201
|
Inokuchi S, Mitoma H, Kawano S, Nakano S, Ayano M, Kimoto Y, Akahoshi M, Arinobu Y, Tsukamoto H, Akashi K, Horiuchi T, Niiro H. Homeostatic Milieu Induces Production of Deoxyribonuclease 1–like 3 from Myeloid Cells. THE JOURNAL OF IMMUNOLOGY 2020; 204:2088-2097. [DOI: 10.4049/jimmunol.1901304] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/12/2020] [Indexed: 12/31/2022]
|
202
|
Samal SK, Qureshi AR, Rahman M, Stenvinkel P, Frostegård J. Antibodies against Malondialdehyde in Haemodialysis Patients and Its Association with Clinical Outcomes: Differences between Subclasses and Isotypes. J Clin Med 2020; 9:jcm9030753. [PMID: 32168733 PMCID: PMC7141181 DOI: 10.3390/jcm9030753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 01/22/2023] Open
Abstract
Patients on haemodialysis (HD-patients) have an increased risk of premature death. Low levels of IgM antibodies against malondialdehyde (anti-MDA) are associated with increased risk of cardiovascular disease (CVD) with underlying potential mechanisms described. Here, we studied subclasses and isotypes of anti-MDA in 210 HD-patients with mortality as outcome (56% men, median age 66, Interquartile range (IQR) 51-74 years, vintage time 29 (15-58) months, mean follow up period of 41 (20-60)months). Patients were also divided into inflamed c-reactive protein (CRP >5.6 mg/mL) and non-inflamed. Antibody levels were measured by ELISA. In multivariate risk analysis, patients in low tertile of IgM anti-MDA sub-distribution hazard ratio (sHR 0.54); 95% confidence interval (CI: 0.34-0.89) inversely and significantly associated with all-cause mortality after five years, after adjusting for confounders. Low tertile of IgG (sHR 0.48, 95%CI: 0.25-0.90, p = 0.02) and IgG1 (sHR 0.50, CI: 0.24-1.04, p = 0.06) was associated low mortality among non-inflamed patients. In contrast, anti-MDA IgG2 among inflamed patients was significantly associated with increased mortality, IgG2(sHR 2.33, CI: 1.16-4.68, p = 0.01). IgM anti-MDA was a novel biomarker among HD-patients with low levels being associated with mortality, while low levels of IgG and IgG1 but not IgA anti-MDA were associated with mortality only among non-inflamed patients. IgG2 anti-MDA was a significant risk marker among inflamed patients, which could be related to infection.
Collapse
Affiliation(s)
- Shailesh Kumar Samal
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden; (S.K.S.); (M.R.)
| | - Abdul Rashid Qureshi
- Divisions of Renal Medicine, Department of Clinical Science, Intervention and Technology, 14152 Huddinge, Sweden; (A.R.Q.); (P.S.)
| | - Mizanur Rahman
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden; (S.K.S.); (M.R.)
| | - Peter Stenvinkel
- Divisions of Renal Medicine, Department of Clinical Science, Intervention and Technology, 14152 Huddinge, Sweden; (A.R.Q.); (P.S.)
| | - Johan Frostegård
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden; (S.K.S.); (M.R.)
- Correspondence:
| |
Collapse
|
203
|
Abstract
Phagocytosis is a pivotal immunological process, and its discovery by Elia Metchnikoff in 1882 was a step toward the establishment of the innate immune system as a separate branch of immunology. Elia Metchnikoff received the Nobel Prize in physiology and medicine for this discovery in 1908. Since its discovery almost 140 years before, phagocytosis remains the hot topic of research in immunology. The phagocytosis research has seen a great advancement since its first discovery. Functionally, phagocytosis is a simple immunological process required to engulf and remove pathogens, dead cells and tumor cells to maintain the immune homeostasis. However, mechanistically, it is a very complex process involving different mechanisms, induced and regulated by several pattern recognition receptors, soluble pattern recognition molecules, scavenger receptors (SRs) and opsonins. These mechanisms involve the formation of phagosomes, their maturation into phagolysosomes causing pathogen destruction or antigen synthesis to present them to major histocompatibility complex molecules for activating an adaptive immune response. Any defect in this mechanism may predispose the host to certain infections and inflammatory diseases (autoinflammatory and autoimmune diseases) along with immunodeficiency. The article is designed to discuss its mechanistic complexity at each level, varying from phagocytosis induction to phagolysosome resolution.
Collapse
Affiliation(s)
- Vijay Kumar
- Faculty of Medicine, Children's Health Queensland Clinical Unit, School of Clinical Medicine, Mater Research, University of Queensland, ST Lucia, Brisbane, Queensland, Australia.,Faculty of Medicine, School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
204
|
Li J, Cai Z, Cheng J, Wang C, Fang Z, Xiao Y, Feng ZG, Gu Y. Characterization of a heparinized decellularized scaffold and its effects on mechanical and structural properties. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2020; 31:999-1023. [PMID: 32138617 DOI: 10.1080/09205063.2020.1736741] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Decellularization is a promising approach in tissue engineering to generate small-diameter blood vessels. However, some challenges still exist. We performed two decellularization phases to develop an optimal decellularized scaffold and analyze the relationship between the extracellular matrix (ECM) composition and mechanical properties. In decellularization phase I, we tested sodium dodecylsulfate (SDS), Triton X-100 (TX100) and trypsin at different concentrations and exposure times. In decellularization phase II, we systematically compared five combined decellularization protocols based on the results of phase I to identify the optimal method. These protocols tested cell removal, ECM preservation, mechanical properties, and residual cytotoxicity. We further immobilized heparin to optimal decellularized scaffolds and determined its anticoagulant activity and mechanical properties. The combined decellularization protocol comprising treatment with 0.5% SDS followed by 1% TX100 could completely remove the cellular contents and preserve the mechanical properties and ECM architecture better. In addition, the heparinized decellularized scaffolds not only had sustained anticoagulant activity, but also similar mechanical properties to native vessels. In conclusion, heparinized decellularized scaffolds represent a promising direction for small-diameter vascular grafts, although further in vivo studies are needed.
Collapse
Affiliation(s)
- Ji Li
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhiwen Cai
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jin Cheng
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Cong Wang
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhiping Fang
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, China
| | - Yonghao Xiao
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, China
| | - Zeng-Guo Feng
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, China
| | - Yongquan Gu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
205
|
Riau AK, Liu YC, Yam GH, Mehta JS. Stromal keratophakia: Corneal inlay implantation. Prog Retin Eye Res 2020; 75:100780. [DOI: 10.1016/j.preteyeres.2019.100780] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/28/2019] [Accepted: 09/02/2019] [Indexed: 12/31/2022]
|
206
|
Phagocytosis assays with different pH-sensitive fluorescent particles and various readouts. Biotechniques 2020; 68:245-250. [PMID: 32079414 DOI: 10.2144/btn-2020-0003] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Phagocytosis is a fundamental mechanism of innate immunity and its impairment is associated with severe chronic diseases, for example, chronic obstructive pulmonary disease. Investigating phagocytosis requires flexible tools and assay conditions, such as different fluorescent particle types, detection colors and readouts. We comprehensively evaluated and optimized phagocytosis assays using particles labeled with fluorescent pH-sensitive pHrodo® dyes, facilitating the specific detection of phagocytosed particles. Beads, bacterial and yeast particles labeled with pHrodo red and green were tested for their uptake by THP-1 cells and primary human macrophages by flow cytometry and high-content imaging. Whereas the latter allowed kinetic phagocytosis measurement, the former demonstrated the feasibility of using cell sorting for periods of up to 6 h, enabling downstream applications such as pooled genetic screens.
Collapse
|
207
|
Abstract
Despite an increase in the rates of survival in patients suffering myocardial infarction, as yet there is no therapy specifically targeting ischaemia and reperfusion injury of the myocardium. With a greater understanding of immune activation during infarction, more potential treatment targets are now being identified. The innate immune system is believed to play an important role in the myocardium after ischaemia-driven cardiomyocyte death. The release of intracellular contents including DNA into the extracellular space during necrosis and cell rupture is now believed to create a pro-inflammatory milieu which propagates the inflammatory process. DNA and DNA fragments have been shown to activate the innate immune system by acting as Danger-Associated Molecular Patterns (DAMPs), which act as ligands on toll-like receptors (TLRs). Stimulation of TLRs, in turn, can activate intracellular cell death pathways such as pyroptosis. Here, we review the role of DNA fragments during ischaemia and reperfusion, and assess their potential as a target in the quest to preserve cardiomyocyte viability following myocardial infarction.
Collapse
Affiliation(s)
- Mohammed Shah
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
208
|
Nagata S, Sakuragi T, Segawa K. Flippase and scramblase for phosphatidylserine exposure. Curr Opin Immunol 2020; 62:31-38. [DOI: 10.1016/j.coi.2019.11.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 11/25/2019] [Indexed: 01/30/2023]
|
209
|
Lipoxin A 4 suppresses angiotensin II type 1 receptor autoantibody in preeclampsia via modulating caspase-1. Cell Death Dis 2020; 11:78. [PMID: 32001671 PMCID: PMC6992755 DOI: 10.1038/s41419-020-2281-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 02/06/2023]
Abstract
Preeclampsia (PE) remains a leading cause of maternal and neonatal morbidity and mortality. Numerous studies have shown that women with PE develop autoantibody, termed angiotensin II type 1 receptor autoantibody (AT1-AA), and key features of the disease result from it. Emerging evidence has indicated that inflammatory cell necrosis, such as pyroptosis, could lead to autoantigen exposure and stimulate autoantibody production. Caspase-1, the central enzyme of inflammasome and key target of pyroptosis, may play roles in AT1R exposure and AT1-AA production. Exploring endogenous regulator that could inhibit AT1-AA production by targeting pyroptosis will be essential for treating PE. Lipoxin A4 (LXA4), endogenous dual anti-inflammatory and proresolving lipid mediator, may inhibit AT1-AA production via modulating caspase-1. Thus, we explore whether caspase-1 is essential for AT1-AA production and LXA4 inhibits AT1-AA via modulating caspase-1. PE patients and mice developed AT1-AA associated with caspase-1 activation. Caspase-1 deletion leaded to AT1-AA decrease in PE mice. Consistent with these findings, we confirmed caspase-1 activation, trophoblast pyroptosis and AT1R exposure in PE mice and trophoblast model, while caspase-1 deficiency showed decreased trophoblast pyroptosis and AT1R exposure in vitro and in vivo. Interestingly, LXA4 could suppress AT1-AA production via regulating caspase-1 as well as enhancing phagocytosis of dead trophoblasts by macrophages. These results suggest that caspase-1 promotes AT1-AA production via inducing trophoblast pyroptosis and AT1R exposure, while LXA4 suppresses AT1-AA production via modulating caspase-1, supporting caspase-1 serving as a therapeutic target for attenuating AT1-AA and LXA4 protecting patients from AT1-AA and PE.
Collapse
|
210
|
Liu H, Liu S, Qiu X, Yang X, Bao L, Pu F, Liu X, Li C, Xuan K, Zhou J, Deng Z, Liu S, Jin Y. Donor MSCs release apoptotic bodies to improve myocardial infarction via autophagy regulation in recipient cells. Autophagy 2020; 16:2140-2155. [PMID: 31959090 DOI: 10.1080/15548627.2020.1717128] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation has been widely applied as a potential therapeutic for multiple diseases. However, the underlying therapeutic mechanisms are not fully understood, especially the paradox between the low survival rate of transplanted cells and the beneficial therapeutic effects generated by these cells. Herein, in a myocardial infarction (MI) model, we found that transplanted MSCs released apoptotic bodies (ABs) to enhance angiogenesis and improve cardiac functional reclovery via regulating macroautophagy/autophagy in the recipient endothelial cells (ECs). Mechanistically, after local transplantation, MSCs underwent extensive apoptosis in the short term and released ABs, which were engulfed by the recipient ECs. Then, in the ECs, ABs activated lysosome functions and promoted the expression of TFEB (transcription factor EB), which is a master gene in lysosomal biogenesis and autophagy. Finally, the increase in TFEB enhanced autophagy-related gene expression in ECs and promoted angiogenesis and cardiac functional recovery after MI. Collectively, we found that apoptotic donor MSCs promote angiogenesis via regulating autophagy in the recipient ECs, unveiling the role of donor cell apoptosis in the therapeutic effects generated by cell transplantation. Abbreviations: 3-MA: 3-methyladenine; ABs: apoptotic bodies; BECN1: beclin 1; CASP3: caspase 3; CQ: chloroquine; ECs: endothelial cells; EVs: extracellular vesicles; LAMP1: lysosomal-associated membrane protein 1; LVEF: left ventricular ejection fraction; LVFS: left ventricular fractional shortening; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MI: myocardial infarction; MSC: mesenchymal stem cell; NO: nitric oxide; TFEB: transcription factor EB; TUNEL: TdT-mediated dUTP Nick-End Labeling.
Collapse
Affiliation(s)
- Huan Liu
- Department of Otolaryngology, Xijing Hospital, The Fourth Military Medical University , Xi'an, Shaanxi, China.,State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University , Xi'an, Shaanxi, China
| | - Siying Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, the Fourth Military Medical University , Xi'an, Shaanxi, China
| | - Xinyu Qiu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University , Xi'an, Shaanxi, China
| | - Xiaoshan Yang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University , Xi'an, Shaanxi, China
| | - Lili Bao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University , Xi'an, Shaanxi, China
| | - Fengxing Pu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University , Xi'an, Shaanxi, China
| | - Xuemei Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University , Xi'an, Shaanxi, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University , Xi'an, Shaanxi, China
| | - Kun Xuan
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University , Xi'an, Shaanxi, China
| | - Jun Zhou
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University , Xi'an, Shaanxi, China
| | - Zhihong Deng
- Department of Otolaryngology, Xijing Hospital, The Fourth Military Medical University , Xi'an, Shaanxi, China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University , Xi'an, Shaanxi, China
| | - Yan Jin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, The Fourth Military Medical University , Xi'an, Shaanxi, China
| |
Collapse
|
211
|
Chan LLY, Rice WL, Qiu J. Observation and quantification of the morphological effect of trypan blue rupturing dead or dying cells. PLoS One 2020; 15:e0227950. [PMID: 31978129 PMCID: PMC6980413 DOI: 10.1371/journal.pone.0227950] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/03/2020] [Indexed: 11/19/2022] Open
Abstract
Trypan blue has long been the gold standard for staining dead cell to determine cell viability. The dye is excluded from membrane-intact live cells, but can enter and concentrate in membrane-compromised dead cells, rendering the cells dark blue. Over the years, there has been an understanding that trypan blue is inaccurate for cell viability under 80% without scientific support. We previously showed that trypan blue can alter the morphology of dead cells to a diffuse shape, which can lead to over-estimation of viability. Here, we investigate the origin of the dim and diffuse objects after trypan blue staining. Utilizing image and video acquisition, we show real-time transformation of cells into diffuse objects when stained with trypan blue. The same phenomenon was not observed when staining cells with propidium iodide. We also demonstrate the co-localization of trypan blue and propidium iodide, confirming these diffuse objects as cells that contain nuclei. The videos clearly show immediate cell rupturing after trypan blue contact. The formation of these diffuse objects was monitored and counted over time as cells die outside of the incubator. We hypothesize and demonstrate that rapid water influx may have caused the cells to rupture and disappear. Since some dead cells disappear after trypan blue staining, the total can be under-counted, leading to over-estimation of cell viability. This inaccuracy could affect the outcomes of cellular therapies, which require accurate measurements of immune cells that will be infused back into patients.
Collapse
Affiliation(s)
- Leo Li-Ying Chan
- Department of Advanced Technology R&D, Nexcelom Bioscience LLC., Lawrence, Massachusetts, United States of America
- * E-mail:
| | - William L. Rice
- Department of Advanced Technology R&D, Nexcelom Bioscience LLC., Lawrence, Massachusetts, United States of America
| | - Jean Qiu
- Department of Advanced Technology R&D, Nexcelom Bioscience LLC., Lawrence, Massachusetts, United States of America
| |
Collapse
|
212
|
Bäck M, Yurdagul A, Tabas I, Öörni K, Kovanen PT. Inflammation and its resolution in atherosclerosis: mediators and therapeutic opportunities. Nat Rev Cardiol 2020; 16:389-406. [PMID: 30846875 DOI: 10.1038/s41569-019-0169-2] [Citation(s) in RCA: 641] [Impact Index Per Article: 128.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a lipid-driven inflammatory disease of the arterial intima in which the balance of pro-inflammatory and inflammation-resolving mechanisms dictates the final clinical outcome. Intimal infiltration and modification of plasma-derived lipoproteins and their uptake mainly by macrophages, with ensuing formation of lipid-filled foam cells, initiate atherosclerotic lesion formation, and deficient efferocytotic removal of apoptotic cells and foam cells sustains lesion progression. Defective efferocytosis, as a sign of inadequate inflammation resolution, leads to accumulation of secondarily necrotic macrophages and foam cells and the formation of an advanced lesion with a necrotic lipid core, indicative of plaque vulnerability. Resolution of inflammation is mediated by specialized pro-resolving lipid mediators derived from omega-3 fatty acids or arachidonic acid and by relevant proteins and signalling gaseous molecules. One of the major effects of inflammation resolution mediators is phenotypic conversion of pro-inflammatory macrophages into macrophages that suppress inflammation and promote healing. In advanced atherosclerotic lesions, the ratio between specialized pro-resolving mediators and pro-inflammatory lipids (in particular leukotrienes) is strikingly low, providing a molecular explanation for the defective inflammation resolution features of these lesions. In this Review, we discuss the mechanisms of the formation of clinically dangerous atherosclerotic lesions and the potential of pro-resolving mediator therapy to inhibit this process.
Collapse
Affiliation(s)
- Magnus Bäck
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Arif Yurdagul
- Columbia University Irving Medical Center, New York, NY, USA
| | - Ira Tabas
- Columbia University Irving Medical Center, New York, NY, USA
| | - Katariina Öörni
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland.,Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Petri T Kovanen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland.
| |
Collapse
|
213
|
Microglia Actively Remodel Adult Hippocampal Neurogenesis through the Phagocytosis Secretome. J Neurosci 2020; 40:1453-1482. [PMID: 31896673 PMCID: PMC7044727 DOI: 10.1523/jneurosci.0993-19.2019] [Citation(s) in RCA: 222] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/15/2022] Open
Abstract
During adult hippocampal neurogenesis, most newborn cells undergo apoptosis and are rapidly phagocytosed by resident microglia to prevent the spillover of intracellular contents. Here, we propose that phagocytosis is not merely passive corpse removal but has an active role in maintaining neurogenesis. First, we found that neurogenesis was disrupted in male and female mice chronically deficient for two phagocytosis pathways: the purinergic receptor P2Y12, and the tyrosine kinases of the TAM family Mer tyrosine kinase (MerTK)/Axl. In contrast, neurogenesis was transiently increased in mice in which MerTK expression was conditionally downregulated. Next, we performed a transcriptomic analysis of the changes induced by phagocytosis in microglia in vitro and identified genes involved in metabolism, chromatin remodeling, and neurogenesis-related functions. Finally, we discovered that the secretome of phagocytic microglia limits the production of new neurons both in vivo and in vitro Our data suggest that microglia act as a sensor of local cell death, modulating the balance between proliferation and survival in the neurogenic niche through the phagocytosis secretome, thereby supporting the long-term maintenance of adult hippocampal neurogenesis.SIGNIFICANCE STATEMENT Microglia are the brain professional phagocytes and, in the adult hippocampal neurogenic niche, they remove newborn cells naturally undergoing apoptosis. Here we show that phagocytosis of apoptotic cells triggers a coordinated transcriptional program that alters their secretome, limiting neurogenesis both in vivo and in vitro In addition, chronic phagocytosis disruption in mice deficient for receptors P2Y12 and MerTK/Axl reduces adult hippocampal neurogenesis. In contrast, inducible MerTK downregulation transiently increases neurogenesis, suggesting that microglial phagocytosis provides a negative feedback loop that is necessary for the long-term maintenance of adult hippocampal neurogenesis. Therefore, we speculate that the effects of promoting engulfment/degradation of cell debris may go beyond merely removing corpses to actively promoting regeneration in development, aging, and neurodegenerative diseases.
Collapse
|
214
|
Park EJ, Han JS, Seong E, Park EJ, Lee BS, Lee SJ, Lee K. Inhaled Kathon may induce eosinophilia-mediated disease in the lung. ENVIRONMENTAL TOXICOLOGY 2020; 35:27-36. [PMID: 31498972 DOI: 10.1002/tox.22839] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 08/09/2019] [Accepted: 08/09/2019] [Indexed: 06/10/2023]
Abstract
In 2011, a link between humidifier disinfectants and patients with idiopathic pulmonary fibrosis was identified in Korea, and Kathon was suggested as one of the causative agents. In this study, Kathon induced apoptotic cell death along with membrane damage at 24 h post-exposure. Additionally, on day 14 after a single instillation with Kathon, the total number of pulmonary cells and the levels of TNF-α, IL-5, IL-13, MIP-1α, and MCP-1α clearly increased in the lung of mice. The proportion of natural killer cells and eosinophils were significantly elevated in the spleen and the bloodstream, respectively, and the level of immunoglobulin (Ig) A, but not IgG, IgM, and IgE, dose-dependently increased. Therefore, we suggest that inhaled Kathon may induce eosinophilia-mediated disease in the lung by disrupting homeostasis of pulmonary surfactants. Considering that eosinophilia is closely related to cancer and fibrosis, further studies are needed to understand the relationship between them.
Collapse
Affiliation(s)
- Eun-Jung Park
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si, Gyeonggi-do, South Korea
| | - Ji-Seok Han
- Toxicologic Pathology Center, Korea Institute of Toxicology, Daejeon-si, South Korea
| | - Eunsol Seong
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si, Gyeonggi-do, South Korea
| | - Eun-Jun Park
- Graduate School of East-West Medical Science, Kyung Hee University, Yongin-si, Gyeonggi-do, South Korea
| | - Byoung-Seok Lee
- Toxicologic Pathology Center, Korea Institute of Toxicology, Daejeon-si, South Korea
| | - Sang Jin Lee
- Respiratory Disease Research Center, Korea Institute of Toxicology, Jeongeup, Jellobuk-do, South Korea
| | - Kyuhong Lee
- Respiratory Disease Research Center, Korea Institute of Toxicology, Jeongeup, Jellobuk-do, South Korea
| |
Collapse
|
215
|
Han S, Zhuang H, Lee PY, Li M, Yang L, Nigrovic PA, Reeves WH. Differential Responsiveness of Monocyte and Macrophage Subsets to Interferon. Arthritis Rheumatol 2020; 72:100-113. [PMID: 31390156 PMCID: PMC6935410 DOI: 10.1002/art.41072] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/01/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Peripheral blood mononuclear cells (PBMCs) in systemic lupus erythematosus (SLE) patients exhibit a gene expression program (interferon [IFN] signature) that is attributed to overproduction of type I IFNs by plasmacytoid dendritic cells. Type I IFNs have been thought to play a role in the pathogenesis of SLE. This study was undertaken to examine an unexpected influence of monocyte/macrophages on the IFN signature. METHODS Proinflammatory (classic) and antiinflammatory (nonclassic) monocyte/macrophages were sorted from mice and analyzed by RNA sequencing and quantitative polymerase chain reaction (qPCR). Type I IFN-α/β/ω receptor (IFNAR-1) expression was determined by qPCR and flow cytometry. Macrophages were stimulated in vitro with IFNα, and pSTAT1was measured. RESULTS Transcriptional profiling of peritoneal macrophages from mice with pristane-induced SLE unexpectedly indicated a strong IFN signature in classic, but not nonclassic, monocyte/macrophages exposed to the same type I IFN concentrations. Ifnar1 messenger RNA and IFNAR surface staining were higher in classic monocyte/macrophages versus nonclassic monocyte/macrophages (P < 0.0001 and P < 0.05, respectively, by Student's t-test). Nonclassic monocyte/macrophages were also relatively insensitive to IFNα-driven STAT1 phosphorylation. Humans exhibited a similar pattern: higher IFNAR expression (P < 0.0001 by Student's t-test) and IFNα-stimulated gene expression (P < 0.01 by paired Wilcoxon's rank sum test) in classic monocyte/macrophages and lower levels in nonclassic monocyte/macrophages. CONCLUSION This study revealed that the relative abundance of different monocyte/macrophage subsets helps determine the magnitude of the IFN signature. Responsiveness to IFNα signaling reflects differences in IFNAR expression in classic (high IFNAR) compared to nonclassic (low IFNAR) monocyte/macrophages. Thus, the IFN signature depends on both type I IFN production and the responsiveness of monocyte/macrophages to IFNAR signaling.
Collapse
Affiliation(s)
| | | | - Pui Y Lee
- Boston Children's Hospital, Boston, Massachusetts
| | | | | | - Peter A Nigrovic
- Boston Children's Hospital and Brigham and Women's Hospital, Boston, Massachusetts
| | | |
Collapse
|
216
|
Pawaria S, Nündel K, Gao KM, Moses S, Busto P, Holt K, Sharma RB, Brehm MA, Gravallese EM, Socolovsky M, Christ A, Marshak-Rothstein A. Role of Interferon-γ-Producing Th1 Cells in a Murine Model of Type I Interferon-Independent Autoinflammation Resulting From DNase II Deficiency. Arthritis Rheumatol 2019; 72:359-370. [PMID: 31464028 DOI: 10.1002/art.41090] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 08/22/2019] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Patients with hypomorphic mutations in DNase II develop a severe and debilitating autoinflammatory disease. This study was undertaken to compare the disease parameters in these patients to those in a murine model of DNase II deficiency, and to evaluate the role of specific nucleic acid sensors and identify the cell types responsible for driving the autoinflammatory response. METHODS To avoid embryonic death, Dnase2-/- mice were intercrossed with mice that lacked the type I interferon (IFN) receptor (Ifnar-/- ). The hematologic changes and immune status of these mice were evaluated using complete blood cell counts, flow cytometry, serum cytokine enzyme-linked immunosorbent assays, and liver histology. Effector cell activity was determined by transferring T cells from Dnase2-/- × Ifnar-/- double-knockout (DKO) mice into Rag1-/- mice, and 4 weeks after cell transfer, induced changes were assessed in the recipient mice. RESULTS In Dnase2-/- × Ifnar-/- DKO mice, many of the disease features found in DNase II-deficient patients were recapitulated, including cytopenia, extramedullary hematopoiesis, and liver fibrosis. Dnase2+/+ × Rag1-/- mice (n > 22) developed a hematologic disorder that was attributed to the transfer of an unusual IFNγ-producing T cell subset from the spleens of donor Dnase2-/- × Ifnar-/- DKO mice. Autoinflammation in this murine model did not depend on the stimulator of IFN genes (STING) pathway but was highly dependent on the chaperone protein Unc93B1. CONCLUSION Dnase2-/- × Ifnar-/- DKO mice may be a valid model for exploring the innate and adaptive immune mechanisms responsible for the autoinflammation similar to that seen in DNASE2-hypomorphic patients. In this murine model, IFNγ is required for T cell activation and the development of clinical manifestations. The role of IFNγ in DNASE2-deficient patient populations remains to be determined, but the ability of Dnase2-/- mouse T cells to transfer disease to Rag1-/- mice suggests that T cells may be a relevant therapeutic target in patients with IFN-related systemic autoinflammatory diseases.
Collapse
Affiliation(s)
| | | | - Kevin M Gao
- University of Massachusetts Medical School, Worcester
| | | | | | - Kevin Holt
- University of Massachusetts Medical School, Worcester
| | | | | | | | | | | | | |
Collapse
|
217
|
Gastrodin Ameliorates Acute Rejection via IRE1 α/TRAF2/NF- κB in Rats Receiving Liver Allografts. BIOMED RESEARCH INTERNATIONAL 2019; 2019:9276831. [PMID: 31828147 PMCID: PMC6886336 DOI: 10.1155/2019/9276831] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/05/2019] [Accepted: 09/16/2019] [Indexed: 12/16/2022]
Abstract
Background Liver transplantation (LT) is currently an effective treatment for end-stage liver disease, but the occurrence of acute rejection (AR) is still the main problem to be solved. The present study aimed to evaluate the effect of gastrodin (GAS) on LT. Methods Rat transplant models were established and divided into SHAM, LT, GAS-L (50 mg/kg GAS), and GAS-H (100 mg/kg GAS) groups. The liver function, inflammatory factors, liver histopathology, survival of rats, number of M2-type macrophages, liver cell apoptosis, and pathway proteins were assayed at 7 days and 14 days after the operations. Results With increasing GAS concentrations, liver function, expression of proinflammatory factors in the liver, and expression of M2-type molecules in macrophages were significantly improved, and the survival time of rats was significantly prolonged (P < 0.05). All rats treated with low or high doses of GAS were judged to have nondeterministic acute rejection. Flow cytometry showed that liver cell apoptosis was decreased significantly in the GAS-L and GAS-H groups after GAS administration compared with apoptosis and differentiation in the LT group (P < 0.05). Expression levels of Caspase-3, Bad, and Bax proteins were decreased, and the expression of the antiapoptotic protein Bcl-2 was increased in the GAS-L and GAS-H groups (P < 0.05). Mechanistically, the ERS-related IRE1α/TRAF2/NF-κB pathway was suppressed by GAS, and GAS acted mainly on intrahepatic macrophages to affect AR and reduce ROS production (P < 0.05). Conclusion GAS ameliorated AR by inhibiting the IRE1α/TRAF2/NF-κB pathway in LT.
Collapse
|
218
|
Fast cyclical-decellularized trachea as a natural 3D scaffold for organ engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 105:110142. [DOI: 10.1016/j.msec.2019.110142] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 08/15/2019] [Accepted: 08/26/2019] [Indexed: 12/16/2022]
|
219
|
Kim GT, Hahn KW, Sohn K, Yoon SY, Kim JW. PLAG enhances macrophage mobility for efferocytosis of apoptotic neutrophils via membrane redistribution of P2Y2. FEBS J 2019; 286:5016-5029. [DOI: 10.1111/febs.15135] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 08/15/2019] [Accepted: 11/10/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Guen Tae Kim
- Cell Factory Research Center Division of Systems Biology and Bioengineering Korea Research Institute of Bioscience and Biotechnology Daejeon South Korea
- Department of Biological Sciences College of Life Science and Nano Technology Hannam University Daejeon South Korea
| | - Kyu Woong Hahn
- Department of Biological Sciences College of Life Science and Nano Technology Hannam University Daejeon South Korea
| | - Ki‐Young Sohn
- Division of Global New Drug Development ENZYCHEM Lifesciences Jecheon South Korea
| | - Sun Young Yoon
- Division of Global New Drug Development ENZYCHEM Lifesciences Jecheon South Korea
| | - Jae Wha Kim
- Cell Factory Research Center Division of Systems Biology and Bioengineering Korea Research Institute of Bioscience and Biotechnology Daejeon South Korea
| |
Collapse
|
220
|
Bollinger AL, Bollinger T, Rupp J, Shima K, Gross N, Padayachy L, Chicheportiche R, Puga Yung GL, Seebach JD. Annexin V expression on CD4 + T cells with regulatory function. Immunology 2019; 159:205-220. [PMID: 31642515 DOI: 10.1111/imm.13140] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 09/28/2019] [Accepted: 10/04/2019] [Indexed: 12/22/2022] Open
Abstract
Regulatory T (Treg) cells induce immunologic tolerance by suppressing effector functions of conventional lymphocytes in the periphery. On the other hand, immune silencing is mediated by recognition of phosphatidylserine (PS) on apoptotic cells by phagocytes. Here we describe expression of the PS-binding protein Annexin V (ANXA5) in CD4+ CD25hi Treg cells at the mRNA and protein levels. CD4+ ANXA5+ T cells constitute about 0·1%-0·6% of peripheral blood CD3+ T cells, exhibit co-expression of several Treg markers, such as Forkhead box P3, programmed cell death protein-1, cytotoxic T-lymphocyte antigen-4 and CD38. In vitro, ANXA5+ Treg cells showed enhanced adhesion to PS+ endothelial cells. Stimulated by anti-CD3 and PS+ syngeneic antigen-presenting cells CD4+ ANXA5+ T cells expanded in the absence of exogenous interleukin-2. CD4+ ANXA5+ T cells suppressed CD4+ ANXA5- T-cell proliferation and mammalian target of rapamycin phosphorylation, partially dependent on cell contact. CD4+ ANXA5+ T-cell-mediated suppression was allo-specific and accompanied by an increased production of anti-inflammatory mediators. In vivo, using a model of delayed type hypersensitivity, murine CD4+ ANXA5+ T cells inhibited T helper type 1 responses. In conclusion, we report for the first time expression of ANXA5 on a subset of Treg cells that might bridge classical regulatory Treg function with immune silencing.
Collapse
Affiliation(s)
- Anna-Lena Bollinger
- Division of Immunology and Allergology, Department of Medicine, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Thomas Bollinger
- Department of Molecular Biology, University of Geneva, Geneva, Switzerland
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Kensuke Shima
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Natalie Gross
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Laura Padayachy
- Division of Immunology and Allergology, Department of Medicine, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Rachel Chicheportiche
- Division of Immunology and Allergology, Department of Medicine, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Gisella L Puga Yung
- Division of Immunology and Allergology, Department of Medicine, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Jörg Dieter Seebach
- Division of Immunology and Allergology, Department of Medicine, University Hospitals and Medical Faculty, Geneva, Switzerland
| |
Collapse
|
221
|
Novel indol-3-yl-thiosemicarbazone derivatives: Obtaining, evaluation of in vitro leishmanicidal activity and ultrastructural studies. Chem Biol Interact 2019; 315:108899. [PMID: 31738906 DOI: 10.1016/j.cbi.2019.108899] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 11/12/2019] [Indexed: 12/19/2022]
Abstract
Parasitic diseases still represent serious public health problems, since the high and steady emergence of resistant strains is evident. Because parasitic infections are distributed predominantly in developing countries, less toxic, more efficient, safer and more accessible drugs have become desirable in the treatment of the infected population. This is the case of leishmaniasis, an infectious disease caused by a protozoan of the genus Leishmania sp., responsible for triggering pathological processes from the simplest to the most severe forms leading to high rates of morbidity and mortality throughout the world. In the search for new leishmanicidal drugs, the thiosemicarbazones and the indole fragments have been identified as promising structures for leishmanicidal activity. The present study proposes the synthesis and structural characterization of new indole-thiosemicarbazone derivatives (2a-j), in addition to performing in vitro evaluations through cytotoxicity assays using macrophages (J774) activity against forms of Leishmania infantum and Leishmania amazonensis promastigote as well as ultrastructural analyzes in promastigotes of L. infantum. Results show that the indole-thiosemicarbazone derivatives were obtained with yield values varying from 32.09 to 94.64%. In the evaluation of cytotoxicity, the indole-thiosemicarbazone compounds presented CC50 values between 53.23 and 357.97 μM. Concerning the evaluation against L. amazonensis promastigote forms, IC50 values ranged between 12.31 and > 481.52 μM, while the activity against L. infantum promastigotes obtained IC50 values between 4.36 and 23.35 μM. The compounds 2d and 2i tested against L. infantum were the most promising in the series, as they showed the lowest IC50 values: 5.60 and 4.36 respectively. The parasites treated with the compounds 2d and 2i showed several structural alterations, such as shrinkage of the cell body, shortening and loss of the flagellum, intense mitochondrial swelling and vacuolization of the cytoplasm leading the parasite to cellular unviability. Therefore, the indole-thiosemicarbazone compounds are promising because they yield considerable synthesis, have low cytotoxicity to mammalian cells and act as leishmanicidal agents.
Collapse
|
222
|
Naik A, Griffin M, Szarko M, Butler PE. Optimizing the decellularization process of an upper limb skeletal muscle; implications for muscle tissue engineering. Artif Organs 2019; 44:178-183. [PMID: 31571221 DOI: 10.1111/aor.13575] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 08/05/2019] [Accepted: 09/24/2019] [Indexed: 12/27/2022]
Abstract
Upper limb muscle reconstruction is required following cancer resection, trauma, and congenital deformities. Current surgical reconstruction of the muscle involves local, regional and free flaps. However, muscle reconstruction is not always possible due to the size of the defect and functional donor site morbidity. These challenges could be addressed with the production of scaffolds composed of an extracellular matrix (ECM) derived from decellularized human skeletal muscle. This study aimed to find an optimal technique to decellularize a flexor digitorum superficialis muscle. The first two protocols were based on a detergent only (DOT) and a detergent-enzymatic protocol (DET). The third protocol avoided the use of detergents and proteolytic enzymes (NDNET). The decellularized scaffolds were characterized using qualitative techniques including histological and immunofluorescent staining and quantitative techniques assessing deoxyribonucleic acid (DNA), glycosaminoglycan (GAG), and collagen content. The DOT protocol consisting of 2% SDS for 4 hours was successful at decellularizing human FDS, as shown by DNA content assay and nuclei immunofluorescence staining. The DOT protocol maintained the microstructure of the scaffolds as shown by Masson's trichrome staining and collagen and GAG content. DET and NDNET protocols maintained the ECM, but were unsuccessful in removing all DNA content after two cycles of decellularization. Decellularization of skeletal muscle is a viable option for muscle reconstruction using a detergent only technique for upper limb defects. Further testing in vivo will assess the effectiveness of decellularized scaffolds for upper limb muscle skeletal tissue engineering.
Collapse
Affiliation(s)
- Anish Naik
- Division of Surgery, University College London, London, United Kingdom.,Charles Wolfson Center for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom.,Department of Plastic Surgery, Royal Free Hospital, London, United Kingdom
| | - Michelle Griffin
- Division of Surgery, University College London, London, United Kingdom.,Charles Wolfson Center for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom.,Department of Plastic Surgery, Royal Free Hospital, London, United Kingdom
| | - Matthew Szarko
- Division of Surgery, University College London, London, United Kingdom.,Charles Wolfson Center for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom.,Department of Plastic Surgery, Royal Free Hospital, London, United Kingdom
| | - Peter E Butler
- Division of Surgery, University College London, London, United Kingdom.,Charles Wolfson Center for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom.,Department of Plastic Surgery, Royal Free Hospital, London, United Kingdom
| |
Collapse
|
223
|
Friedrich SK, Lang PA, Friebus-Kardash J, Duhan V, Bezgovsek J, Lang KS. Mechanisms of lymphatic system-specific viral replication and its potential role in autoimmune disease. Clin Exp Immunol 2019; 195:64-73. [PMID: 30444956 DOI: 10.1111/cei.13241] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2018] [Indexed: 12/15/2022] Open
Abstract
Viral infections can be fatal because of the direct cytopathic effects of the virus or the induction of a strong, uncontrolled inflammatory response. Virus and host intrinsic characteristics strongly modulate the outcome of viral infections. Recently we determined the circumstances under which enhanced replication of virus within the lymphoid tissue is beneficial for the outcome of a disease. This enforced viral replication promotes anti-viral immune activation and, counterintuitively, accelerates virus control. In this review we summarize the mechanisms that contribute to enforced viral replication. Antigen-presenting cells and CD169+ macrophages exhibit enforced viral replication after infection with the model viruses lymphocytic choriomeningitis virus (LCMV) and vesicular stomatitis virus (VSV). Ubiquitin-specific peptidase 18 (Usp18), an endogenous type I interferon blocker in CD169+ macrophages, has been identified as a proviral gene, as are B cell activating factor (BAFF) and carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1). Lymphotoxins (LT) strongly enhance viral replication in the spleen and lymph nodes. All these factors modulate splenic architecture and thereby promote the development of CD169+ macrophages. Tumor necrosis factor alpha (TNF-α) and nuclear factor kappa-light-chain-enhancer of activated B cell signaling (NF-κB) have been found to promote the survival of infected CD169+ macrophages, thereby similarly promoting enforced viral replication. Association of autoimmune disease with infections is evident from (1) autoimmune phenomena described during a chronic virus infection; (2) onset of autoimmune disease simultaneous to viral infections; and (3) experimental evidence. Involvement of virus infection during onset of type I diabetes is strongly evident. Epstein-Bar virus (EBV) infection was discussed to be involved in the pathogenesis of systemic lupus erythematosus. In conclusion, several mechanisms promote viral replication in secondary lymphatic organs. Identifying such factors in humans is a challenge for future studies.
Collapse
Affiliation(s)
- S-K Friedrich
- University of Duisburg-Essen, Institute of Immunology, Medical Faculty, Essen, Germany
| | - P A Lang
- Heinrich-Heine-University, Insitute of Molecular Medicine II, Düsseldorf, Germany
| | - J Friebus-Kardash
- University of Duisburg-Essen, Institute of Immunology, Medical Faculty, Essen, Germany
| | - V Duhan
- University of Duisburg-Essen, Institute of Immunology, Medical Faculty, Essen, Germany
| | - J Bezgovsek
- University of Duisburg-Essen, Institute of Immunology, Medical Faculty, Essen, Germany
| | - K S Lang
- University of Duisburg-Essen, Institute of Immunology, Medical Faculty, Essen, Germany
| |
Collapse
|
224
|
Chua BA, Ngo JA, Situ K, Morizono K. Roles of phosphatidylserine exposed on the viral envelope and cell membrane in HIV-1 replication. Cell Commun Signal 2019; 17:132. [PMID: 31638994 PMCID: PMC6805584 DOI: 10.1186/s12964-019-0452-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 10/04/2019] [Indexed: 02/07/2023] Open
Abstract
Phosphatidylserine (PtdSer) is usually present only in the inner leaf of the lipid bilayers of the cell membrane, but is exposed on the outer leaf when cells are activated and/or die. Exposure of PtdSer has physiological functions. For example, the PtdSer exposed on dead cells can serve as “eat-me signals” for phagocytes to clear dead cells by phagocytosis, which prevents autoimmune reactions and inflammation. HIV-1 induces PtdSer exposure on infected and target cells and it also exposes PtdSer on its envelope. Recent studies showed that PtdSer exposed on the HIV-1 envelope and infected and target cells can facilitate or inhibit multiple steps of HIV-1 replication. At the virus binding and entry steps, interaction of the envelope PtdSer and the host’s PtdSer-binding molecules can enhance HIV-1 infection of cells by facilitating virus attachment. At the virus budding step, HIV-1 can be trapped on the cell surface by one family of PtdSer-binding receptors, T-cell immunoglobulin mucin domain proteins (TIM)-1, 3, and 4 expressed on virus producer cells. Although this trapping can inhibit release of HIV-1, one of the HIV-1 accessory gene products, Negative Factor (Nef), can counteract virus trapping by TIM family receptors (TIMs) by inducing the internalization of these receptors. HIV-1 infection can induce exposure of PtdSer on infected cells by inducing cell death. A soluble PtdSer-binding protein in serum, protein S, bridges PtdSer exposed on HIV-1-infected cells and a receptor tyrosine kinase, Mer, expressed on macrophages and mediate phagocytic clearance of HIV-1 infected cells. HIV-1 can also induce exposure of PtdSer on target cells at the virus binding step. Binding of HIV-1 envelope proteins to its receptor (CD4) and co-receptors (CXCR4 or CCR5) elicit signals that induce PtdSer exposure on target cells by activating TMEM16F, a phospholipid scramblase. PtdSer exposed on target cells enhances HIV-1 infection by facilitating fusion between the viral envelope and target cell membrane. Because various other phospholipid channels mediating PtdSer exposure have recently been identified, it will be of interest to examine how HIV-1 actively interacts with these molecules to manipulate PtdSer exposure levels on cells and viral envelope to support its replication.
Collapse
|
225
|
Canuto J, Lima D, Menezes R, Batista A, Nogueira P, Silveira E, Grangeiro T, Nogueira N, Martins A. Antichagasic effect of violacein from
Chromobacterium violaceum. J Appl Microbiol 2019; 127:1373-1380. [DOI: 10.1111/jam.14391] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/03/2019] [Accepted: 07/11/2019] [Indexed: 12/26/2022]
Affiliation(s)
- J.A. Canuto
- Departamento de Análises Clínicas e Toxicológicas Universidade Federal do Ceará Fortaleza CE Brazil
| | - D.B. Lima
- Departamento de Análises Clínicas e Toxicológicas Universidade Federal do Ceará Fortaleza CE Brazil
| | - R.R.P.P.B. Menezes
- Departamento de Análises Clínicas e Toxicológicas Universidade Federal do Ceará Fortaleza CE Brazil
| | - A.H.M. Batista
- Departamento de Análises Clínicas e Toxicológicas Universidade Federal do Ceará Fortaleza CE Brazil
| | - P.C.D.N. Nogueira
- Departamento de Química Universidade Federal do Ceará Fortaleza CE Brazil
| | - E.R. Silveira
- Departamento de Química Universidade Federal do Ceará Fortaleza CE Brazil
| | - T.B. Grangeiro
- Departamento de Biologia Universidade Federal do Ceará Fortaleza CE Brazil
| | - N.A.P. Nogueira
- Departamento de Análises Clínicas e Toxicológicas Universidade Federal do Ceará Fortaleza CE Brazil
| | - A.M.C. Martins
- Departamento de Análises Clínicas e Toxicológicas Universidade Federal do Ceará Fortaleza CE Brazil
| |
Collapse
|
226
|
de Couto G, Jaghatspanyan E, DeBerge M, Liu W, Luther K, Wang Y, Tang J, Thorp EB, Marbán E. Mechanism of Enhanced MerTK-Dependent Macrophage Efferocytosis by Extracellular Vesicles. Arterioscler Thromb Vasc Biol 2019; 39:2082-2096. [PMID: 31434491 DOI: 10.1161/atvbaha.119.313115] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Extracellular vesicles secreted by cardiosphere-derived cells (CDCev) polarize macrophages toward a distinctive phenotype with enhanced phagocytic capacity (MCDCev). These changes underlie cardioprotection by CDCev and by the parent CDCs, notably attenuating the no-reflow phenomenon following myocardial infarction, but the mechanisms are unclear. Here, we tested the hypothesis that MCDCev are especially effective at scavenging debris from dying cells (ie, efferocytosis) to attenuate irreversible damage post-myocardial infarction. Approach and Results: In vitro efferocytosis assays with bone marrow-derived macrophages, and in vivo transgenic rodent models of myocardial infarction, demonstrate enhanced apoptotic cell clearance with MCDCev. CDCev exposure induces sustained MerTK expression in MCDCev through extracellular vesicle transfer of microRNA-26a (via suppression of Adam17); the cardioprotective response is lost in animals deficient in MerTK. Single-cell RNA-sequencing revealed phagocytic pathway activation in MCDCev, with increased expression of complement factor C1qa, a phagocytosis facilitator. CONCLUSIONS Together, these data demonstrate that extracellular vesicle modulation of MerTK and C1qa expression leads to enhanced macrophage efferocytosis and cardioprotection.
Collapse
Affiliation(s)
- Geoffrey de Couto
- From the Smidt Heart Institute (G.d.C., E.J., W.L., K.L., E.M.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Ervin Jaghatspanyan
- From the Smidt Heart Institute (G.d.C., E.J., W.L., K.L., E.M.), Cedars-Sinai Medical Center, Los Angeles, CA
| | | | - Weixin Liu
- From the Smidt Heart Institute (G.d.C., E.J., W.L., K.L., E.M.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Kristin Luther
- From the Smidt Heart Institute (G.d.C., E.J., W.L., K.L., E.M.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Yizhou Wang
- Genomics Core (Y.W., J.T.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Jie Tang
- Genomics Core (Y.W., J.T.), Cedars-Sinai Medical Center, Los Angeles, CA
| | | | - Eduardo Marbán
- From the Smidt Heart Institute (G.d.C., E.J., W.L., K.L., E.M.), Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
227
|
Stabilin Receptors: Role as Phosphatidylserine Receptors. Biomolecules 2019; 9:biom9080387. [PMID: 31434355 PMCID: PMC6723754 DOI: 10.3390/biom9080387] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 08/16/2019] [Accepted: 08/18/2019] [Indexed: 12/18/2022] Open
Abstract
Phosphatidylserine is a membrane phospholipid that is localized to the inner leaflet of the plasma membrane. Phosphatidylserine externalization to the outer leaflet of the plasma membrane is an important signal for various physiological processes, including apoptosis, platelet activation, cell fusion, lymphocyte activation, and regenerative axonal fusion. Stabilin-1 and stabilin-2 are membrane receptors that recognize phosphatidylserine on the cell surface. Here, we discuss the functions of Stabilin-1 and stabilin-2 as phosphatidylserine receptors in apoptotic cell clearance (efferocytosis) and cell fusion, and their ligand-recognition and signaling pathways.
Collapse
|
228
|
Efficient engulfment of necroptotic and pyroptotic cells by nonprofessional and professional phagocytes. Cell Discov 2019; 5:39. [PMID: 31632688 PMCID: PMC6796833 DOI: 10.1038/s41421-019-0108-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 04/18/2019] [Accepted: 06/05/2019] [Indexed: 12/20/2022] Open
|
229
|
Nonaka S, Sono M, Hoshi C, Kanetani T, Nakayama H, Dohmae N, Nakanishi Y. Transcription repressor-mediated control of engulfment receptor expression in Drosophila phagocytes. Exp Cell Res 2019; 381:10-17. [DOI: 10.1016/j.yexcr.2019.04.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/03/2019] [Accepted: 04/29/2019] [Indexed: 01/04/2023]
|
230
|
Gorgulho CM, Romagnoli GG, Bharthi R, Lotze MT. Johnny on the Spot-Chronic Inflammation Is Driven by HMGB1. Front Immunol 2019; 10:1561. [PMID: 31379812 PMCID: PMC6660267 DOI: 10.3389/fimmu.2019.01561] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 06/24/2019] [Indexed: 12/24/2022] Open
Abstract
Although much has been made of the role of HMGB1 acting as an acute damage associated molecular pattern (DAMP) molecule, prompting the response to tissue damage or injury, it is also released at sites of chronic inflammation including sites of infection, autoimmunity, and cancer. As such, the biology is distinguished from homeostasis and acute inflammation by the recruitment and persistence of myeloid derived suppressor cells, T regulatory cells, fibrosis and/or exuberant angiogenesis depending on the antecedents and the other individual inflammatory partners that HMGB1 binds and focuses, including IL-1β, CXCL12/SDF1, LPS, DNA, RNA, and sRAGE. High levels of HMGB1 released into the extracellular milieu and its persistence in the microenvironment can contribute to the pathogenesis of many if not all autoimmune disorders and is a key factor that drives inflammation further and worsens symptoms. HMGB1 is also pivotal in the maintenance of chronic inflammation and a “wound healing” type of immune response that ultimately contributes to the onset of carcinogenesis and tumor progression. Exosomes carrying HMGB1 and other instructive molecules are released and shape the response of various cells in the chronic inflammatory environment. Understanding the defining roles of REDOX, DAMPs and PAMPs, and the host response in chronic inflammation requires an alternative means for positing HMGB1's central role in limiting and focusing inflammation, distinguishing chronic from acute inflammation.
Collapse
Affiliation(s)
- Carolina M Gorgulho
- Tumor Immunology Laboratory, Department of Microbiology and Immunology, Botucatu Institute of Biosciences, São Paulo State University, Botucatu, Brazil.,DAMP Laboratory, Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Graziela G Romagnoli
- Tumor Immunology Laboratory, Department of Microbiology and Immunology, Botucatu Institute of Biosciences, São Paulo State University, Botucatu, Brazil
| | - Rosh Bharthi
- DAMP Laboratory, Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael T Lotze
- DAMP Laboratory, Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Immunology, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
231
|
Riis S, Hansen AC, Johansen L, Lund K, Pedersen C, Pitsa A, Hyldig K, Zachar V, Fink T, Pennisi CP. Fabrication and characterization of extracellular matrix scaffolds obtained from adipose-derived stem cells. Methods 2019; 171:68-76. [PMID: 31299290 DOI: 10.1016/j.ymeth.2019.07.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/17/2019] [Accepted: 07/06/2019] [Indexed: 02/08/2023] Open
Abstract
Chronic non-healing wounds are detrimental for the quality of life of the affected individuals and represent a major burden for the health care systems. Adipose-derived stem cells (ASCs) are being investigated for the development of novel treatments of chronic wounds, as they have shown several positive effects on wound healing. While these effects appear to be mediated by the release of soluble factors, it is has also become apparent that the extracellular matrix (ECM) deposited by ASCs is essential in several phases of the wound healing process. In this work, we describe an approach to produce ECM scaffolds derived from ASCs in culture. Upon growth of ASCs into an overconfluent cell layer, a detergent-based cell extraction approach is applied to remove the cellular components. The extraction is followed by an enzymatic treatment to remove the residual DNA. The resultant cell-derived scaffolds are depleted of cellular components, display low DNA remnant, and retain the native fibrillar organization of the ECM. Analysis of the molecular composition of the ECM scaffolds revealed that they are composed of collagens type I and III, and fibronectin. The decellularized scaffolds represent a substrate that supports adhesion and proliferation of primary human fibroblasts and dermal microvascular endothelial cells, indicating their potential as platforms for wound healing studies.
Collapse
Affiliation(s)
- Simone Riis
- Laboratory for Stem Cell Research, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Anne Cathrine Hansen
- Laboratory for Stem Cell Research, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Lonnie Johansen
- Laboratory for Stem Cell Research, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Kaya Lund
- Laboratory for Stem Cell Research, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Cecilie Pedersen
- Laboratory for Stem Cell Research, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Aikaterini Pitsa
- Laboratory for Stem Cell Research, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Kathrine Hyldig
- Laboratory for Stem Cell Research, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Vladimir Zachar
- Laboratory for Stem Cell Research, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Trine Fink
- Laboratory for Stem Cell Research, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Cristian Pablo Pennisi
- Laboratory for Stem Cell Research, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
232
|
Simon D, Gilicze O, Farkas N, Najbauer J, Németh P, Lénárd L, Berki T. Correlation of natural autoantibodies and cardiovascular disease-related anti-bacterial antibodies in pericardial fluid of cardiac surgery patients. Clin Exp Immunol 2019; 193:55-63. [PMID: 29573404 DOI: 10.1111/cei.13127] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2018] [Indexed: 12/30/2022] Open
Abstract
Our previous studies showed that anti-citrate synthase (anti-CS) immunoglobulin (Ig)M natural autoantibodies are present in healthy individuals without previous antigen stimulation, but no studies have investigated their presence in the pericardial fluid (PF). Therefore, we detected the natural anti-CS IgG/M autoantibody levels in plasma and PF of cardiac surgery patients and investigated their relationship with cardiovascular disease-associated bacterial pathogens. PF and blood samples of 22 coronary artery bypass graft (CABG) and 10 aortic valve replacement (AVR) patients were tested for total Ig levels, natural autoantibodies and infection-related antibodies using enzyme-linked immunosorbent assay (ELISA) and Luminex methods. The B cell subsets were measured by flow cytometry. The total Ig subclass levels were four to eight times lower in PF than in plasma, but the natural anti-CS IgM autoantibodies showed a relative increase in PF. The frequency of CD19+ B lymphocytes was significantly lower in PF than in blood (P = 0·01), with a significant relative increase of B1 cells (P = 0·005). Mycoplasma pneumoniae antibody-positive patients had significantly higher anti-CS IgM levels. In CABG patients we found a correlation between anti-CS IgG levels and M. pneumoniae, Chlamydia pneumoniae and Borrelia burgdorferi antibody titres. Our results provide the first evidence that natural autoantibodies are present in the PF, and they show a significant correlation with certain anti-bacterial antibody titres in a disease-specific manner.
Collapse
Affiliation(s)
- D Simon
- Department of Immunology and Biotechnology, Clinical Center, Pécs, Hungary
| | - O Gilicze
- Department of Immunology and Biotechnology, Clinical Center, Pécs, Hungary.,Heart Institute, University of Pécs Medical School, Pécs, Hungary
| | - N Farkas
- Institute of Bioanalysis, University of Pécs Medical School, Pécs, Hungary
| | - J Najbauer
- Department of Immunology and Biotechnology, Clinical Center, Pécs, Hungary
| | - P Németh
- Department of Immunology and Biotechnology, Clinical Center, Pécs, Hungary
| | - L Lénárd
- Heart Institute, University of Pécs Medical School, Pécs, Hungary
| | - T Berki
- Department of Immunology and Biotechnology, Clinical Center, Pécs, Hungary
| |
Collapse
|
233
|
Wang N, Chen M, Wang T. Liposomes used as a vaccine adjuvant-delivery system: From basics to clinical immunization. J Control Release 2019; 303:130-150. [PMID: 31022431 PMCID: PMC7111479 DOI: 10.1016/j.jconrel.2019.04.025] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/17/2019] [Accepted: 04/17/2019] [Indexed: 12/14/2022]
Abstract
Liposomes are widely utilized as a carrier to improve therapeutic efficacy of agents thanks to their merits of high loading capacity, targeting delivery, reliable protection of agents, good biocompatibility, versatile structure modification and adjustable characteristics, such as size, surface charge, membrane flexibility and the agent loading mode. In particular, in recent years, through modification with immunopotentiators and targeting molecules, and in combination with innovative immunization devices, liposomes are rapidly developed as a multifunctional vaccine adjuvant-delivery system (VADS) that has a high capability in inducing desired immunoresponses, as they can target immune cells and even cellular organelles, engender lysosome escape, and promote Ag cross-presentation, thus enormously enhancing vaccination efficacy. Moreover, after decades of development, several products developed on liposome VADS have already been authorized for clinical immunization and are showing great advantages over conventional vaccines. This article describes in depth some critical issues relevant to the development of liposomes as a VADS, including principles underlying immunization, physicochemical properties of liposomes as the immunity-influencing factors, functional material modification to enhance immunostimulatory functions, the state-of-the-art liposome VADSs, as well as the marketed vaccines based on a liposome VADS. Therefore, this article provides a comprehensive reference to the development of novel liposome vaccines.
Collapse
Affiliation(s)
- Ning Wang
- School of Food and Bioengineering, Hefei University of Technology, 193 Tun Brook Road, Hefei, Anhui Province 230009, China
| | - Minnan Chen
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Ting Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China.
| |
Collapse
|
234
|
Ma Q, Liang M, Wu Y, Ding N, Duan L, Yu T, Bai Y, Kang F, Dong S, Xu J, Dou C. Mature osteoclast-derived apoptotic bodies promote osteogenic differentiation via RANKL-mediated reverse signaling. J Biol Chem 2019; 294:11240-11247. [PMID: 31167789 DOI: 10.1074/jbc.ra119.007625] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/21/2019] [Indexed: 11/06/2022] Open
Abstract
In bone remodeling, after a lifespan of ∼2 weeks, osteoclasts undergo apoptosis in each bone turnover cycle, resulting in generation of a large number of apoptotic bodies (ABs). However, the biological roles of osteoclast-derived ABs (OC-ABs) in bone remodeling have not been investigated and remain unknown. In this study, we stimulated bone marrow macrophages with receptor activator of NF-κB ligand (RANKL) to obtain both preosteoclasts and mature osteoclasts (mOCs). We then used alendronate to induce apoptosis in preosteoclasts and mOCs and generate the respective ABs and used flow cytometry and immunoblotting to characterize the sizes and immunogenic characteristics of the extracted ABs. We show that mOC-ABs are engulfed by preosteoblastic MC3T3-E1 cells and promote the viability of these cells. Among all osteoclast-derived extracellular vesicles, mOC-ABs had the highest osteogenic potency. We further observed that mOC-ABs had the highest vesicular receptor activator of NF-κB (RANK) levels among all types of osteoclast-derived extracellular vesicles. Of note, masking of vesicular RANK by soluble RANKL strongly abolished the osteogenic potency of osteoclast-derived ABs. Mechanistically, we found that mOC-ABs induce osteoblast differentiation by activatingPI3K/AKT/mechanistic target of rapamycin (mTOR)/ribosomal protein S6 kinase signaling. In conclusion, OC-ABs promote osteogenic differentiation by stimulating osteoblast differentiation via activation of RANKL reverse signaling. These findings provide important insights into the reversal phase between the bone resorption and formation stages during bone remodeling and identify an AB-dependent cellular signaling mechanism in osteoclast-osteoblast coupling.
Collapse
Affiliation(s)
- Qinyu Ma
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Mengmeng Liang
- Department of Biomedical Materials Science, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yutong Wu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ning Ding
- Department of Blood Purification, General Hospital of Shenyang Military Area Command, Shenyang 110000, China
| | - Lianli Duan
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Tao Yu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yun Bai
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Fei Kang
- Department of Biomedical Materials Science, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jianzhong Xu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ce Dou
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China .,Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
235
|
Bahrini I, Hanayama R. Development of a Method That Delivers Drugs to Enveloped Viruses. Biol Pharm Bull 2019; 42:977-981. [DOI: 10.1248/bpb.b18-01000] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Insaf Bahrini
- Department of Immunology, Kanazawa University Graduate School of Medical Sciences
| | - Rikinari Hanayama
- Department of Immunology, Kanazawa University Graduate School of Medical Sciences
- WPI Nano Life Science Institute (NanoLSI), Kanazawa University
| |
Collapse
|
236
|
Fernández-Pérez J, Ahearne M. Decellularization and recellularization of cornea: Progress towards a donor alternative. Methods 2019; 171:86-96. [PMID: 31128238 DOI: 10.1016/j.ymeth.2019.05.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 12/20/2022] Open
Abstract
The global shortage of donor corneas for transplantation has led to corneal bioengineering being investigated as a method to generate transplantable tissues. Decellularized corneas are among the most promising materials for engineering corneal tissue since they replicate the complex structure and composition of real corneas. Decellularization is a process that aims to remove cells from organs or tissues resulting in a cell-free scaffold consisting of the tissues extracellular matrix. Here different decellularization techniques are described, including physical, chemical and biological methods. Analytical techniques to confirm decellularization efficiency are also discussed. Different cell sources for the recellularization of the three layers of the cornea, recellularization methods used in the literature and techniques used to assess the outcome of the implantation of such scaffolds are examined. Studies involving the application of decellularized corneas in animal models and human clinical studies are discussed. Finally, challenges for this technology are explored involving scalability, automatization and regulatory affairs.
Collapse
Affiliation(s)
- Julia Fernández-Pérez
- Dept of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, University of Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Science Institute, Trinity College Dublin, University of Dublin, Ireland
| | - Mark Ahearne
- Dept of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, University of Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Science Institute, Trinity College Dublin, University of Dublin, Ireland.
| |
Collapse
|
237
|
Guo M, Härtlova A, Gierliński M, Prescott A, Castellvi J, Losa JH, Petersen SK, Wenzel UA, Dill BD, Emmerich CH, Ramon Y Cajal S, Russell DG, Trost M. Triggering MSR1 promotes JNK-mediated inflammation in IL-4-activated macrophages. EMBO J 2019; 38:embj.2018100299. [PMID: 31028084 PMCID: PMC6545745 DOI: 10.15252/embj.2018100299] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/14/2022] Open
Abstract
Alternatively activated M2 macrophages play an important role in maintenance of tissue homeostasis by scavenging dead cells, cell debris and lipoprotein aggregates via phagocytosis. Using proteomics, we investigated how alternative activation, driven by IL‐4, modulated the phagosomal proteome to control macrophage function. Our data indicate that alternative activation enhances homeostatic functions such as proteolysis, lipolysis and nutrient transport. Intriguingly, we identified the enhanced recruitment of the TAK1/MKK7/JNK signalling complex to phagosomes of IL‐4‐activated macrophages. The recruitment of this signalling complex was mediated through K63 polyubiquitylation of the macrophage scavenger receptor 1 (MSR1). Triggering of MSR1 in IL‐4‐activated macrophages leads to enhanced JNK activation, thereby promoting a phenotypic switch from an anti‐inflammatory to a pro‐inflammatory state, which was abolished upon MSR1 deletion or JNK inhibition. Moreover, MSR1 K63 polyubiquitylation correlated with the activation of JNK signalling in ovarian cancer tissue from human patients, suggesting that it may be relevant for macrophage phenotypic shift in vivo. Altogether, we identified that MSR1 signals through JNK via K63 polyubiquitylation and provides evidence for the receptor's involvement in macrophage polarization.
Collapse
Affiliation(s)
- Manman Guo
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Anetta Härtlova
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK .,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Microbiology and Immunology, Institute for Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Marek Gierliński
- Data Analysis Group, School of Life Sciences, University of Dundee, Dundee, UK
| | - Alan Prescott
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Josep Castellvi
- Department of Pathology, Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Javier Hernandez Losa
- Department of Pathology, Hospital Universitario Vall d'Hebron, Barcelona, Spain.,Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Barcelona, Spain
| | - Sine K Petersen
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Microbiology and Immunology, Institute for Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf A Wenzel
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Microbiology and Immunology, Institute for Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Brian D Dill
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Christoph H Emmerich
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Santiago Ramon Y Cajal
- Department of Pathology, Hospital Universitario Vall d'Hebron, Barcelona, Spain.,Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Barcelona, Spain
| | - David G Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Matthias Trost
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK .,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
238
|
Ölander M, Handin N, Artursson P. Image-Based Quantification of Cell Debris as a Measure of Apoptosis. Anal Chem 2019; 91:5548-5552. [PMID: 31001971 DOI: 10.1021/acs.analchem.9b01243] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Apoptosis is a controlled form of cell death that can be induced by various diseases and exogenous toxicants. Common apoptosis-detection methods rely on fluorescent markers, which necessitate the use of costly reagents and time-consuming labeling procedures. Label-free methods avoid these problems, but often require specialized instruments instead. Here, we utilize apoptotic-cell disintegration to develop a novel label-free detection method based on the quantification of subcellular debris particles in bright-field-microscopy images. Debris counts show strong correlations with fluorescence-based annexin V staining and can be used to study concentration-dependent and temporal apoptosis activation. The method is rapid, low-cost, and easy to apply, as the only experimental step comprises bright-field imaging of culture-media samples followed by automated image processing. The late-stage nature of the debris measurement means that the method can complement other, established apoptosis assays, and its accessibility will allow a wider community of researchers to study apoptotic cell death.
Collapse
Affiliation(s)
- Magnus Ölander
- Department of Pharmacy , Uppsala University , SE-75123 Uppsala , Sweden
| | - Niklas Handin
- Department of Pharmacy , Uppsala University , SE-75123 Uppsala , Sweden
| | - Per Artursson
- Department of Pharmacy and Science for Life Laboratory , Uppsala University , SE-75123 Uppsala , Sweden
| |
Collapse
|
239
|
Galloway DA, Phillips AEM, Owen DRJ, Moore CS. Phagocytosis in the Brain: Homeostasis and Disease. Front Immunol 2019; 10:790. [PMID: 31040847 PMCID: PMC6477030 DOI: 10.3389/fimmu.2019.00790] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/26/2019] [Indexed: 12/28/2022] Open
Abstract
Microglia are resident macrophages of the central nervous system and significantly contribute to overall brain function by participating in phagocytosis during development, homeostasis, and diseased states. Phagocytosis is a highly complex process that is specialized for the uptake and removal of opsonized and non-opsonized targets, such as pathogens, apoptotic cells, and cellular debris. While the role of phagocytosis in mediating classical innate and adaptive immune responses has been known for decades, it is now appreciated that phagocytosis is also critical throughout early neural development, homeostasis, and initiating repair mechanisms. As such, modulating phagocytic processes has provided unexplored avenues with the intent of developing novel therapeutics that promote repair and regeneration in the CNS. Here, we review the functional consequences that phagocytosis plays in both the healthy and diseased CNS, and summarize how phagocytosis contributes to overall pathophysiological mechanisms involved in brain injury and repair.
Collapse
Affiliation(s)
- Dylan A Galloway
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Alexandra E M Phillips
- Division of Brain Sciences, Department of Medicine Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - David R J Owen
- Division of Brain Sciences, Department of Medicine Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Craig S Moore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
240
|
Huang W, Lin ET, Hsu Y, Lin S. Anagen hair follicle repair: Timely regenerative attempts from plastic extra‐bulge epithelial cells. Exp Dermatol 2019; 28:406-412. [DOI: 10.1111/exd.13889] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/26/2018] [Accepted: 01/15/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Wen‐Yen Huang
- Department of Biomedical EngineeringNational Taiwan University Taipei Taiwan
| | - Edrick Tai‐Yu Lin
- Department of Biomedical EngineeringNational Taiwan University Taipei Taiwan
- Department of DermatologyNational Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan
| | - Ya‐Chieh Hsu
- Department of Stem Cell and Regenerative BiologyHarvard University and Harvard Stem Cell Institute Cambridge Massachusetts
| | - Sung‐Jan Lin
- Department of Biomedical EngineeringNational Taiwan University Taipei Taiwan
- Department of DermatologyNational Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan
- Research Center for Developmental Biology and Regenerative MedicineNational Taiwan University Taipei Taiwan
- Graduate Institute of Clinical MedicineCollege of MedicineNational Taiwan University Taipei Taiwan
| |
Collapse
|
241
|
Andrade H, Lin W, Zhang Y. Specificity from nonspecific interaction: regulation of tumor necrosis factor-α activity by DNA. J Biol Chem 2019; 294:6397-6404. [PMID: 30814250 DOI: 10.1074/jbc.ra119.007586] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/20/2019] [Indexed: 11/06/2022] Open
Abstract
As anionic biopolymers, oligonucleotides can have biological functions independent from their roles as the medium for the storage and flow of genetic information. In this paper, we investigated the interaction between DNA and the pro-inflammatory cytokine tumor necrosis factor-α (TNFα). Although various forms of DNA bind to TNFα with low μm dissociation constants, the interaction stabilizes the trimeric form of TNFα and enhances its cytotoxic effect. Based on this mechanism, a photoswitchable TNFα (TNFα-2-nitroveratryloxycarbonyl) has been designed whose sensitivity to DNA-mediated up-regulation of TNFα activity can be tuned by light irradiation. The mechanism described in this study represents a general model to understand the involvement of nonspecific interactions among biomolecules in regulating their biological functions. Because the interaction is not DNA sequence-specific, the resulting effect should be considered for oligonucleotide-based therapeutics in general.
Collapse
Affiliation(s)
- Helena Andrade
- From the B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, 01307 Dresden, Germany
| | - Weilin Lin
- From the B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, 01307 Dresden, Germany
| | - Yixin Zhang
- From the B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
242
|
Kim Y, Lee J, Lee D, Othmer HG. Synergistic Effects of Bortezomib-OV Therapy and Anti-Invasive Strategies in Glioblastoma: A Mathematical Model. Cancers (Basel) 2019; 11:E215. [PMID: 30781871 PMCID: PMC6406513 DOI: 10.3390/cancers11020215] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 12/18/2022] Open
Abstract
It is well-known that the tumor microenvironment (TME) plays an important role in the regulation of tumor growth and the efficacy of anti-tumor therapies. Recent studies have demonstrated the potential of combination therapies, using oncolytic viruses (OVs) in conjunction with proteosome inhibitors for the treatment of glioblastoma, but the role of the TME in such therapies has not been studied. In this paper, we develop a mathematical model for combination therapies based on the proteosome inhibitor bortezomib and the oncolytic herpes simplex virus (oHSV), with the goal of understanding their roles in bortezomib-induced endoplasmic reticulum (ER) stress, and how the balance between apoptosis and necroptosis is affected by the treatment protocol. We show that the TME plays a significant role in anti-tumor efficacy in OV combination therapy, and illustrate the effect of different spatial patterns of OV injection. The results illustrate a possible phenotypic switch within tumor populations in a given microenvironment, and suggest new anti-invasion therapies.
Collapse
Affiliation(s)
- Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul 05029, Korea.
| | - Junho Lee
- Department of Mathematics, Konkuk University, Seoul 05029, Korea.
| | - Donggu Lee
- Department of Mathematics, Konkuk University, Seoul 05029, Korea.
| | - Hans G Othmer
- School of Mathematics, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
243
|
TRADD regulates perinatal development and adulthood survival in mice lacking RIPK1 and RIPK3. Nat Commun 2019; 10:705. [PMID: 30741936 PMCID: PMC6370879 DOI: 10.1038/s41467-019-08584-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 01/18/2019] [Indexed: 11/08/2022] Open
Abstract
TRADD is an adaptor for TNFR1-induced apoptosis and NFκB activation. However, TRADD-deficient mice undergo normal development and contain normal lymphoid populations, which contrasts with an embryonic defect in mice lacking FADD, the shared adaptor mediating apoptosis. Recent studies indicate FADD suppresses embryonic necroptosis mediated by RIPK1. TRADD was suggested to also mediate necroptosis. Here we report that targeting TRADD fails to rescue Fadd-/- embryos from necroptosis, and ablation of TRADD rescues Ripk1-/- mice from perinatal lethality when RIPK3-mediated necroptosis is disabled. The resulting Ripk1-/-Ripk3-/-Tradd-/- mice survive until early adulthood, but die thereafter. A single allele of Tradd is optimal for survival of Ripk1-/-Ripk3-/-Tradd+/- mice. We show that TRADD plays a more dominating role in NFκB-signaling than RIPK1. While RIPK1 protects thymocytes from TNFα-induced apoptosis, TRADD promotes this process. The data demonstrate that TRADD is critical in perinatal and adult mice lacking RIPK1 and RIPK3, which has not been appreciated in prior studies.
Collapse
|
244
|
Phosphorylation-mediated activation of mouse Xkr8 scramblase for phosphatidylserine exposure. Proc Natl Acad Sci U S A 2019; 116:2907-2912. [PMID: 30718401 DOI: 10.1073/pnas.1820499116] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The exposure of phosphatidylserine (PtdSer) to the cell surface is regulated by the down-regulation of flippases and the activation of scramblases. Xkr8 has been identified as a scramblase that is activated during apoptosis, but its exogenous expression in the mouse Ba/F3 pro B cell line induces constitutive PtdSer exposure. Here we found that this Xkr8-mediated PtdSer exposure occurred at 4 °C, but not at 20 °C, although its scramblase activity was observed at 20 °C. The Xkr8-mediated PtdSer exposure was inhibited by a kinase inhibitor and enhanced by phosphatase inhibitors. Phosphorylated Xkr8 was detected by Phos-tag PAGE, and a mass spectrometric and mutational analysis identified three phosphorylation sites. Their phosphomimic mutation rendered Xkr8 resistant to the kinase inhibitor for PtdSer exposure at 4 °C, but unlike phosphatase inhibitors, it did not induce constitutive PtdSer exposure at 20 °C. On the other hand, when the flippase genes were deleted, the Xkr8 induced constitutive PtdSer exposure at high temperature, indicating that the flippase activity normally counteracted Xkr8's ability to expose PtdSer. These results indicate that PtdSer exposure can be increased by the phosphorylation-mediated activation of Xkr8 scramblase and flippase down-regulation.
Collapse
|
245
|
A noncanonical role for the engulfment gene ELMO1 in neutrophils that promotes inflammatory arthritis. Nat Immunol 2019; 20:141-151. [PMID: 30643265 PMCID: PMC6402828 DOI: 10.1038/s41590-018-0293-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 12/04/2018] [Indexed: 12/15/2022]
Abstract
Rheumatoid arthritis is characterized by progressive joint inflammation and affects ~1% of the human population. We noted single nucleotide polymorphisms (SNPs) in the apoptotic cell engulfment genes ELMO1, DOCK2, and RAC1 linked to rheumatoid arthritis. As ELMO1 promotes cytoskeletal reorganization during engulfment, we hypothesized that ELMO1 loss would worsen inflammatory arthritis. Surprisingly, Elmo1-deficient mice showed reduced joint inflammation in acute and chronic arthritis models. Genetic and cell biological studies revealed that ELMO1 associates with receptors linked to neutrophil function in arthritis and regulates activation and early neutrophil recruitment to the joints, without general inhibition of inflammatory responses. Further, neutrophils from peripheral blood of human donors that carry the SNP in ELMO1 associated with arthritis display increased migratory capacity, whereas ELMO1 knockdown reduces human neutrophil migration to chemokines linked to arthritis. These data identify ‘non-canonical’ roles for ELMO1 as an important cytoplasmic regulator of specific neutrophil receptors and promoter of arthritis.
Collapse
|
246
|
Shi Y, Zhu R, Xue Z, Han J, Han S. An in cellulo-activated multicolor cell labeling approach used to image dying cell clearance. Analyst 2019; 144:4687-4693. [DOI: 10.1039/c9an00904c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dying cell clearance is critical for myriad biological processes such as tissue homeostasis.
Collapse
Affiliation(s)
- Yilong Shi
- State key Laboratory of Cellular Stress Biology
- Innovation Center for Cell Signalling Network
- School of Life Sciences
- Xiamen University
- Xiamen, Fujian
| | - Rui Zhu
- State Key Laboratory for Physical Chemistry of Solid Surfaces
- Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- the Key Laboratory for Chemical Biology of Fujian Province
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
| | - Zhongwei Xue
- State Key Laboratory for Physical Chemistry of Solid Surfaces
- Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- the Key Laboratory for Chemical Biology of Fujian Province
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
| | - Jiahuai Han
- State key Laboratory of Cellular Stress Biology
- Innovation Center for Cell Signalling Network
- School of Life Sciences
- Xiamen University
- Xiamen, Fujian
| | - Shoufa Han
- State Key Laboratory for Physical Chemistry of Solid Surfaces
- Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- the Key Laboratory for Chemical Biology of Fujian Province
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
| |
Collapse
|
247
|
Kim YS, Majid M, Melchiorri AJ, Mikos AG. Applications of decellularized extracellular matrix in bone and cartilage tissue engineering. Bioeng Transl Med 2019; 4:83-95. [PMID: 30680321 PMCID: PMC6336671 DOI: 10.1002/btm2.10110] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 12/12/2022] Open
Abstract
Regenerative therapies for bone and cartilage injuries are currently unable to replicate the complex microenvironment of native tissue. There are many tissue engineering approaches attempting to address this issue through the use of synthetic materials. Although synthetic materials can be modified to simulate the mechanical and biochemical properties of the cell microenvironment, they do not mimic in full the multitude of interactions that take place within tissue. Decellularized extracellular matrix (dECM) has been established as a biomaterial that preserves a tissue's native environment, promotes cell proliferation, and provides cues for cell differentiation. The potential of dECM as a therapeutic agent is rising, but there are many limitations of dECM restricting its use. This review discusses the recent progress in the utilization of bone and cartilage dECM through applications as scaffolds, particles, and supplementary factors in bone and cartilage tissue engineering.
Collapse
Affiliation(s)
- Yu Seon Kim
- Dept. of BioengineeringRice UniversityHoustonTX 77005
| | - Marjan Majid
- Dept. of BioengineeringRice UniversityHoustonTX 77005
| | | | - Antonios G. Mikos
- Dept. of BioengineeringRice UniversityHoustonTX 77005
- Biomaterials LabRice UniversityHoustonTX 77005
| |
Collapse
|
248
|
Zhang HL, Zhu YM, Zhou XY. Coordination of Autophagy and Other Cellular Activities. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1206:697-727. [PMID: 31777007 DOI: 10.1007/978-981-15-0602-4_30] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Conventionally, autophagy (=self-eating) is thought to be a catabolic cellular process that is responsible for regulating cell homeostasis. However, the newly evidence have expanded the range of the impact of autophagy in biology. Autophagy interplays with endocytosis through shared factors such as phosphatidylinositol 3 kinase complex (PI(3)K complex), autophagy associated gene (Atg), and lysosome. Autophagy and phagocytosis orchestrate in maintaining homeostasis, in MHC class II antigen processing, in the removal of pathogens, in cell death, immunity, and inflammation. There are numerous cross talks of autophagy with biosynthetic processes such as conventional and unconventional secretion of biologically active cargo and trafficking of integral membrane proteins, as well as the exosome secretion. There are also links between autophagy and trafficking events from plasma membrane, including lateral plasma membrane proteins connexins, cell connections, and ciliogenesis.
Collapse
Affiliation(s)
- Hui-Ling Zhang
- Department of Pharmacology, Laboratory of Cerebrovascular Pharmacology, Soochow University, 199 Ren-Ai Road, Suzhou, Jiangsu, 215123, China.
| | - Yong-Ming Zhu
- Department of Pharmacology, Laboratory of Cerebrovascular Pharmacology, Soochow University, 199 Ren-Ai Road, Suzhou, Jiangsu, 215123, China
| | - Xian-Yong Zhou
- Department of Pharmacology, Laboratory of Cerebrovascular Pharmacology, Soochow University, 199 Ren-Ai Road, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
249
|
Ramezani G, Pourgheysari B, Shirzad H, Sourani Z. Pterostilbene increases Fas expression in T-lymphoblastic leukemia cell lines. Res Pharm Sci 2019; 14:55-63. [PMID: 30936933 PMCID: PMC6407337 DOI: 10.4103/1735-5362.251853] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Treatment of acute lymphoblastic leukemia (ALL) has been promising in last decades, but side effects still persist and searching for the least toxic agents continue. Pterostilbene (PTE) is a natural compound with several anti-cancer and anti-oxidant properties. Fas, as a member of death inducing family of tumor necrosis factor (TNF) receptors with an intracellular death domain, can initiate the extrinsic apoptosis signaling pathway. Here after the half maximal inhibitory concentration (IC50) determination in cell lines, we searched for PTE effects on Fas, both in mRNA and surface levels in two ALL cell lines, Jurkat and Molt-4. After harvesting cells in optimum situations, MTS assay was used to determine IC50 concentrations. Real-time polymerase chain reaction (RT-PCR) and flow cytometry were performed for Fas mRNA and surface expression variations after exposure to PTE. The findings showed that PTE decreases cell viability with different extent in two ALL cell lines. In addition to inducing apoptosis, it can increase Fas in both gene and cell surface expression in the same concentrations. Pterostilbene as a natural anti-cancer agent can increase Fas expression both in mRNA and surface levels that results in apoptosis signal transduction improvement which sensitizes cells to apoptosis by immune effector cells. As a result, abnormal cells removal would be more efficiently with the minimum side effects on normal cells.
Collapse
Affiliation(s)
- Gelareh Ramezani
- Immunology Department, Shahrekord University of Medical Sciences, Shahrekord, I.R. Iran
| | - Batoul Pourgheysari
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, I.R. Iran.,Pathology and Hematology Department, Shahrekord University of Medical Sciences, Shahrekord, I.R. Iran
| | - Hedayatollah Shirzad
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, I.R. Iran
| | - Zahra Sourani
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, I.R. Iran.,Department of Medical Laboratory Sciences, Shahrekord University of Medical Sciences, Shahrekord, I.R. Iran
| |
Collapse
|
250
|
Wang H, Wang G, Ansari GAS, Khan MF. Trichloroethene metabolite dichloroacetyl chloride induces apoptosis and compromises phagocytosis in Kupffer Cells: Activation of inflammasome and MAPKs. PLoS One 2018; 13:e0210200. [PMID: 30596806 PMCID: PMC6312261 DOI: 10.1371/journal.pone.0210200] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 12/18/2018] [Indexed: 12/20/2022] Open
Abstract
Exposure to trichloroethene (TCE), an occupational and ubiquitous environmental contaminant, is associated with the development of several autoimmune diseases, including autoimmune hepatitis (AIH). However, mechanisms contributing to TCE-mediated AIH are not known. Earlier, we have shown that dichloroacetyl chloride (DCAC), one of the reactive metabolites of TCE with strong acylating capability, can elicit an autoimmune response at much lower dose than TCE in female MRL+/+ mice. Furthermore, Kupffer cells (KCs), the liver resident macrophages, are crucial for hepatic homeostasis, but can also participate in the immunopathogenesis of AIH. However, contribution of KCs in TCE-mediated AIH and the underlying mechanisms are not understood. We hypothesized that increased apoptosis and delayed clearance of apoptotic bodies, due to compromised KC function, will result in the breakdown of self-tolerance, autoimmunity, and ultimately AIH. Therefore, using an in vitro model of immortalized mouse KCs, we investigated the contribution of DCAC in TCE-mediated AIH. KCs were treated with different concentrations of DCAC and apoptosis was measured by Annexin V and PI staining. Also, the impact of DCAC on phagocytic potential of KCs was evaluated. Furthermore, markers of inflammasome (NLRP3 and caspase1) were analyzed by real-time PCR and Western blot analysis. DCAC treatment resulted in significantly increased early and late-stage apoptosis, accompanied with inflammasome activation (NLRP3 increases). DCAC treatment resulted in decreased phagocytic function of KCs in a dose-dependent manner, with reduced MFG-E8 levels (phagocytotic function). Furthermore, DCAC exposure led to induction of phos-ERK and phos-AKT signaling. These findings suggest that DCAC induces apoptosis and inflammasome activation, while compromising the phagocytic function of KCs. Our data support that increased apoptosis and impaired KC function by DCAC could be contributory to TCE-mediated AIH.
Collapse
Affiliation(s)
- Hui Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| | - Gangduo Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| | | | - M. Firoze Khan
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|