fooyin University Kaohsiung Taiwan
- Department of Medical Laboratory Sciences and Biotechnology Fooyin University Kaohsiung Taiwan
- Biomed Analysis Center Fooyin University Hospital Pingtung Taiwan
| |
Collapse
|
18
|
G6PD is a critical enabler of hypoxia-induced accumulation of macrophages and platelets in mice lungs and contributor to lung inflammation. Vascul Pharmacol 2022; 144:106976. [DOI: 10.1016/j.vph.2022.106976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/27/2022] [Accepted: 03/03/2022] [Indexed: 02/05/2023]
|
19
|
Auguste M, Melillo D, Corteggio A, Marino R, Canesi L, Pinsino A, Italiani P, Boraschi D. Methodological Approaches To Assess Innate Immunity and Innate Memory in Marine Invertebrates and Humans. FRONTIERS IN TOXICOLOGY 2022; 4:842469. [PMID: 35295223 PMCID: PMC8915809 DOI: 10.3389/ftox.2022.842469] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/20/2022] [Indexed: 12/17/2022] Open
Abstract
Assessing the impact of drugs and contaminants on immune responses requires methodological approaches able to represent real-life conditions and predict long-term effects. Innate immunity/inflammation is the evolutionarily most widespread and conserved defensive mechanism in living organisms, and therefore we will focus here on immunotoxicological methods that specifically target such processes. By exploiting the conserved mechanisms of innate immunity, we have examined the most representative immunotoxicity methodological approaches across living species, to identify common features and human proxy models/assays. Three marine invertebrate organisms are examined in comparison with humans, i.e., bivalve molluscs, tunicates and sea urchins. In vivo and in vitro approaches are compared, highlighting common mechanisms and species-specific endpoints, to be applied in predictive human and environmental immunotoxicity assessment. Emphasis is given to the 3R principle of Replacement, Refinement and Reduction of Animals in Research and to the application of the ARRIVE guidelines on reporting animal research, in order to strengthen the quality and usability of immunotoxicology research data.
Collapse
Affiliation(s)
- Manon Auguste
- Department of Earth, Environment and Life Sciences, University of Genova, Genova, Italy
| | - Daniela Melillo
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Annunziata Corteggio
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
| | - Rita Marino
- Stazione Zoologica Anton Dohrn, Napoli, Italy
| | - Laura Canesi
- Department of Earth, Environment and Life Sciences, University of Genova, Genova, Italy
| | - Annalisa Pinsino
- Institute of Translational Pharmacology (IFT), CNR, Palermo, Italy
| | - Paola Italiani
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
- Stazione Zoologica Anton Dohrn, Napoli, Italy
- *Correspondence: Paola Italiani, ; Diana Boraschi,
| | - Diana Boraschi
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Napoli, Italy
- Stazione Zoologica Anton Dohrn, Napoli, Italy
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Science (CAS), Shenzhen, China
- *Correspondence: Paola Italiani, ; Diana Boraschi,
| |
Collapse
|
20
|
Zhang B, Xue Y, Zhao J, Jiang H, Zhu J, Yin H, Qiu Y, Hu A, Xu L, Song Y, Wang X. Shionone Attenuates Sepsis-Induced Acute Kidney Injury by Regulating Macrophage Polarization via the ECM1/STAT5 Pathway. Front Med (Lausanne) 2022; 8:796743. [PMID: 35141243 PMCID: PMC8818860 DOI: 10.3389/fmed.2021.796743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/28/2021] [Indexed: 12/16/2022] Open
Abstract
Backgrounds To date, there are no specific drugs approved for the treatment of sepsis associated acute kidney injury (AKI). Shionone is a natural component with anti-inflammatory activity. In this study, we sought to determine the functional role of Shionone in sepsis-induced AKI. Methods Animal models of AKI were constructed by cecum ligation and puncture (CLP) surgery. C57BL/6 mice were randomly assigned to the Sham, CLP, 10 mg/kg DXM, 50 mg/kg Shionone and 100 mg/kg Shionone groups. RAW264.7 treated with lipopolysaccharides (LPS) was used as an in vitro sepsis model and cells were divided into control, LPS, 1 μg/mL Shionone and 2 μg/mL Shionone groups. The pathological status was assessed by Hematoxylin-Eosin (HE) staining assay, protein expressions were detected by immunofluorescence staining and Western blot, macrophage typing was detected by flow, and the levels of pro-inflammatory factors (IL-6, IL-12, IL-1β, TNF-α) and anti-inflammatory factors (IL-10 and TGF-β) were measured using the corresponding kits. Results ECM1 is highly expressed in tissue-infiltrating macrophages under inflammatory conditions. It has been observed that Shionone inhibits the expression of ECM1 and attenuates sepsis-induced injury in kidney and inflammatory factor levels in serum. In addition, Shionone may reduce inflammatory factor levels through the promotion of M2 macrophages by GM-CSF/STAT5/Arg1 pathway to alleviate sepsis induced inflammation in vitro. Conclusion These findings demonstrate that Shionone can alleviate sepsis-induced AKI by promoting M2 macrophage polarization through regulating the ECM1/STAT5 pathway.
Collapse
Affiliation(s)
- Biao Zhang
- Department of Critical Care Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Yi Xue
- Department of Nephrology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Jin Zhao
- Department of Critical Care Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Huojun Jiang
- Department of Critical Care Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Jiaoli Zhu
- Department of Nephrology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Hao Yin
- Li Shicai School Inheritance Studio, Suzhou Hospital of Integrated Traditional and Western Medicine, Suzhou, China
| | - Yizhen Qiu
- Department of Critical Care Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Aihao Hu
- Department of Critical Care Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Lingqi Xu
- Department of Critical Care Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Yi Song
- Department of Critical Care Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
- Yi Song
| | - Xin Wang
- Li Shicai School Inheritance Studio, Suzhou Hospital of Integrated Traditional and Western Medicine, Suzhou, China
- *Correspondence: Xin Wang
| |
Collapse
|
21
|
Kumar R, Bhatia M, Pai K. Role of Chemokines in the Pathogenesis of Visceral Leishmaniasis. Curr Med Chem 2022; 29:5441-5461. [PMID: 35579167 DOI: 10.2174/0929867329666220509171244] [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: 08/04/2021] [Revised: 12/23/2021] [Accepted: 03/02/2022] [Indexed: 11/22/2022]
Abstract
Visceral leishmaniasis (VL; also known as kala-azar), caused by the protozoan parasite Leishmania donovani, is characterized by the inability of the host to generate an effective immune response. The manifestations of the disease depend on the involvement of various immune components such as activation of macrophages, cell mediated immunity, secretion of cytokines and chemokines, etc. Macrophages are the final host cells for Leishmania parasites to multiply, and they are the key to a controlled or aggravated response that leads to clinical symptoms. The two most common macrophage phenotypes are M1 and M2. The pro-inflammatory microenvironment (mainly by IL-1β, IL-6, IL-12, IL-23, and TNF-α cytokines) and tissue injury driven by classically activated macrophages (M1-like) and wound healing driven by alternatively activated macrophages (M2-like) in an anti-inflammatory environment (mainly by IL-10, TGF-β, chemokine ligand (CCL)1, CCL2, CCL17, CCL18, and CCL22). Moreover, on polarized Th cells, chemokine receptors are expressed differently. Typically, CXCR3 and CCR5 are preferentially expressed on polarized Th1 cells, whereas CCR3, CCR4, and CCR8 have been associated with the Th2 phenotype. Further, the ability of the host to produce a cell-mediated immune response capable of regulating and/or eliminating the parasite is critical in the fight against the disease. Here, we review the interactions between parasites and chemokines and chemokine receptors in the pathogenesis of VL.
Collapse
Affiliation(s)
- Ramesh Kumar
- Department of Biochemistry, Bundelkhand University, Jhansi (UP), India
| | - Madhav Bhatia
- Department of Pathology and Biomedical Sciences, University of Otago, Christchurch, New Zealand
| | - Kalpana Pai
- Department of Zoology, Savitribai Phule Pune University, Pune, Maharashtra, India
| |
Collapse
|
22
|
Shanmugam G, Sundaramoorthy A, Shanmugam N. Biosynthesis of Silver Nanoparticles from Leaf Extract of Salvia coccinea and Its Effects of Anti-inflammatory Potential in Human Monocytic THP-1 Cells. ACS APPLIED BIO MATERIALS 2021; 4:8433-8442. [DOI: 10.1021/acsabm.1c00963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Gobinath Shanmugam
- Department of Biomedical Science, Bharathidasan University, Tiruchirappalli 620 024, Tamil Nadu, India
| | - Arun Sundaramoorthy
- Department of Biomedical Science, Bharathidasan University, Tiruchirappalli 620 024, Tamil Nadu, India
| | - Narkunaraja Shanmugam
- Department of Biomedical Science, Bharathidasan University, Tiruchirappalli 620 024, Tamil Nadu, India
| |
Collapse
|
23
|
Chisari E, Rehak L, Khan WS, Maffulli N. Tendon healing is adversely affected by low-grade inflammation. J Orthop Surg Res 2021; 16:700. [PMID: 34863223 PMCID: PMC8642928 DOI: 10.1186/s13018-021-02811-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 10/06/2020] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Tendinopathy is common, presents with pain and activity limitation, and is associated with a high risk of recurrence of the injury. Tendinopathy usually occurs as a results of a disrupted healing response to a primary injury where cellular and molecular pathways lead to low grade chronic inflammation. MAIN FINDINGS There has been a renewed interest in investigating the role of Inflammation in the pathogenesis of tendinopathy, in particular during the initial phases of the condition where it may not be clinically evident. Understanding the early and late stages of tendon injury pathogenesis would help develop new and effective treatments addressed at targeting the inflammatory pathways. CONCLUSION This review outlines the role of low-grade Inflammation in the pathogenesis of tendinopathy, stressing the role of proinflammatory cytokines, proteolytic enzymes and growth factors, and explores how Inflammation exerts a negative influence on the process of tendon healing.
Collapse
Affiliation(s)
| | - Laura Rehak
- Athena Biomedical Innovations, Florence, Italy
| | - Wasim S Khan
- Division of Trauma and Orthopaedic Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Nicola Maffulli
- Department of Musculoskeletal Disorders, School of Medicine and Surgery, University of Salerno, Salerno, Italy.
- Clinica Ortopedica, Ospedale San Giovanni di Dio e Ruggi D'Aragona, 84131, Salerno, Italy.
- Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Mile End Hospital, 275 Bancroft Road, London, E1 4DG, UK.
- School of Medicine, Institute of Science and Technology in Medicine, Guy Hilton Research Centre, Keele University, Thornburrow Drive, Hartshill, Stoke-on-Trent, ST4 7QB, UK.
| |
Collapse
|
24
|
Boraschi D, Li D, Li Y, Italiani P. In Vitro and In Vivo Models to Assess the Immune-Related Effects of Nanomaterials. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182211769. [PMID: 34831525 PMCID: PMC8623312 DOI: 10.3390/ijerph182211769] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/13/2021] [Accepted: 10/13/2021] [Indexed: 12/12/2022]
Abstract
The immunological safety of drugs, nanomaterials and contaminants is a central point in the regulatory evaluation and safety monitoring of working and public places and of the environment. In fact, anomalies in immune responses may cause diseases and hamper the physical and functional integrity of living organisms, from plants to human beings. In the case of nanomaterials, many experimental models are used for assessing their immunosafety, some of which have been adopted by regulatory bodies. All of them, however, suffer from shortcomings and approximations, and may be inaccurate in representing real-life responses, thereby leading to incomplete, incorrect or even misleading predictions. Here, we review the advantages and disadvantages of current nanoimmunosafety models, comparing in vivo vs. in vitro models and examining the use of animal vs. human cells, primary vs. transformed cells, complex multicellular and 3D models, organoids and organs-on-chip, in view of implementing a reliable and personalized nanoimmunosafety testing. The general conclusion is that the choice of testing models is key for obtaining reliable predictive information, and therefore special attention should be devoted to selecting the most relevant and realistic suite of models in order to generate relevant information that can allow for safer-by-design nanotechnological developments.
Collapse
Affiliation(s)
- Diana Boraschi
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518055, China; (D.L.); (Y.L.)
- Institute of Biochemistry and Cell Biology (IBBC), Consiglio Nazionale delle Ricerche (CNR), 80131 Napoli, Italy;
- Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
- Correspondence:
| | - Dongjie Li
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518055, China; (D.L.); (Y.L.)
| | - Yang Li
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518055, China; (D.L.); (Y.L.)
| | - Paola Italiani
- Institute of Biochemistry and Cell Biology (IBBC), Consiglio Nazionale delle Ricerche (CNR), 80131 Napoli, Italy;
| |
Collapse
|
25
|
Goswami KK, Bose A, Baral R. Macrophages in tumor: An inflammatory perspective. Clin Immunol 2021; 232:108875. [PMID: 34740843 DOI: 10.1016/j.clim.2021.108875] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 10/07/2021] [Accepted: 10/27/2021] [Indexed: 01/08/2023]
Abstract
Inflammation is a part of carefully co-ordinated healing immune exercise to eliminate injurious stimuli. However, in substantial number of cancer types, it contributes in shaping up of robust tumor microenvironment (TME). Solid TME promotes infiltration of tumor associated macrophages (TAMs) that contributes to cancer promotion. TAMs are functionally heterogeneous and display an extraordinary degree of plasticity, which allow 'Switching' of macrophages into an 'M2', phenotype, linked with immunosuppression, advancement of tumor angiogenesis with metastatic consequences. In contrary to the classical M1 macrophages, these M2 TAMs are high-IL-10, TGF-β secreting-'anti-inflammatory'. In this review, we will discuss the modes of infiltration and switching of TAMs into M2 anti-inflammatory state in the TME to promote immunosuppression and inflammation-driven cancer.
Collapse
Affiliation(s)
- Kuntal Kanti Goswami
- Department of Microbiology, Asutosh College, 92, S. P. Mukherjee Road, Kolkata 700026, India.
| | - Anamika Bose
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Rathindranath Baral
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| |
Collapse
|
26
|
Foo SS, Cambou MC, Mok T, Fajardo VM, Jung KL, Fuller T, Chen W, Kerin T, Mei J, Bhattacharya D, Choi Y, Wu X, Xia T, Shin WJ, Cranston J, Aldrovandi G, Tobin N, Contreras D, Ibarrondo FJ, Yang O, Yang S, Garner O, Cortado R, Bryson Y, Janzen C, Ghosh S, Devaskar S, Asilnejad B, Moreira ME, Vasconcelos Z, Soni PR, Gibson LC, Brasil P, Comhair SA, Arumugaswami V, Erzurum SC, Rao R, Jung JU, Nielsen-Saines K. The systemic inflammatory landscape of COVID-19 in pregnancy: Extensive serum proteomic profiling of mother-infant dyads with in utero SARS-CoV-2. Cell Rep Med 2021; 2:100453. [PMID: 34723226 PMCID: PMC8549189 DOI: 10.1016/j.xcrm.2021.100453] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 08/30/2021] [Accepted: 10/21/2021] [Indexed: 12/22/2022]
Abstract
While pregnancy increases the risk for severe COVID-19, the clinical and immunological implications of COVID-19 on maternal-fetal health remain unknown. Here, we present the clinical and immunological landscapes of 93 COVID-19 mothers and 45 of their SARS-CoV-2-exposed infants through comprehensive serum proteomics profiling for >1,400 cytokines of their peripheral and cord blood specimens. Prenatal SARS-CoV-2 infection triggers NF-κB-dependent proinflammatory immune activation. Pregnant women with severe COVID-19 show increased inflammation and unique IFN-λ antiviral signaling, with elevated levels of IFNL1 and IFNLR1. Furthermore, SARS-CoV-2 infection re-shapes maternal immunity at delivery, altering the expression of pregnancy complication-associated cytokines, inducing MMP7, MDK, and ESM1 and reducing BGN and CD209. Finally, COVID-19-exposed infants exhibit induction of T cell-associated cytokines (IL33, NFATC3, and CCL21), while some undergo IL-1β/IL-18/CASP1 axis-driven neonatal respiratory distress despite birth at term. Our findings demonstrate COVID-19-induced immune rewiring in both mothers and neonates, warranting long-term clinical follow-up to mitigate potential health risks.
Collapse
Affiliation(s)
- Suan-Sin Foo
- Department of Cancer Biology, Infection Biology Program, and Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Mary Catherine Cambou
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Thalia Mok
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Viviana M. Fajardo
- Department of Pediatrics, Division of Neonatology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kyle L. Jung
- Department of Cancer Biology, Infection Biology Program, and Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Trevon Fuller
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Manguinhos, Rio de Janeiro 21040-360, Brazil
| | - Weiqiang Chen
- Department of Cancer Biology, Infection Biology Program, and Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Tara Kerin
- Department of Pediatrics, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jenny Mei
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Debika Bhattacharya
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Younho Choi
- Department of Cancer Biology, Infection Biology Program, and Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Xin Wu
- Department of Cancer Biology, Infection Biology Program, and Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Tian Xia
- Department of Cancer Biology, Infection Biology Program, and Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Woo-Jin Shin
- Department of Cancer Biology, Infection Biology Program, and Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jessica Cranston
- Department of Pediatrics, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Grace Aldrovandi
- Department of Pediatrics, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nicole Tobin
- Department of Pediatrics, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Deisy Contreras
- Department of Pediatrics, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Francisco J. Ibarrondo
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Otto Yang
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shangxin Yang
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Omai Garner
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ruth Cortado
- Department of Pediatrics, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yvonne Bryson
- Department of Pediatrics, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Carla Janzen
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shubhamoy Ghosh
- Department of Pediatrics, Division of Neonatology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sherin Devaskar
- Department of Pediatrics, Division of Neonatology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Brenda Asilnejad
- Georgetown University School of Medicine, Washington, DC 20007, USA
| | | | - Zilton Vasconcelos
- Instituto Fernades Figueira, Fiocruz, Flamengo, Rio de Janeiro 20140-360, Brazil
| | - Priya R. Soni
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90049, USA
| | - L. Caroline Gibson
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90049, USA
| | - Patricia Brasil
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Manguinhos, Rio de Janeiro 21040-360, Brazil
| | - Suzy A.A. Comhair
- Respiratory Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Serpil C. Erzurum
- Respiratory Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Rashmi Rao
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jae U. Jung
- Department of Cancer Biology, Infection Biology Program, and Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA,Corresponding author
| | - Karin Nielsen-Saines
- Department of Pediatrics, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA,Corresponding author
| |
Collapse
|
27
|
Nikovics K, Favier AL. Macrophage Identification In Situ. Biomedicines 2021; 9:1393. [PMID: 34680510 PMCID: PMC8533306 DOI: 10.3390/biomedicines9101393] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022] Open
Abstract
Understanding the processes of inflammation and tissue regeneration after injury is of great importance. For a long time, macrophages have been known to play a central role during different stages of inflammation and tissue regeneration. However, the molecular and cellular mechanisms by which they exert their effects are as yet mostly unknown. While in vitro macrophages have been characterized, recent progress in macrophage biology studies revealed that macrophages in vivo exhibited distinctive features. Actually, the precise characterization of the macrophages in vivo is essential to develop new healing treatments and can be approached via in situ analyses. Nowadays, the characterization of macrophages in situ has improved significantly using antigen surface markers and cytokine secretion identification resulting in specific patterns. This review aims for a comprehensive overview of different tools used for in situ macrophage identification, reporter genes, immunolabeling and in situ hybridization, discussing their advantages and limitations.
Collapse
Affiliation(s)
- Krisztina Nikovics
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223 Brétigny-sur-Orge, France;
| | | |
Collapse
|
28
|
Boyd A, Byrne S, Middleton RJ, Banati RB, Liu GJ. Control of Neuroinflammation through Radiation-Induced Microglial Changes. Cells 2021; 10:2381. [PMID: 34572030 PMCID: PMC8468704 DOI: 10.3390/cells10092381] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 12/15/2022] Open
Abstract
Microglia, the innate immune cells of the central nervous system, play a pivotal role in the modulation of neuroinflammation. Neuroinflammation has been implicated in many diseases of the CNS, including Alzheimer's disease and Parkinson's disease. It is well documented that microglial activation, initiated by a variety of stressors, can trigger a potentially destructive neuroinflammatory response via the release of pro-inflammatory molecules, and reactive oxygen and nitrogen species. However, the potential anti-inflammatory and neuroprotective effects that microglia are also thought to exhibit have been under-investigated. The application of ionising radiation at different doses and dose schedules may reveal novel methods for the control of microglial response to stressors, potentially highlighting avenues for treatment of neuroinflammation associated CNS disorders, such as Alzheimer's disease and Parkinson's disease. There remains a need to characterise the response of microglia to radiation, particularly low dose ionising radiation.
Collapse
Affiliation(s)
- Alexandra Boyd
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia; (A.B.); (S.B.); (R.J.M.); (R.B.B.)
| | - Sarah Byrne
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia; (A.B.); (S.B.); (R.J.M.); (R.B.B.)
| | - Ryan J. Middleton
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia; (A.B.); (S.B.); (R.J.M.); (R.B.B.)
| | - Richard B. Banati
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia; (A.B.); (S.B.); (R.J.M.); (R.B.B.)
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, Sydney, NSW 2050, Australia
| | - Guo-Jun Liu
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia; (A.B.); (S.B.); (R.J.M.); (R.B.B.)
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, Sydney, NSW 2050, Australia
| |
Collapse
|
29
|
Mehta AK, Kadel S, Townsend MG, Oliwa M, Guerriero JL. Macrophage Biology and Mechanisms of Immune Suppression in Breast Cancer. Front Immunol 2021; 12:643771. [PMID: 33968034 PMCID: PMC8102870 DOI: 10.3389/fimmu.2021.643771] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/17/2021] [Indexed: 12/14/2022] Open
Abstract
Macrophages are crucial innate immune cells that maintain tissue homeostasis and defend against pathogens; however, their infiltration into tumors has been associated with adverse outcomes. Tumor-associated macrophages (TAMs) represent a significant component of the inflammatory infiltrate in breast tumors, and extensive infiltration of TAMs has been linked to poor prognosis in breast cancer. Here, we detail how TAMs impede a productive tumor immunity cycle by limiting antigen presentation and reducing activation of cytotoxic T lymphocytes (CTLs) while simultaneously supporting tumor cell survival, angiogenesis, and metastasis. There is an urgent need to overcome TAM-mediated immune suppression for durable anti-tumor immunity in breast cancer. To date, failure to fully characterize TAM biology and classify multiple subsets has hindered advancement in therapeutic targeting. In this regard, the complexity of TAMs has recently taken center stage owing to their subset diversity and tightly regulated molecular and metabolic phenotypes. In this review, we reveal major gaps in our knowledge of the functional and phenotypic characterization of TAM subsets associated with breast cancer, before and after treatment. Future work to characterize TAM subsets, location, and crosstalk with neighboring cells will be critical to counteract TAM pro-tumor functions and to identify novel TAM-modulating strategies and combinations that are likely to enhance current therapies and overcome chemo- and immuno-therapy resistance.
Collapse
Affiliation(s)
- Anita K Mehta
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, United States.,Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, United States
| | - Sapana Kadel
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Madeline G Townsend
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, United States.,Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, United States
| | - Madisson Oliwa
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, United States.,Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, United States
| | - Jennifer L Guerriero
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, United States.,Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, United States
| |
Collapse
|
30
|
Barbosa MMF, Kanno AI, Farias LP, Madej M, Sipos G, Sbrana S, Romani L, Boraschi D, Leite LCC, Italiani P. Primary and Memory Response of Human Monocytes to Vaccines: Role of Nanoparticulate Antigens in Inducing Innate Memory. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:931. [PMID: 33917456 PMCID: PMC8067467 DOI: 10.3390/nano11040931] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/25/2021] [Accepted: 04/01/2021] [Indexed: 01/09/2023]
Abstract
Innate immune cells such as monocytes and macrophages are activated in response to microbial and other challenges and mount an inflammatory defensive response. Exposed cells develop the so-called innate memory, which allows them to react differently to a subsequent challenge, aiming at better protection. In this study, using human primary monocytes in vitro, we have assessed the memory-inducing capacity of two antigenic molecules of Schistosoma mansoni in soluble form compared to the same molecules coupled to outer membrane vesicles of Neisseria lactamica. The results show that particulate challenges are much more efficient than soluble molecules in inducing innate memory, which is measured as the production of inflammatory and anti-inflammatory cytokines (TNFα, IL-6, IL-10). Controls run with LPS from Klebsiella pneumoniae compared to the whole bacteria show that while LPS alone has strong memory-inducing capacity, the entire bacteria are more efficient. These data suggest that microbial antigens that are unable to induce innate immune activation can nevertheless participate in innate activation and memory when in a particulate form, which is a notion that supports the use of nanoparticulate antigens in vaccination strategies for achieving adjuvant-like effects of innate activation as well as priming for improved reactivity to future challenges.
Collapse
Affiliation(s)
- Mayra M. Ferrari Barbosa
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP 05503-900, Brazil; (M.M.F.B.); (A.I.K.)
| | - Alex Issamu Kanno
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP 05503-900, Brazil; (M.M.F.B.); (A.I.K.)
| | - Leonardo Paiva Farias
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA 40296-710, Brazil;
| | - Mariusz Madej
- Istituto di Biochimica e Biologia Cellulare, Consiglio Nazionale delle Ricerche, 80131 Napoli, Italy; (M.M.); (G.S.)
| | - Gergö Sipos
- Istituto di Biochimica e Biologia Cellulare, Consiglio Nazionale delle Ricerche, 80131 Napoli, Italy; (M.M.); (G.S.)
| | - Silverio Sbrana
- Istituto di Fisiologia Clinica, Consiglio Nazionale delle Ricerche, 54100 Massa, Italy;
| | - Luigina Romani
- Dipartimento di Medicina e Chirurgia, University of Perugia, 06132 Perugia, Italy;
| | - Diana Boraschi
- Istituto di Biochimica e Biologia Cellulare, Consiglio Nazionale delle Ricerche, 80131 Napoli, Italy; (M.M.); (G.S.)
- Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
| | - Luciana C. C. Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP 05503-900, Brazil; (M.M.F.B.); (A.I.K.)
| | - Paola Italiani
- Istituto di Biochimica e Biologia Cellulare, Consiglio Nazionale delle Ricerche, 80131 Napoli, Italy; (M.M.); (G.S.)
- Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
| |
Collapse
|
31
|
The Role of Creatine in the Development and Activation of Immune Responses. Nutrients 2021; 13:nu13030751. [PMID: 33652752 PMCID: PMC7996722 DOI: 10.3390/nu13030751] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/10/2021] [Accepted: 02/13/2021] [Indexed: 02/06/2023] Open
Abstract
The use of dietary supplements has become increasingly common over the past 20 years. Whereas supplements were formerly used mainly by elite athletes, age and fitness status no longer dictates who uses these substances. Indeed, many nutritional supplements are recommended by health care professionals to their patients. Creatine (CR) is a widely used dietary supplement that has been well-studied for its effects on performance and health. CR also aids in recovery from strenuous bouts of exercise by reducing inflammation. Although CR is considered to be very safe in recommended doses, a caveat is that a preponderance of the studies have focused upon young athletic individuals; thus there is limited knowledge regarding the effects of CR on children or the elderly. In this review, we examine the potential of CR to impact the host outside of the musculoskeletal system, specifically, the immune system, and discuss the available data demonstrating that CR can impact both innate and adaptive immune responses, together with how the effects on the immune system might be exploited to enhance human health.
Collapse
|
32
|
Swartzwelter BJ, Verde A, Rehak L, Madej M, Puntes VF, De Luca AC, Boraschi D, Italiani P. Interaction between Macrophages and Nanoparticles: In Vitro 3D Cultures for the Realistic Assessment of Inflammatory Activation and Modulation of Innate Memory. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:207. [PMID: 33467414 PMCID: PMC7830034 DOI: 10.3390/nano11010207] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/11/2022]
Abstract
Understanding the modes of interaction between human monocytes/macrophages and engineered nanoparticles is the basis for assessing particle safety, in terms of activation of innate/inflammatory reactions, and their possible exploitation for medical applications. In vitro assessment of nanoparticle-macrophage interaction allows for examining the response of primary human cells, but the conventional 2D cultures do not reproduce the three-dimensional spacing of a tissue and the interaction of macrophages with the extracellular tissue matrix, conditions that shape macrophage recognition capacity and reactivity. Here, we have compared traditional 2D cultures with cultures on a 3D collagen matrix for evaluating the capacity gold nanoparticles to induce monocyte activation and subsequent innate memory in human blood monocytes in comparison to bacterial LPS. Results show that monocytes react to stimuli almost in the same way in 2D and 3D cultures in terms of production of TNFα and IL-6, but that notable differences are found when IL-8 and IL-1Ra are examined, in particular in the recall/memory response of primed cells to a second stimulation, with the 3D cultures showing cell activation and memory effects of nanoparticles better. In addition, the response variations in monocytes/macrophages from different donors point towards a personalized assessment of the nanoparticle effects on macrophage activation.
Collapse
Affiliation(s)
- Benjamin J. Swartzwelter
- Institute of Biochemistry and Cell Biology, National Research Council, 80131 Napoli, Italy; (B.J.S.); (A.V.); (M.M.); (A.C.D.L.)
| | - Alessandro Verde
- Institute of Biochemistry and Cell Biology, National Research Council, 80131 Napoli, Italy; (B.J.S.); (A.V.); (M.M.); (A.C.D.L.)
| | - Laura Rehak
- Athena Biomedical Innovations, 00100 Roma, Italy;
| | - Mariusz Madej
- Institute of Biochemistry and Cell Biology, National Research Council, 80131 Napoli, Italy; (B.J.S.); (A.V.); (M.M.); (A.C.D.L.)
| | - Victor. F. Puntes
- Institut Català de Nanociència i Nanotecnologia (ICN2), CSIC and The Barcelona Institute of Science and Technology (BIST), Campus UAB, 08193 Bellaterra, Barcelona, Spain;
| | - Anna Chiara De Luca
- Institute of Biochemistry and Cell Biology, National Research Council, 80131 Napoli, Italy; (B.J.S.); (A.V.); (M.M.); (A.C.D.L.)
| | - Diana Boraschi
- Institute of Biochemistry and Cell Biology, National Research Council, 80131 Napoli, Italy; (B.J.S.); (A.V.); (M.M.); (A.C.D.L.)
- Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
| | - Paola Italiani
- Institute of Biochemistry and Cell Biology, National Research Council, 80131 Napoli, Italy; (B.J.S.); (A.V.); (M.M.); (A.C.D.L.)
- Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
| |
Collapse
|
33
|
Melbourne JK, Rosen C, Chase KA, Feiner B, Sharma RP. Monocyte Transcriptional Profiling Highlights a Shift in Immune Signatures Over the Course of Illness in Schizophrenia. Front Psychiatry 2021; 12:649494. [PMID: 34054608 PMCID: PMC8160367 DOI: 10.3389/fpsyt.2021.649494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/19/2021] [Indexed: 02/05/2023] Open
Abstract
With advanced understanding of the intricate interplay between the immune and central nervous systems in neurological and neuropsychiatric illness, there is renewed interest in the potential contribution of immune dysregulation to the development and progression of schizophrenia. To inform this line of inquiry requires a more nuanced understanding of specific immune changes throughout the course of illness. Here, we utilized a genome-wide sequencing approach to transcriptionally profile circulating monocytes in participants with chronic schizophrenia. These myeloid cells, isolated from whole blood samples, are highly plastic with potentially important disease-modifying functions. Differential gene expression and gene set enrichment analyses, focusing on established monocyte phenotypic signatures, including those related to proinflammatory ("M1-like") and protective or tissue remodeling ("M2-like") functions, were carried out. We demonstrate an overall enrichment of both "M1-like" (interferon-alpha, interferon-gamma, lipopolysaccharide acute) and "M2-like" (endotoxin tolerance, glucocorticoid acute) monocyte signatures in the participants with schizophrenia compared to non-psychiatric controls. There was no enrichment of the "M1-like" chronic stress signature or the "M2-like" interleukin-4 signature. Using the Molecular Signatures Database Hallmark gene sets list, the "interferon response" was most strongly enriched in schizophrenia compared to controls. Additionally, an exploratory subgroup analysis based on illness duration suggests a shift in monocyte phenotype with illness progression. Specifically, the "M1-like" interferon-gamma signature shows decreased enrichment accompanied by increased enrichment of opposing "M2-like" signatures in participants with a medium illness duration shifting to a strong enrichment of interferon response signatures only in participants with a long illness duration. These findings related to circulating immune cell phenotype have potentially important implications for understanding the role of immune dysregulation in schizophrenia and are a critical consideration for future study design and immune-targeting treatment strategies.
Collapse
Affiliation(s)
- Jennifer K Melbourne
- Department of Psychiatry, The Psychiatric Institute, University of Illinois at Chicago, Chicago, IL, United States
| | - Cherise Rosen
- Department of Psychiatry, The Psychiatric Institute, University of Illinois at Chicago, Chicago, IL, United States
| | - Kayla A Chase
- Department of Psychiatry, The Psychiatric Institute, University of Illinois at Chicago, Chicago, IL, United States
| | - Benjamin Feiner
- Department of Psychiatry, The Psychiatric Institute, University of Illinois at Chicago, Chicago, IL, United States
| | - Rajiv P Sharma
- Department of Psychiatry, The Psychiatric Institute, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
34
|
Tamaki S, Kuroshima S, Hayano H, Nakajima K, Kakehashi H, Ishisaki A, Sawase T. Dynamic polarization shifting from M1 to M2 macrophages in reduced osteonecrosis of the jaw-like lesions by cessation of anti-RANKL antibody in mice. Bone 2020; 141:115560. [PMID: 32730924 DOI: 10.1016/j.bone.2020.115560] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 07/08/2020] [Accepted: 07/10/2020] [Indexed: 11/28/2022]
Abstract
Denosumab-related osteonecrosis of the jaw (DRONJ), which mainly occurs in cancer patients receiving anti-receptor activator NF-kappaB ligand (RANKL) antibody, reduces oral health-related quality of life. However, the exact mechanisms of and definitive treatment strategies for DRONJ remain unknown. We hypothesized that cessation of denosumab heals and/or ameliorates DRONJ, since it is a protein-based antibody agent, although stopping denosumab should be avoided in clinical situations. Therefore, the aims of this study were: 1) to create a healing and/or amelioration murine model of DRONJ-like lesions induced by chemotherapy/anti-RANKL antibody (mAb) combination therapy and tooth extraction; and 2) to investigate histopathology and immunopathology in the extraction sockets by comparing the murine model of DRONJ-like lesions with the amelioration/healing model of DRONJ-like lesions. Eight-week-old, female C57B/6J mice received chemotherapeutic drug (cyclophosphamide: CY) and mAb combination therapy (CY/mAb) with tooth extraction. Open wounds were sustained in CY/mAb-treated mice at 2 and 4 weeks post-extraction. Impaired socket healing was diagnosed as CY/mAb-related ONJ-like lesions at 3 weeks post-extraction in this study. Next, mAb was discontinued for 2 and 4 weeks after diagnosis of CY/mAb-related ONJ-like lesions. mAb cessation for 2 weeks induced partial osseous wound healing and significantly improved soft tissue wound healing of the extraction sockets. Anti-angiogenesis and normal lymphangiogenesis with CY/mAb combination therapy was not changed by mAb discontinuation. However, mAb cessation for 2 weeks significantly increased the number of CD38+F4/80+ M1 and CD163+F4/80+ M2 macrophages, which significantly increased the M2/M1 ratio in the connective tissue of extraction sockets. No direct effects of mAb on macrophages were noted both in vivo and in vitro. Therefore, the developed healing and/or amelioration murine model of CY/mAb-related ONJ-like lesions is a useful tool to investigate the histopathology and immunopathology of DRONJ in humans. Dynamic polarization shifting from M1 to M2 macrophages induced by mAb cessation may play an important role in wound healing, rather than angiogenesis and lymphangiogenesis, in DRONJ.
Collapse
Affiliation(s)
- Saki Tamaki
- Department of Applied Prosthodontics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| | - Shinichiro Kuroshima
- Department of Applied Prosthodontics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan.
| | - Hiroki Hayano
- Department of Applied Prosthodontics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| | - Kazunori Nakajima
- Department of Applied Prosthodontics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| | - Hiroe Kakehashi
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnosis and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Akira Ishisaki
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Shiwa-gun, Iwate 028-3694, Japan
| | - Takashi Sawase
- Department of Applied Prosthodontics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| |
Collapse
|
35
|
Kotliar D, Lin AE, Logue J, Hughes TK, Khoury NM, Raju SS, Wadsworth MH, Chen H, Kurtz JR, Dighero-Kemp B, Bjornson ZB, Mukherjee N, Sellers BA, Tran N, Bauer MR, Adams GC, Adams R, Rinn JL, Melé M, Schaffner SF, Nolan GP, Barnes KG, Hensley LE, McIlwain DR, Shalek AK, Sabeti PC, Bennett RS. Single-Cell Profiling of Ebola Virus Disease In Vivo Reveals Viral and Host Dynamics. Cell 2020; 183:1383-1401.e19. [PMID: 33159858 PMCID: PMC7707107 DOI: 10.1016/j.cell.2020.10.002] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/10/2020] [Accepted: 10/02/2020] [Indexed: 12/14/2022]
Abstract
Ebola virus (EBOV) causes epidemics with high mortality yet remains understudied due to the challenge of experimentation in high-containment and outbreak settings. Here, we used single-cell transcriptomics and CyTOF-based single-cell protein quantification to characterize peripheral immune cells during EBOV infection in rhesus monkeys. We obtained 100,000 transcriptomes and 15,000,000 protein profiles, finding that immature, proliferative monocyte-lineage cells with reduced antigen-presentation capacity replace conventional monocyte subsets, while lymphocytes upregulate apoptosis genes and decline in abundance. By quantifying intracellular viral RNA, we identify molecular determinants of tropism among circulating immune cells and examine temporal dynamics in viral and host gene expression. Within infected cells, EBOV downregulates STAT1 mRNA and interferon signaling, and it upregulates putative pro-viral genes (e.g., DYNLL1 and HSPA5), nominating pathways the virus manipulates for its replication. This study sheds light on EBOV tropism, replication dynamics, and elicited immune response and provides a framework for characterizing host-virus interactions under maximum containment.
Collapse
Affiliation(s)
- Dylan Kotliar
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| | - Aaron E Lin
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Program in Virology, Harvard Medical School, Boston, MA 02115, USA.
| | - James Logue
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Travis K Hughes
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Chemistry, Institute for Medical Engineering and Sciences (IMES), and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02142, USA; Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Nadine M Khoury
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Siddharth S Raju
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Marc H Wadsworth
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry, Institute for Medical Engineering and Sciences (IMES), and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02142, USA; Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Han Chen
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Jonathan R Kurtz
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Bonnie Dighero-Kemp
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Zach B Bjornson
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | | | - Brian A Sellers
- Trans-NIH Center for Human Immunology, Autoimmunity, and Inflammation, National Institutes of Health, Bethesda, MD 20814, USA
| | - Nancy Tran
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Matthew R Bauer
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Gordon C Adams
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ricky Adams
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - John L Rinn
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Marta Melé
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Catalonia 08034, Spain
| | - Stephen F Schaffner
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
| | - Garry P Nolan
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Kayla G Barnes
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA; MRC-University of Glasgow Centre for Virus Research, Glasgow G61 1QH, UK
| | - Lisa E Hensley
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA.
| | - David R McIlwain
- Department of Pathology, Stanford University, Stanford, CA 94305, USA.
| | - Alex K Shalek
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Chemistry, Institute for Medical Engineering and Sciences (IMES), and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02142, USA; Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Pardis C Sabeti
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Richard S Bennett
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| |
Collapse
|
36
|
Liu L, Guo S, Shi W, Liu Q, Huo F, Wu Y, Tian W. Bone Marrow Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Promote Periodontal Regeneration. Tissue Eng Part A 2020; 27:962-976. [PMID: 32962564 DOI: 10.1089/ten.tea.2020.0141] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Bone marrow mesenchymal stem cell-derived small extracellular vesicles (BMSC-sEVs) can be used as a potential cell-free strategy for periodontal tissue regeneration, and we aim to investigate the effect and possible mechanism of BMSC-sEV in periodontal tissue regeneration in this study. The BMSC-sEV was isolated by the Exosome Isolation™ reagent and identified by transmission electron microscopy, nanoparticle tracking analysis, and Western blotting. The human periodontal ligament cells (hPDLCs) were cocultured with BMSC-sEV in vitro to detect the effects of BMSC-sEV on hPDLC migration, proliferation, and differentiation. The BMSC-sEV loaded by hydrogel was injected into experimental periodontitis rats to verify the therapeutic effect and possible mechanism. The results showed that BMSC-sEVs were 30-150 nm vesicles and expressed the exosome protein CD63 and tumor susceptibility 101 (TSG101), which could promote the migration, proliferation, osteogenic differentiation of hPDLCs. The BMSC-sEV-hydrogel had a therapeutic effect on periodontitis rats. After administration for 4-8 weeks, microcomputed tomography and histological analysis showed that alveolar bone loss, inflammatory infiltration, and collagen destruction in the BMSC-sEV-hydrogel group were less than that in the phosphate-buffered saline (PBS)-hydrogel group and periodontitis group. Further immunohistochemical and immunofluorescent staining revealed that the number of tartrate-resistant acid phosphatase-positive cells and the expression ratio of osteoprotegerin (OPG) and receptor-activator of nuclear factor kappa beta ligand (RANKL) in the BMSC-sEV-hydrogel group were lower than that in the PBS-hydrogel group and periodontitis group, while the expression of transforming growth factor-beta 1 (TGF-β1) and the ratio of macrophage type 2 and macrophage type 1 (M2/M1) were opposite. Therefore, BMSC-sEV can promote the regeneration of periodontal tissues, and that may be partly due to BMSC-sEV involvement in the OPG-RANKL-RANK signaling pathway to regulate the function of osteoclasts and affect the macrophage polarization and TGF-β1 expression to modulate the inflammatory immune response, thereby inhibiting the development of periodontitis and immune damage of periodontal tissue. Impact statement Bone marrow mesenchymal stem cell-derived small extracellular vesicles (BMSCs-sEVs) have been proven to have similar functions to BMSCs, such as promoting the regeneration of heart, liver, kidney, and bone tissue and regulating immune responses. BMSCs are candidate seed cells of periodontal regeneration, but it is unclear about the role of BMSC-sEV on periodontal regeneration. In this study, we explored the effects and possible mechanism of BMSC-sEV on periodontal regeneration. The results of this study provide the evidence of BMSC-sEV as a cell-free strategy for periodontal regeneration.
Collapse
Affiliation(s)
- Li Liu
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Shujuan Guo
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Weiwei Shi
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Qian Liu
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Fangjun Huo
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Yafei Wu
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Weidong Tian
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
37
|
Dwyer CM, Nguyen LTT, Healy LM, Dutta R, Ludwin S, Antel J, Binder MD, Kilpatrick TJ. Multiple Sclerosis as a Syndrome-Implications for Future Management. Front Neurol 2020; 11:784. [PMID: 32982904 PMCID: PMC7483755 DOI: 10.3389/fneur.2020.00784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/25/2020] [Indexed: 12/25/2022] Open
Abstract
We propose that multiple sclerosis (MS) is best characterized as a syndrome rather than a single disease because different pathogenetic mechanisms can result in the constellation of symptoms and signs by which MS is clinically characterized. We describe several cellular mechanisms that could generate inflammatory demyelination through disruption of homeostatic interactions between immune and neural cells. We illustrate that genomics is important in identifying phenocopies, in particular for primary progressive MS. We posit that molecular profiling, rather than traditional clinical phenotyping, will facilitate meaningful patient stratification, as illustrated by interactions between HLA and a regulator of homeostatic phagocytosis, MERTK. We envisage a personalized approach to MS management where genetic, molecular, and cellular information guides management.
Collapse
Affiliation(s)
- Christopher M Dwyer
- Florey Institute of Neuroscience and Mental Health, Florey Department, The University of Melbourne, Parkville, VIC, Australia.,Department of Neurology, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Linda Thien-Trang Nguyen
- Florey Institute of Neuroscience and Mental Health, Florey Department, The University of Melbourne, Parkville, VIC, Australia
| | - Luke M Healy
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Ranjan Dutta
- Department of Neurosciences, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Samuel Ludwin
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Jack Antel
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Michele D Binder
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, Australia
| | - Trevor J Kilpatrick
- Florey Institute of Neuroscience and Mental Health, Florey Department, The University of Melbourne, Parkville, VIC, Australia.,Department of Neurology, Royal Melbourne Hospital, Parkville, VIC, Australia
| |
Collapse
|
38
|
Carvalheiro T, Garcia S, Pascoal Ramos MI, Giovannone B, Radstake TRDJ, Marut W, Meyaard L. Leukocyte Associated Immunoglobulin Like Receptor 1 Regulation and Function on Monocytes and Dendritic Cells During Inflammation. Front Immunol 2020; 11:1793. [PMID: 32973751 PMCID: PMC7466540 DOI: 10.3389/fimmu.2020.01793] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/06/2020] [Indexed: 12/30/2022] Open
Abstract
Inhibitory receptors are crucial immune regulators and are essential to prevent exacerbated responses, thus contributing to immune homeostasis. Leukocyte associated immunoglobulin like receptor 1 (LAIR-1) is an immune inhibitory receptor which has collagen and collagen domain containing proteins as ligands. LAIR-1 is broadly expressed on immune cells and has a large availability of ligands in both circulation and tissues, implicating a need for tight regulation of this interaction. In the current study, we sought to examine the regulation and function of LAIR-1 on monocyte, dendritic cell (DC) and macrophage subtypes, using different in vitro models. We found that LAIR-1 is highly expressed on intermediate monocytes as well as on plasmacytoid DCs. LAIR-1 is also expressed on skin immune cells, mainly on tissue CD14+ cells, macrophages and CD1c+ DCs. In vitro, monocyte and type-2 conventional DC stimulation leads to LAIR-1 upregulation, which may reflect the importance of LAIR-1 as negative regulator under inflammatory conditions. Indeed, we demonstrate that LAIR-1 ligation on monocytes inhibits toll like receptor (TLR)4 and Interferon (IFN)-α- induced signals. Furthermore, LAIR-1 is downregulated on GM-CSF and IFN-γ monocyte-derived macrophages and monocyte-derived DCs. In addition, LAIR-1 triggering during monocyte derived-DC differentiation results in significant phenotypic changes, as well as a different response to TLR4 and IFN-α stimulation. This indicates a role for LAIR-1 in skewing DC function, which impacts the cytokine expression profile of these cells. In conclusion, we demonstrate that LAIR-1 is consistently upregulated on monocytes and DC during the inflammatory phase of the immune response and tends to restore its expression during the resolution phase. Under inflammatory conditions, LAIR-1 has an inhibitory function, pointing toward to a potential intervention opportunity targeting LAIR-1 in inflammatory conditions.
Collapse
Affiliation(s)
- Tiago Carvalheiro
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.,Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Samuel Garcia
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.,Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.,Rheumatology & Immuno-Mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain.,Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - M Inês Pascoal Ramos
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| | - Barbara Giovannone
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.,Department of Dermatology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Timothy R D J Radstake
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.,Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Wioleta Marut
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.,Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Linde Meyaard
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| |
Collapse
|
39
|
Scheurlen KM, Billeter AT, O'Brien SJ, Galandiuk S. Metabolic dysfunction and early-onset colorectal cancer - how macrophages build the bridge. Cancer Med 2020; 9:6679-6693. [PMID: 33624450 PMCID: PMC7520341 DOI: 10.1002/cam4.3315] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022] Open
Abstract
Background The incidence of colorectal cancer (CRC) among patients <50 years of age has increased dramatically over the last decades. At the same time, the growing proportion of obese children and adolescents and the increasing proportion of young and obese patients with CRC suggests an association between metabolic dysfunction and carcinogenesis. Tumor‐associated macrophages (TAMs) are able to orchestrate tumor promoting and suppressing mechanisms in CRC. The aim of this review was to discuss the different roles of TAMs in CRC and their phenotype‐specific metabolic pathways to identify potential new targets for CRC treatment. Methods A literature search was performed using PubMed, Cochrane and Embase to identify studies on TAMs and their metabolism in CRC. The following search terms were used in various combinations: (obesity OR adiposity OR obese) AND (macrophage OR polarization OR macrophage metabolism) AND ((colon cancer*) OR (colon carcinoma) OR (colonic tumor*) OR (colonic neoplasm[MeSH]) OR (rectal cancer*) OR (rectal carcinoma) OR (rectal tumor*) OR (rectal neoplasm[MeSH]) OR (colorectal cancer*) OR (colorectal carcinoma) OR (colorectal tumor*) OR (colorectal neoplasm[MeSH])). Studies including data on the phenotype and metabolism of TAMs in CRC were analyzed. Results Evidence for the prognostic utility of macrophage markers in CRC is currently evolving, with a particular role of stage‐dependent cellular metabolism profiles of TAMs. Itaconate is one of the metabolites produced by proinflammatory subtypes of TAMs and it is known to have tumor promoting effects. Metabolic pathways that are involved in macrophage activation and reprogramming play a role in a chronic inflammatory setting, consequently affecting the onset and development of CRC. Conclusions Tumor‐promoting metabolites, such as itaconate, are directly regulating these mechanisms, thereby triggering carcinogenesis. Metabolic reprogramming in TAMs can build a bridge between metabolic dysfunction and the onset and progression of CRC through inflammatory pathways, particularly in younger patients with early‐onset CRC.
Collapse
Affiliation(s)
- Katharina M Scheurlen
- Price Institute of Surgical Research, Department of Surgery, University of Louisville, Louisville, KY, USA
| | - Adrian T Billeter
- Department of General, Visceral, and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Baden-Wuerttemberg, Germany
| | - Stephen J O'Brien
- Price Institute of Surgical Research, Department of Surgery, University of Louisville, Louisville, KY, USA
| | - Susan Galandiuk
- Price Institute of Surgical Research, Department of Surgery, University of Louisville, Louisville, KY, USA
| |
Collapse
|
40
|
Italiani P, Mosca E, Della Camera G, Melillo D, Migliorini P, Milanesi L, Boraschi D. Profiling the Course of Resolving vs. Persistent Inflammation in Human Monocytes: The Role of IL-1 Family Molecules. Front Immunol 2020; 11:1426. [PMID: 32754155 PMCID: PMC7365847 DOI: 10.3389/fimmu.2020.01426] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 06/03/2020] [Indexed: 12/18/2022] Open
Abstract
Monocytes and macrophages have a central role in all phases of an inflammatory reaction. To understanding the regulation of monocyte activation during a physiological or pathological inflammation, we propose two in vitro models that recapitulate the different phases of the reaction (recruitment, initiation, development, and resolution vs. persistence of inflammation), based on human primary blood monocytes exposed to sequential modifications of microenvironmental conditions. These models exclusively describe the functional development of blood-derived monocytes that first enter an inflammatory site. All reaction phases were profiled by RNA-Seq, and the two models were validated by studying the modulation of IL-1 family members. Genes were differentially modulated, and distinct clusters were identified during the various phases of inflammation. Pathway analysis revealed that both models were enriched in pathways involved in innate immune activation. We observe that monocytes acquire an M1-like profile during early inflammation, and switch to a deactivated M2-like profile during both the resolving and persistent phases. However, during persistent inflammation they partially maintain an M1 profile, although they lose the ability to produce inflammatory cytokines compared to M1 cells. The production of IL-1 family molecules by ELISA reflected the transcriptomic profiles in the distinct phases of the two inflammatory reactions. Based on the results, we hypothesize that persistence of inflammatory stimuli cannot maintain the M1 activated phenotype of incoming monocytes for long, suggesting that the persistent presence of M1 cells and effects in a chronically inflamed tissue is mainly due to activation of newly incoming cells. Moreover, being IL-1 family molecules mainly expressed and secreted by monocytes during the early stages of the inflammatory response (within 4-14 h), and the rate of their production decreasing during the late phase of both resolving and persistent inflammation, we suppose that IL-1 factors are key regulators of the acute defensive innate inflammatory reaction that precedes establishment of longer-term adaptive immunity, and are mainly related to the presence of recently recruited blood monocytes. The well-described role of IL-1 family cytokines and receptors in chronic inflammation is therefore most likely dependent on the continuous influx of blood monocytes into a chronically inflamed site.
Collapse
Affiliation(s)
- Paola Italiani
- Institute of Protein Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Ettore Mosca
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
| | - Giacomo Della Camera
- Institute of Protein Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Daniela Melillo
- Institute of Protein Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Paola Migliorini
- Clinical Immunology Unit, Department Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luciano Milanesi
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
| | - Diana Boraschi
- Institute of Protein Biochemistry and Cell Biology, National Research Council, Naples, Italy
| |
Collapse
|
41
|
OGT suppresses S6K1-mediated macrophage inflammation and metabolic disturbance. Proc Natl Acad Sci U S A 2020; 117:16616-16625. [PMID: 32601203 DOI: 10.1073/pnas.1916121117] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Enhanced inflammation is believed to contribute to overnutrition-induced metabolic disturbance. Nutrient flux has also been shown to be essential for immune cell activation. Here, we report an unexpected role of nutrient-sensing O-linked β-N-acetylglucosamine (O-GlcNAc) signaling in suppressing macrophage proinflammatory activation and preventing diet-induced metabolic dysfunction. Overnutrition stimulates an increase in O-GlcNAc signaling in macrophages. O-GlcNAc signaling is down-regulated during macrophage proinflammatory activation. Suppressing O-GlcNAc signaling by O-GlcNAc transferase (OGT) knockout enhances macrophage proinflammatory polarization, promotes adipose tissue inflammation and lipolysis, increases lipid accumulation in peripheral tissues, and exacerbates tissue-specific and whole-body insulin resistance in high-fat-diet-induced obese mice. OGT inhibits macrophage proinflammatory activation by catalyzing ribosomal protein S6 kinase beta-1 (S6K1) O-GlcNAcylation and suppressing S6K1 phosphorylation and mTORC1 signaling. These findings thus identify macrophage O-GlcNAc signaling as a homeostatic mechanism maintaining whole-body metabolism under overnutrition.
Collapse
|
42
|
Bezold V, Rosenstock P, Scheffler J, Geyer H, Horstkorte R, Bork K. Glycation of macrophages induces expression of pro-inflammatory cytokines and reduces phagocytic efficiency. Aging (Albany NY) 2020; 11:5258-5275. [PMID: 31386629 PMCID: PMC6682540 DOI: 10.18632/aging.102123] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/21/2019] [Indexed: 02/06/2023]
Abstract
Glycation and the accumulation of advanced glycation end products (AGEs) are known to occur during normal aging but also in the progression of several diseases, such as diabetes. Diabetes type II and aging both lead to impaired wound healing. It has been demonstrated that macrophages play an important role in impaired wound healing, however, the underlying causes remain unknown. Elevated blood glucose levels as well as elevated methylglyoxal (MGO) levels in diabetic patients result in glycation and increase of AGEs. We used MGO to investigate the influence of glycation and AGEs on macrophages. We could show that glycation, but not treatment with AGE-modified serum proteins, increased expression of pro-inflammatory cytokines interleukin 1β (IL-1β) and IL-8 but also affected IL-10 and TNF-α expression, resulting in increased inflammation. At the same time, glycation reduced phagocytic efficiency and led to impaired clearance rates of invading microbes and cellular debris. Our data suggest that glycation contributes to changes of macrophage activity and cytokine expression and therefore could support the understanding of disturbed wound healing during aging and diabetes.
Collapse
Affiliation(s)
- Veronika Bezold
- Institute for Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Philip Rosenstock
- Institute for Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Jonas Scheffler
- Institute for Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Henriette Geyer
- Octapharma Biopharmaceuticals GmbH, Molecular Biochemistry, Berlin, Germany
| | - Rüdiger Horstkorte
- Institute for Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Kaya Bork
- Institute for Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
43
|
Zhu G, Zhao G, Lin J, Li C, Wang Q, Xu Q, Peng X, Zheng H. FCN-A mediates the inflammatory response and the macrophage polarization in Aspergillus fumigatus keratitis of mice by activating the MAPK signaling pathway. Int Immunopharmacol 2020; 83:106473. [PMID: 32272397 DOI: 10.1016/j.intimp.2020.106473] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/26/2020] [Accepted: 04/01/2020] [Indexed: 02/05/2023]
Abstract
Fungal keratitis (FK) is a severe corneal disease that may cause vision loss. Previous studies indicate that the innate immune response produces the most effective anti-Aspergillus immune resistance. Ficolin-A (FCN-A), a soluble pattern-recognition receptor (PRR) family plays an important role in the innate immunity. In this study, we aimed to study the role of FCN-A in the A. fumigatus infected cornea. Here for the first time, we reported that the expression of FCN-A increases after A. fumigatus infection in the cornea of mice. Then, our results showed that the down-regulation of FCN-A reduced the inflammatory response of the cornea infected mice and decreased the expression of the TNF-a, p-p38, p-JNK. We also found that FCN-A can affect the recruitment of macrophages in the cornea of mice with A. fumigatus keratitis. In the mouse model of A. fumigatus keratitis and the A. fumigatus stimulation of RAW 264.7 cells, knocking down of FCN-A expression promoted the macrophage polarization toward M2. Furthermore, we observed that both the p38 and JNK inhibitors pretreatment decreased the proportion of M1/M2 in RAW 264.7 cells. Taken together, our data provide evidence that FCN-A participated in the inflammatory response of A. fumigatus keratitis in mice. Moreover, FCN-A mediates the inflammatory response and the polarization of the macrophages by activating the MAPK signaling pathway in A. fumigatus keratitis.
Collapse
Affiliation(s)
- Guoqiang Zhu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China.
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Cui Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Qian Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Qiang Xu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Xudong Peng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| | - Hengrui Zheng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266003, China
| |
Collapse
|
44
|
Bianchetto-Aguilera F, Tamassia N, Gasperini S, Calzetti F, Finotti G, Gardiman E, Montioli R, Bresciani D, Vermi W, Cassatella MA. Deciphering the fate of slan + -monocytes in human tonsils by gene expression profiling. FASEB J 2020; 34:9269-9284. [PMID: 32413173 DOI: 10.1096/fj.202000181r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/24/2020] [Accepted: 04/24/2020] [Indexed: 12/24/2022]
Abstract
Monocytic cells perform crucial homeostatic and defensive functions. However, their fate and characterization at the transcriptomic level in human tissues are partially understood, often as a consequence of the lack of specific markers allowing their unequivocal identification. The 6-sulfo LacNAc (slan) antigen identifies a subset of non-classical (NC) monocytes in the bloodstream, namely the slan+ -monocytes. In recent studies, we and other groups have reported that, in tonsils, slan marks dendritic cell (DC)-like cells, as defined by morphological, phenotypical, and functional criteria. However, subsequent investigations in lymphomas have uncovered a significant heterogeneity of tumor-infiltrating slan+ -cells, including a macrophage-like state. Based on their emerging role in tissue inflammation and cancer, herein we investigated slan+ -cell fate in tonsils by using a molecular-based approach. Hence, RNA from tonsil slan+ -cells, conventional CD1c+ DCs (cDC2) and CD11b+ CD14+ -macrophages was subjected to gene expression analysis. For comparison, transcriptomes were also obtained from blood cDC2, classical (CL), intermediate (INT), NC, and slan+ -monocytes. Data demonstrate that the main trajectory of human slan+ -monocytes infiltrating the tonsil tissue is toward a macrophage-like population, displaying molecular features distinct from those of tonsil CD11b+ CD14+ -macrophages and cDC2. These findings provide a novel view on the terminal differentiation path of slan+ -monocytes, which is relevant for inflammatory diseases and lymphomas.
Collapse
Affiliation(s)
| | - Nicola Tamassia
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Sara Gasperini
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Federica Calzetti
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Giulia Finotti
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Elisa Gardiman
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Riccardo Montioli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Debora Bresciani
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, Brescia, Italy
| | - William Vermi
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, Brescia, Italy.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | |
Collapse
|
45
|
Duncan SE, Gao S, Sarhene M, Coffie JW, Linhua D, Bao X, Jing Z, Li S, Guo R, Su J, Fan G. Macrophage Activities in Myocardial Infarction and Heart Failure. Cardiol Res Pract 2020; 2020:4375127. [PMID: 32377427 PMCID: PMC7193281 DOI: 10.1155/2020/4375127] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/21/2020] [Accepted: 01/27/2020] [Indexed: 02/06/2023] Open
Abstract
Heart diseases remain the major cause of death worldwide. Advances in pharmacological and biomedical management have resulted in an increasing proportion of patients surviving acute heart failure (HF). However, many survivors of HF in the early stages end up increasing the disease to chronic HF (CHF). HF is an established frequent complication of myocardial infarction (MI), and numerous influences including persistent myocardial ischemia, shocked myocardium, ventricular remodeling, infarct size, and mechanical impairments, as well as hibernating myocardium trigger the development of left ventricular systolic dysfunction following MI. Macrophage population is active in inflammatory process, yet the clear understanding of the causative roles for these macrophage cells in HF development and progression is actually incomplete. Long ago, it was thought that macrophages are of importance in the heart after MI. Also, though inflammation is as a result of adverse HF in patients, but despite the fact that broad immunosuppression therapeutic target has been used in various clinical trials, no positive results have showed up, but rather, the focus on proinflammatory cytokines has proved more benefits in patients with HF. Therefore, in this review, we discuss the recent findings and new development about macrophage activations in HF, its role in the healthy heart, and some therapeutic targets for myocardial repair. We have a strong believe that there is a need to give maximum attention to cardiac resident macrophages due to the fact that they perform various tasks in wound healing, self-renewal of the heart, and tissue remodeling. Currently, it has been discovered that the study of macrophages goes far beyond its phagocytotic roles. If researchers in future confirm that macrophages play a vital role in the heart, they can be therapeutically targeted for cardiac healing.
Collapse
Affiliation(s)
- Sophia Esi Duncan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Shan Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Michael Sarhene
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Joel Wake Coffie
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Deng Linhua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Xingru Bao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Zhang Jing
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Sheng Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Rui Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Jing Su
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| |
Collapse
|
46
|
Kim BS, Tilstam PV, Arnke K, Leng L, Ruhl T, Piecychna M, Schulte W, Sauler M, Frueh FS, Storti G, Lindenblatt N, Giovanoli P, Pallua N, Bernhagen J, Bucala R. Differential regulation of macrophage activation by the MIF cytokine superfamily members MIF and MIF-2 in adipose tissue during endotoxemia. FASEB J 2020; 34:4219-4233. [PMID: 31961019 PMCID: PMC7060131 DOI: 10.1096/fj.201901511r] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/27/2019] [Accepted: 01/07/2020] [Indexed: 01/01/2023]
Abstract
Sepsis is a leading cause of death worldwide and recent studies have shown white adipose tissue (WAT) to be an important regulator in septic conditions. In the present study, the role of the inflammatory cytokine macrophage migration inhibitory factor (MIF) and its structural homolog D-dopachrome tautomerase (D-DT/MIF-2) were investigated in WAT in a murine endotoxemia model. Both MIF and MIF-2 levels were increased in the peritoneal fluid of LPS-challenged wild-type mice, yet, in visceral WAT, the proteins were differentially regulated, with elevated MIF but downregulated MIF-2 expression in adipocytes. Mif gene deletion polarized adipose tissue macrophages (ATM) toward an anti-inflammatory phenotype while Mif-2 gene knockout drove ATMs toward a pro-inflammatory phenotype and Mif-deficiency was found to increase fibroblast viability. Additionally, we observed the same differential regulation of these two MIF family proteins in human adipose tissue in septic vs healthy patients. Taken together, these data suggest an inverse relationship between adipocyte MIF and MIF-2 expression during systemic inflammation, with the downregulation of MIF-2 in fat tissue potentially increasing pro-inflammatory macrophage polarization to further drive adipose inflammation.
Collapse
Affiliation(s)
- Bong-Sung Kim
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
- Department of Plastic, Reconstructive and Hand Surgery, RWTH Aachen University, Aachen, Germany
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Switzerland
| | - Pathricia V. Tilstam
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Kevin Arnke
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Switzerland
| | - Lin Leng
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Tim Ruhl
- Department of Plastic, Reconstructive and Hand Surgery, RWTH Aachen University, Aachen, Germany
| | - Marta Piecychna
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Wibke Schulte
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
- Department of Surgery, Yale University School of Medicine, New Haven, CT
- Department of Surgery, Campus Charité Mitte | Campus Virchow-Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Maor Sauler
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, CT
| | - Florian S. Frueh
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Switzerland
| | - Gabriele Storti
- Unit of Plastic and Reconstructive Surgery University of Rome- “Tor Vergata”
| | - Nicole Lindenblatt
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Switzerland
| | - Pietro Giovanoli
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Switzerland
| | - Norbert Pallua
- Department of Plastic, Reconstructive and Hand Surgery, RWTH Aachen University, Aachen, Germany
| | - Jürgen Bernhagen
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig-Maximilians-University Munich, Munich, Germany
- Munich Cluster for Systems Neurology, 81377 Munich, Germany
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
47
|
Rao Muvva J, Parasa VR, Lerm M, Svensson M, Brighenti S. Polarization of Human Monocyte-Derived Cells With Vitamin D Promotes Control of Mycobacterium tuberculosis Infection. Front Immunol 2020; 10:3157. [PMID: 32038652 PMCID: PMC6987394 DOI: 10.3389/fimmu.2019.03157] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 12/31/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Understanding macrophage behavior is key to decipher Mycobacterium tuberculosis (Mtb) pathogenesis. We studied the phenotype and ability of human monocyte-derived cells polarized with active vitamin D [1,25(OH)2D3] to control intracellular Mtb infection compared with polarization of conventional subsets, classical M1 or alternative M2. Methods: Human blood-derived monocytes were treated with active vitamin D or different cytokines to obtain 1,25(OH)2D3-polarized as well as M1- and M2-like cells or fully polarized M1 and M2 subsets. We used an in vitro macrophage Mtb infection model to assess both phenotype and functional markers i.e., inhibitory and scavenger receptors, costimulatory molecules, cytokines, chemokines, and effector molecules using flow cytometry and quantitative mRNA analysis. Intracellular uptake of bacilli and Mtb growth was monitored using flow cytometry and colony forming units. Results: Uninfected M1 subsets typically expressed higher levels of CCR7, TLR2, and CD86, while M2 subsets expressed higher CD163, CD200R, and CD206. Most of the investigated markers were up-regulated in all subsets after Mtb infection, generating a mixed M1/M2 phenotype, while the expression of CD206, HLADR, and CD80 was specifically up-regulated (P < 0.05) on 1,25(OH)2D3-polarized macrophages. Consistent with the pro-inflammatory features of M1 cells, Mtb uptake and intracellular Mtb growth was significantly (P < 0.01–0.001 and P < 0.05–0.01) lower in the M1 (19.3%) compared with the M2 (82.7%) subsets 4 h post-infection. However, infectivity rapidly and gradually increased in M1 cells at 24–72 h. 1,25(OH)2D3-polarized monocyte-derived cells was the most potent subset to inhibit Mtb growth at both 4 and 72 h (P < 0.05–0.01) post-Mtb infection. This ability was associated with high mRNA levels of pro-inflammatory cytokines and the antimicrobial peptide LL-37 but also anti-inflammatory IL-10, while expression of the immunosuppressive enzyme IDO (indoleamine 2,3-dioxygenase) remained low in Mtb-infected 1,25(OH)2D3-polarized cells compared with the other subsets. Conclusions: Mtb infection promoted a mixed M1/M2 macrophage activation, and 1,25(OH)2D3-polarized monocyte-derived cells expressing LL-37 but not IDO, were most effective to control intracellular Mtb growth. Macrophage polarization in the presence of vitamin D may provide the capacity to mount an antimicrobial response against Mtb and simultaneously prevent expression of inhibitory molecules that could accelerate local immunosuppression in the microenvironment of infected tissue.
Collapse
Affiliation(s)
- Jagadeeswara Rao Muvva
- Department of Medicine, Center for Infectious Medicine (CIM), ANA Futura, Karolinska Institutet, Stockholm, Sweden
| | | | - Maria Lerm
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Mattias Svensson
- Department of Medicine, Center for Infectious Medicine (CIM), ANA Futura, Karolinska Institutet, Stockholm, Sweden
| | - Susanna Brighenti
- Department of Medicine, Center for Infectious Medicine (CIM), ANA Futura, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
48
|
Chisari E, Rehak L, Khan WS, Maffulli N. Tendon healing in presence of chronic low-level inflammation: a systematic review. Br Med Bull 2019; 132:97-116. [PMID: 31838495 DOI: 10.1093/bmb/ldz035] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/19/2019] [Accepted: 10/21/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Tendinopathy is a common musculoskeletal condition affecting subjects regardless of their activity level. Multiple inflammatory molecules found in ex vivo samples of human tendons are related to the initiation or progression of tendinopathy. Their role in tendon healing is the subject of this review. SOURCES OF DATA An extensive review of current literature was conducted using PubMed, Embase and Cochrane Library using the term 'tendon', as well as some common terms of tendon conditions such as 'tendon injury OR (tendon damage) OR tendonitis OR tendinopathy OR (chronic tendonitis) OR tendinosis OR (chronic tendinopathy) OR enthesitis' AND 'healing' AND '(inflammation OR immune response)' as either key words or MeSH terms. AREAS OF AGREEMENT An environment characterized by a low level of chronic inflammation, together with increased expression of inflammatory cytokines and growth factors, may influence the physiological tendon healing response after treatment. AREAS OF CONTROVERSY Most studies on this topic exhibited limited scientific translational value because of their heterogeneity. The evidence associated with preclinical studies is limited. GROWING POINTS The role of inflammation in tendon healing is still unclear, though it seems to affect the overall outcome. A thorough understanding of the biochemical mediators of healing and their pathway of pain could be used to target tendinopathy and possibly guide its management. AREAS TIMELY FOR DEVELOPING RESEARCH We require further studies with improved designs to effectively evaluate the pathogenesis and progression of tendinopathy to identify cellular and molecular targets to improve outcomes.
Collapse
Affiliation(s)
- Emanuele Chisari
- University of Catania, Departmento of General Surgery and Medical Specialities, Via Santa Sofia 78, Catania 95123, Italy
| | - Laura Rehak
- Athena Biomedical Innovations, Viale Europa 139, Florence, 50126, Italy
| | - Wasim S Khan
- Division of Trauma and Orthopaedics, Addenbrooke's Hospital, University of Cambridge, Hills Rd, Cambridge CB2 0QQ, United Kingdom
| | - Nicola Maffulli
- Department of Musculoskeletal Disorders, Via Salvador Allende, 43, 84081 Baronissi SA, Italy, Salerno, Italy.,Clinica Ortopedica, Ospedale San Giovanni di Dio e Ruggi D'Aragona, Largo Città di Ippocrate, Salerno, 84131, Italy.,Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Mile End Hospital, Queen Mary University of London, 275 Bancroft Road, London E1 4DG, England.,School of Medicine, Institute of Science and Technology in Medicine, Guy Hilton Research Centre, Keele University, Thornburrow Drive, Hartshill, Stoke-on-Trent ST4 7QB, England
| |
Collapse
|
49
|
Österlund P, Jiang M, Westenius V, Kuivanen S, Järvi R, Kakkola L, Lundberg R, Melén K, Korva M, Avšič-Županc T, Vapalahti O, Julkunen I. Asian and African lineage Zika viruses show differential replication and innate immune responses in human dendritic cells and macrophages. Sci Rep 2019; 9:15710. [PMID: 31673117 PMCID: PMC6823455 DOI: 10.1038/s41598-019-52307-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/16/2019] [Indexed: 12/14/2022] Open
Abstract
Zika virus (ZIKV) infections in humans are considered to be mild or subclinical. However, during the recent epidemics in the Pacific Islands and the Americas, the infection was associated with Quillain-Barré syndrome and congenital infections with fetal brain abnormalities, including microcephaly. Thus, more detailed understanding of ZIKV-host cell interactions and regulation of innate immune responses by strains of differential evolutionary origin is required. Here, we characterized the infection and immune responses triggered by two epidemic Asian/American lineage viruses, including an isolate from fetal brains, and a historical, low passage 1947 African lineage virus in human monocyte-derived dendritic cells (DCs) and macrophages. The epidemic Asian/American ZIKV replicated well and induced relatively good antiviral responses in human DCs whereas the African strain replicated less efficiently and induced weaker immune responses. In macrophages both the African and Asian strains showed limited replication and relatively weak cytokine gene expression. Interestingly, in macrophages we observed host protein degradation, especially IRF3 and STAT2, at early phases of infection with both lineage viruses, suggesting an early proteasomal activation in phagocytic cells. Our data indicates that ZIKV evolution has led to significant phenotypic differences in the replication characteristics leading to differential regulation of host innate immune responses.
Collapse
Affiliation(s)
- Pamela Österlund
- Expert Microbiology Unit, Finnish Institute for Health and Welfare, 00300, Helsinki, Finland.
| | - Miao Jiang
- Expert Microbiology Unit, Finnish Institute for Health and Welfare, 00300, Helsinki, Finland
| | - Veera Westenius
- Expert Microbiology Unit, Finnish Institute for Health and Welfare, 00300, Helsinki, Finland
| | - Suvi Kuivanen
- Department of Virology, University of Helsinki, 00290, Helsinki, Finland
| | - Riia Järvi
- Expert Microbiology Unit, Finnish Institute for Health and Welfare, 00300, Helsinki, Finland
| | - Laura Kakkola
- Institute of Biomedicine, University of Turku and Turku University Hospital, 20520, Turku, Finland
| | - Rickard Lundberg
- Institute of Biomedicine, University of Turku and Turku University Hospital, 20520, Turku, Finland
| | - Krister Melén
- Expert Microbiology Unit, Finnish Institute for Health and Welfare, 00300, Helsinki, Finland.,Institute of Biomedicine, University of Turku and Turku University Hospital, 20520, Turku, Finland
| | - Miša Korva
- Faculty of Medicine, Institute of Microbiology and Immunology, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Tatjana Avšič-Županc
- Faculty of Medicine, Institute of Microbiology and Immunology, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Olli Vapalahti
- Department of Virology, University of Helsinki, 00290, Helsinki, Finland
| | - Ilkka Julkunen
- Institute of Biomedicine, University of Turku and Turku University Hospital, 20520, Turku, Finland
| |
Collapse
|
50
|
Anesi A, Generali L, Sandoni L, Pozzi S, Grande A. From Osteoclast Differentiation to Osteonecrosis of the Jaw: Molecular and Clinical Insights. Int J Mol Sci 2019; 20:ijms20194925. [PMID: 31590328 PMCID: PMC6801843 DOI: 10.3390/ijms20194925] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 01/05/2023] Open
Abstract
Bone physiology relies on the delicate balance between resorption and formation of its tissue. Bone resorption depends on a process called osteoclastogenesis in which bone-resorbing cells, i.e., osteoclasts, are produced by the differentiation of more undifferentiated progenitors and precursors. This process is governed by two main factors, monocyte-colony stimulating factor (M-CSF) and receptor activator of NFκB ligand (RANKL). While the former exerts a proliferating effect on progenitors/precursors, the latter triggers a differentiation effect on more mature cells of the same lineage. Bone homeostasis requires a perfect space–time coordination of the involved signals. When osteoclastogenesis is poorly balanced with the differentiation of the bone forming counterparts, i.e., osteoblasts, physiological bone remodelling can turn into a pathological state, causing the systematic disruption of bone tissue which results in osteopenia or osteolysis. Examples of these conditions are represented by osteoporosis, Paget’s disease, bone metastasis, and multiple myeloma. Therefore, drugs targeting osteoclastogenesis, such as bisphosphonates and an anti-RANKL monoclonal antibody, have been developed and are currently used in the treatment of such diseases. Despite their demonstrated therapeutic efficacy, these agents are unfortunately not devoid of side effects. In this regard, a condition called osteonecrosis of the jaw (ONJ) has been recently correlated with anti-resorptive therapy. In this review we will address the involvement of osteoclasts and osteoclast-related factors in the pathogenesis of ONJ. It is to be hoped that a better understanding of the biological mechanisms underlying bone remodelling will help in the design a medical therapeutic approach for ONJ as an alternative to surgical procedures.
Collapse
Affiliation(s)
- Alexandre Anesi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy.
| | - Luigi Generali
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Transplant Surgery, Oncology and Regenerative Medicine Relevance, University of Modena and Reggio Emilia, 41121 Modena, Italy.
| | - Laura Sandoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 287, 41125 Modena, Italy.
| | - Samantha Pozzi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy.
| | - Alexis Grande
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 287, 41125 Modena, Italy.
| |
Collapse
|