201
|
Rosowski EE. Determining macrophage versus neutrophil contributions to innate immunity using larval zebrafish. Dis Model Mech 2020; 13:13/1/dmm041889. [PMID: 31932292 PMCID: PMC6994940 DOI: 10.1242/dmm.041889] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The specific roles of the two major innate immune cell types – neutrophils and macrophages – in response to infection and sterile inflammation are areas of great interest. The larval zebrafish model of innate immunity, and the imaging capabilities it provides, is a source of new research and discoveries in this field. Multiple methods have been developed in larval zebrafish to specifically deplete functional macrophages or neutrophils. Each of these has pros and cons, as well as caveats, that often make it difficult to directly compare results from different studies. The purpose of this Review is to (1) explore the pros, cons and caveats of each of these immune cell-depleted models; (2) highlight and place into a broader context recent key findings on the specific functions of innate immune cells using these models; and (3) explore future directions in which immune cell depletion methods are being expanded. Summary: Macrophages and neutrophils are distinct innate immune cells with diverse roles in diverse inflammatory contexts. Recent research in larval zebrafish using cell-specific depletion methods has revealed new insights into these cells' functions.
Collapse
Affiliation(s)
- Emily E Rosowski
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
202
|
Ding X, Chen J, Wu C, Wang G, Zhou C, Chen S, Wang K, Zhang A, Ye P, Wu J, Chen S, Zhang H, Xu K, Wang S, Xia J. Nucleotide-Binding Oligomerization Domain-Like Receptor Protein 3 Deficiency in Vascular Smooth Muscle Cells Prevents Arteriovenous Fistula Failure Despite Chronic Kidney Disease. J Am Heart Assoc 2020; 8:e011211. [PMID: 30587058 PMCID: PMC6405733 DOI: 10.1161/jaha.118.011211] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background The arteriovenous fistula (AVF) is the preferred hemodialysis access for patients with chronic kidney disease. Chronic kidney disease can increase neointima formation, which greatly contributes to AVF failure by an unknown mechanism. Our study aimed to determine the role of nucleotide‐binding oligomerization domain‐like receptor protein 3 (NLRP3) in neointima formation induced by experimental AVFs in the presence of chronic kidney disease. Methods and Results From our findings, NLRP3 was upregulated in the intimal lesions of AVFs in both uremic mice and patients. Smooth muscle–specific knockout NLRP3 mice exhibited markedly decreased neointima formation in the outflow vein of AVFs. Compared with primary vascular smooth muscle cells isolated from control mice, those isolated from smooth muscle–specific knockout NLRP3 mice showed compromised proliferation, migration, phenotypic switching, and a weakened ability to activate mononuclear macrophages. To identify how NLRP3 functions, several small‐molecule inhibitors were used. The results showed that NLRP3 regulates smooth muscle cell proliferation and migration through Smad2/3 phosphorylation rather than through caspase‐1/interleukin‐1 signaling. Unexpectedly, the selective NLRP3‐inflammasome inhibitor MCC950 also repressed Smad2/3 phosphorylation and relieved chronic kidney disease–promoted AVF failure independent of macrophages. Conclusions Our findings suggest that NLRP3 in vascular smooth muscle cells may play a crucial role in uremia‐associated AVF failure and may be a promising therapeutic target for the treatment of AVF failure.
Collapse
Affiliation(s)
- Xiangchao Ding
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jiuling Chen
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Chuangyan Wu
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,2 Department of Thoracic Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Guohua Wang
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Cheng Zhou
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Shanshan Chen
- 3 Key Laboratory for Molecular Diagnosis of Hubei Province Central Hospital of Wuhan Tongji Medical College Huazhong University of Science and Technology Wuhan China.,4 Central Laboratory Central Hospital of Wuhan Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Ke Wang
- 6 Department of Respiratory and Critical Care Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Anchen Zhang
- 5 Department of Cardiovascular Medicine Central Hospital of Wuhan Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Ping Ye
- 5 Department of Cardiovascular Medicine Central Hospital of Wuhan Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jie Wu
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Shanshan Chen
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Hao Zhang
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Kaiying Xu
- 2 Department of Thoracic Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Sihua Wang
- 2 Department of Thoracic Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jiahong Xia
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
203
|
|
204
|
Shi M, Zhang P, Zhao Q, Shen K, Qiu Y, Xiao Y, Yuan Q, Zhang Y. Dual Functional Monocytes Modulate Bactericidal and Anti-Inflammation Process for Severe Osteomyelitis Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1905185. [PMID: 31880088 DOI: 10.1002/smll.201905185] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/04/2019] [Indexed: 06/10/2023]
Abstract
Osteomyelitis is an inflammatory bone disease caused by infection microorganisms which leads to progressive bone destruction and loss. Drug resistance and inflammatory damage make it urgent to develop new dual-functional therapies. Based on the powerful bactericidal effect of monocyte/macrophage cells by nature, a functional monocyte with programed anti-inflammatory ability is promising for osteomyelitis treatment. Herein, gold nanocage (GNC)-modified monocytes are developed which contain aspirin to realize the controlled antibacterial and anti-inflammatory process for bone infection treatment effectively. Aspirin@GNC-laden monocytes inherit the biological functions of origin monocytes such as chemotaxis to bacteria, differentiation potential, and phagocytic ability. The controlled release of aspirin from GNC has a beneficial effect on improving the rate and amount of bone regeneration after the anti-infection stage due to its ability to suppress the activity of natural immunity and induce osteoblast differentiation during the treatment of osteomyelitis. The present work described here is the first to utilize living monocytes to achieve a dual effect to antibacteria and anti-inflammation in a time-oriented and programed way, and provides an inspiration for future therapy based on this concept.
Collapse
Affiliation(s)
- Miusi Shi
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Peng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Qin Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Kailun Shen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yun Qiu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yin Xiao
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, 4059, Australia
| | - Quan Yuan
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Yufeng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
205
|
Hogg C, Horne AW, Greaves E. Endometriosis-Associated Macrophages: Origin, Phenotype, and Function. Front Endocrinol (Lausanne) 2020; 11:7. [PMID: 32038499 PMCID: PMC6989423 DOI: 10.3389/fendo.2020.00007] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/07/2020] [Indexed: 01/03/2023] Open
Abstract
Endometriosis is a complex, heterogeneous, chronic inflammatory condition impacting ~176 million women worldwide. It is associated with chronic pelvic pain, infertility, and fatigue, and has a substantial impact on health-related quality of life. Endometriosis is defined by the growth of endometrial-like tissue outside the uterus, typically on the lining of the pelvic cavity and ovaries (known as "lesions"). Macrophages are complex cells at the center of this enigmatic condition; they are critical for the growth, development, vascularization, and innervation of lesions as well as generation of pain symptoms. In health, tissue-resident macrophages are seeded during early embryonic life are vital for development and homeostasis of tissues. In the adult, under inflammatory challenge, monocytes are recruited from the blood and differentiate into macrophages in tissues where they fulfill functions, such as fighting infection and repairing wounds. The interplay between tissue-resident and recruited macrophages is now at the forefront of macrophage research due to their differential roles in inflammatory disorders. In some cancers, tumor-associated macrophages (TAMs) are comprised of tissue-resident macrophages and recruited inflammatory monocytes that differentiate into macrophages within the tumor. These macrophages of different origins play differential roles in disease progression. Herein, we review the complexities of macrophage dynamics in health and disease and explore the paradigm that under disease-modified conditions, macrophages that normally maintain homeostasis become modified such that they promote disease. We also interrogate the evidence to support the existence of multiple phenotypic populations and origins of macrophages in endometriosis and how this could be exploited for therapy.
Collapse
Affiliation(s)
- Chloe Hogg
- Medical Research Council Centre for Reproductive Health, The University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew W. Horne
- Medical Research Council Centre for Reproductive Health, The University of Edinburgh, Edinburgh, United Kingdom
| | - Erin Greaves
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- *Correspondence: Erin Greaves
| |
Collapse
|
206
|
Abstract
Nanomedicines have historically struggled to find clinical relevance and to achieve translation successfully. In this Perspective, we discuss possible reasons for this difficulty and highlight several key features of nanomedicines that are often overlooked by biomedical engineers. We present the notion of clinical multifunctionality as distinct from multifunctionality as it is traditionally described at the nanoscale and emphasize its importance through examples of nanomedicines that have demonstrated emergent clinical multifunctionality once translated. We also describe a phenomenon in which clinical multifunctionality results in diagonal translation after a nanomedicine is adopted by clinicians to serve as a solution for a clinical problem that it was not designed to solve. Biomedical engineers can take advantage of these phenomena to assist in achieving clinical translation of new nanomedicines.
Collapse
Affiliation(s)
- Keegan Guidolin
- Department of Surgery , University of Toronto , Toronto M5G 1L7 , Canada
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto M5G 1L7 , Canada
- Princess Margaret Cancer Centre , Toronto M5G 1L7 , Canada
| | - Gang Zheng
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto M5G 1L7 , Canada
- Department of Medical Biophysics , University of Toronto , Toronto M5G 1L7 , Canada
- Princess Margaret Cancer Centre , Toronto M5G 1L7 , Canada
| |
Collapse
|
207
|
Lentini G, Famà A, Biondo C, Mohammadi N, Galbo R, Mancuso G, Iannello D, Zummo S, Giardina M, De Gaetano GV, Teti G, Beninati C, Midiri A. Neutrophils Enhance Their Own Influx to Sites of Bacterial Infection via Endosomal TLR-Dependent Cxcl2 Production. THE JOURNAL OF IMMUNOLOGY 2019; 204:660-670. [PMID: 31852751 DOI: 10.4049/jimmunol.1901039] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/19/2019] [Indexed: 12/27/2022]
Abstract
The influx of neutrophils to infection sites is a fundamental step in host defenses against the frequent human pathogen group B Streptococcus (GBS) and other extracellular bacteria. Using a mouse model of GBS-induced peritonitis, we show in this study that the chemokines Cxcl1 and Cxcl2 play distinctive roles in enhancing the recruitment and the antibacterial activities of neutrophils in a manner that is linked to differences in the cellular sources of these mediators. Cell depletion experiments demonstrated that neutrophils make a significant contribution to the in vivo production of Cxcl2 but not Cxcl1. In vitro, neutrophils responded weakly to LPS but released high levels of Cxcl2 after stimulation with GBS or other bacteria. Neutrophil-derived Cxcl2 acted in an autocrinous manner to increase its own production and to enhance antibacterial activities, including the release of oxygen radicals. In both neutrophils and macrophages, the production of Cxcl1/2 largely required the presence of functional UNC93B1, a chaperone protein involved in signaling by endosomal TLRs. Moreover, the phenotype of UNC93B1-defective phagocytes could be recapitulated by the simultaneous absence of TLR7, 9, and 13 but not by the absence of individual TLRs. Collectively, our data show that neutrophils recognize Gram-positive and Gram-negative bacteria by means of multiple phagosomal TLRs, resulting in de novo synthesis of Cxcl2, amplification of neutrophil recruitment, and potentiation of their antibacterial activities. These data may be useful to devise alternative therapeutic strategies aimed at enhancing the recruitment and the functional activities of polymorphonuclear leukocytes during infections caused by antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Germana Lentini
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Agata Famà
- Charybdis Vaccines Srl, 98125 Messina, Italy
| | - Carmelo Biondo
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Nastaran Mohammadi
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Roberta Galbo
- Department of Chemical, Biological, Pharmaceutical Sciences and Environmental Sciences, University of Messina, 98166 Messina, Italy; and
| | - Giuseppe Mancuso
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Daniela Iannello
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Sebastiana Zummo
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Miriam Giardina
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | | | | | - Concetta Beninati
- Department of Human Pathology, University of Messina, 98125 Messina, Italy.,Scylla Biotech SRL, 98125 Messina, Italy
| | - Angelina Midiri
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| |
Collapse
|
208
|
PP2ACα of Alveolar Macrophages Is a Novel Protective Factor for LPS-Induced Acute Respiratory Distress Syndrome. Inflammation 2019; 42:1004-1014. [PMID: 30684253 DOI: 10.1007/s10753-019-00962-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Protein phosphatase 2A (PP2A) is one main serine/threonine phosphatase in eukaryotes, and its activation changes have been linked to modulation of numerous pathological processes, such as cancer, inflammation, fibrosis, and neurodegenerative diseases. Acute respiratory distress syndrome (ARDS), the major cause of respiratory failure, remains with limited therapies available up to now. Alveolar macrophages (AMs) are essential to innate immunity and host defense, participating in the pathogenesis of ARDS. As a result, AMs are considered as a potential therapeutic target for ARDS. In our study, we firstly found that PP2A activity was significantly decreased in the lipopolysaccharide (LPS)-stimulated AMs. Furthermore, adoptive transfer of AMs with enhanced PP2A enzyme activity that was improved by C2-ceramide prior to LPS exposure alleviated acute lung inflammation. Conversely, AM-specific ablation of PP2ACα exacerbated inflammatory responses to LPS. Mechanistically, PP2ACα negatively regulates LPS-induced cytokine secretion of AMs by NF-κB and MAPK pathways. Together, these findings provide the evidence to guide the development of novel therapeutic options targeting PP2ACα for ARDS/acute lung injury.
Collapse
|
209
|
He W, Kapate N, Shields CW, Mitragotri S. Drug delivery to macrophages: A review of targeting drugs and drug carriers to macrophages for inflammatory diseases. Adv Drug Deliv Rev 2019; 165-166:15-40. [PMID: 31816357 DOI: 10.1016/j.addr.2019.12.001] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/16/2022]
Abstract
Macrophages play a key role in defending against foreign pathogens, healing wounds, and regulating tissue homeostasis. Driving this versatility is their phenotypic plasticity, which enables macrophages to respond to subtle cues in tightly coordinated ways. However, when this coordination is disrupted, macrophages can aid the progression of numerous diseases, including cancer, cardiovascular disease, and autoimmune disease. The central link between these disorders is aberrant macrophage polarization, which misguides their functional programs, secretory products, and regulation of the surrounding tissue microenvironment. As a result of their important and deterministic roles in both health and disease, macrophages have gained considerable attention as targets for drug delivery. Here, we discuss the role of macrophages in the initiation and progression of various inflammatory diseases, summarize the leading drugs used to regulate macrophages, and review drug delivery systems designed to target macrophages. We emphasize strategies that are approved for clinical use or are poised for clinical investigation. Finally, we provide a prospectus of the future of macrophage-targeted drug delivery systems.
Collapse
Affiliation(s)
- Wei He
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Neha Kapate
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - C Wyatt Shields
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
210
|
Li W, Zhang X, Wu F, Zhou Y, Bao Z, Li H, Zheng P, Zhao S. Gastric cancer-derived mesenchymal stromal cells trigger M2 macrophage polarization that promotes metastasis and EMT in gastric cancer. Cell Death Dis 2019; 10:918. [PMID: 31801938 PMCID: PMC6892854 DOI: 10.1038/s41419-019-2131-y] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/03/2019] [Accepted: 11/05/2019] [Indexed: 12/16/2022]
Abstract
Resident macrophages in the tumor microenvironment exert a dual role in tumor progression. So far, the mechanism of intratumoral macrophage generation is still largely unknown. In the present study, the importance of macrophages in the pro-tumor role of gastric cancer-derived mesenchymal stromal cells (GC-MSCs) was observed in a mouse xenograft model with macrophage depletion. In gastric cancer tissues, high expression levels of Ym-1, Fizz-1, arginase-1, and CCR-2, as well as a low expression level of iNOS, were verified, and co-localization of GC-MSCs and tumor-associated macrophages (TAMs) was observed by dual immunofluorescence histochemistry. TAMs isolated from gastric cancer tissues predominantly displayed an M2 phenotype. In a co-culture system, the contribution of GC-MSCs to M2 polarization of macrophages was confirmed by the M2-related protein expression, M2-like immunophenotype and cytokine profile of GC-MSC-primed macrophages in vitro. Blockade of IL-6/IL-8 by neutralizing antibodies significantly attenuated the promoting effect of GC-MSCs on M2-like macrophage polarization via the JAK2/STAT3 signaling pathway. In addition, GC-MSC-primed macrophages promoted the migration and invasion of gastric cancer cells, and the process of EMT in gastric cancer cells was significantly enhanced by GC-MSC-primed macrophage treatment. Our study showed that tumor-promoting GC-MSCs contribute to M2 macrophage polarization within the gastric cancer niche through considerable secretion of IL-6 and IL-8. These GC-MSC-primed macrophages can subsequently prompt gastric cancer metastasis via EMT promotion in gastric cancer cells.
Collapse
Affiliation(s)
- Wei Li
- Center of Research Laboratory, The First People's Hospital of Lianyungang, Lianyungang, 222001, China.
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Fenglei Wu
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, 222001, China
| | - Ying Zhou
- Center of Research Laboratory, The First People's Hospital of Lianyungang, Lianyungang, 222001, China
| | - Zengtao Bao
- Department of Gastrointestinal Surgery, The First People's Hospital of Lianyungang, Lianyungang, 222001, China
| | - Haining Li
- Center of Research Laboratory, The First People's Hospital of Lianyungang, Lianyungang, 222001, China
- Department of Clinical Laboratory Diagnostics, Kangda College of Nanjing Medical University, Lianyungang, 222000, China
| | - Ping Zheng
- Center of Research Laboratory, The First People's Hospital of Lianyungang, Lianyungang, 222001, China
| | - Shaolin Zhao
- Center of Research Laboratory, The First People's Hospital of Lianyungang, Lianyungang, 222001, China.
| |
Collapse
|
211
|
Shwartz A, Goessling W, Yin C. Macrophages in Zebrafish Models of Liver Diseases. Front Immunol 2019; 10:2840. [PMID: 31867007 PMCID: PMC6904306 DOI: 10.3389/fimmu.2019.02840] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 11/19/2019] [Indexed: 12/16/2022] Open
Abstract
Hepatic macrophages are key components of the liver immunity and consist of two main populations. Liver resident macrophages, known as Kupffer cells in mammals, are crucial for maintaining normal liver homeostasis. Upon injury, they become activated to release proinflammatory cytokines and chemokines and recruit a large population of inflammatory monocyte-derived macrophages to the liver. During the progression of liver diseases, macrophages are highly plastic and have opposing functions depending on the signaling cues that they receive from the microenvironment. A comprehensive understanding of liver macrophages is essential for developing therapeutic interventions that target these cells in acute and chronic liver diseases. Mouse studies have provided the bulk of our current knowledge of liver macrophages. The emergence of various liver disease models and availability of transgenic tools to visualize and manipulate macrophages have made the teleost zebrafish (Danio rerio) an attractive new vertebrate model to study liver macrophages. In this review, we summarize the origin and behaviors of macrophages in healthy and injured livers in zebrafish. We highlight the roles of macrophages in zebrafish models of alcoholic and non-alcoholic liver diseases, hepatocellular carcinoma, and liver regeneration, and how they compare with the roles that have been described in mammals. We also discuss the advantages and challenges of using zebrafish to study liver macrophages.
Collapse
Affiliation(s)
- Arkadi Shwartz
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Wolfram Goessling
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Harvard Stem Cell Institute, Cambridge, MA, United States
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
- Broad Institute, Massachusetts Institute of Technology and Harvard, Cambridge, MA, United States
- Division of Health Sciences and Technology, Harvard and Massachusetts Institute of Technology, Boston, MA, United States
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Chunyue Yin
- Division of Gastroenterology, Hepatology and Nutrition and Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
212
|
Liu F, Dai S, Feng D, Peng X, Qin Z, Kearns AC, Huang W, Chen Y, Ergün S, Wang H, Rappaport J, Bryda EC, Chandrasekhar A, Aktas B, Hu H, Chang SL, Gao B, Qin X. Versatile cell ablation tools and their applications to study loss of cell functions. Cell Mol Life Sci 2019; 76:4725-4743. [PMID: 31359086 PMCID: PMC6858955 DOI: 10.1007/s00018-019-03243-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/17/2019] [Accepted: 07/22/2019] [Indexed: 12/22/2022]
Abstract
Targeted cell ablation is a powerful approach for studying the role of specific cell populations in a variety of organotypic functions, including cell differentiation, and organ generation and regeneration. Emerging tools for permanently or conditionally ablating targeted cell populations and transiently inhibiting neuronal activities exhibit a diversity of application and utility. Each tool has distinct features, and none can be universally applied to study different cell types in various tissue compartments. Although these tools have been developed for over 30 years, they require additional improvement. Currently, there is no consensus on how to select the tools to answer the specific scientific questions of interest. Selecting the appropriate cell ablation technique to study the function of a targeted cell population is less straightforward than selecting the method to study a gene's functions. In this review, we discuss the features of the various tools for targeted cell ablation and provide recommendations for optimal application of specific approaches.
Collapse
Affiliation(s)
- Fengming Liu
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Shen Dai
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xiao Peng
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Zhongnan Qin
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Alison C Kearns
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Wenfei Huang
- Institute of NeuroImmune Pharmacology, Seton Hall University, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Yong Chen
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Key Lab for Immunology in Universities of Shandong Province, School of Clinical Medicine, Weifang Medical University, 261053, Weifang, People's Republic of China
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximillan University, 97070, Wurzburg, Germany
| | - Hong Wang
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Jay Rappaport
- Division of Pathology, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Elizabeth C Bryda
- Rat Resource and Research Center, University of Missouri, 4011 Discovery Drive, Columbia, MO, 65201, USA
| | - Anand Chandrasekhar
- Division of Biological Sciences, 340D Life Sciences Center, University of Missouri, 1201 Rollins St, Columbia, MO, USA
| | - Bertal Aktas
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Hongzhen Hu
- Department of Anesthesiology, Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sulie L Chang
- Institute of NeuroImmune Pharmacology, Seton Hall University, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xuebin Qin
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA.
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA.
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
213
|
Men Y, Li W, Tu Y, Peng F, Janssen GJA, Nolte RJM, Wilson DA. Nonequilibrium Reshaping of Polymersomes via Polymer Addition. ACS NANO 2019; 13:12767-12773. [PMID: 31697471 PMCID: PMC6887890 DOI: 10.1021/acsnano.9b04740] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Polymersomes are a class of artificial liposomes, assembled from amphiphilic synthetic block copolymers, holding great promise toward applications in nanomedicine. The diversity in polymersome morphological shapes and, in particular, the precise control of these shapes, which is an important aspect in drug delivery studies, remains a great challenge. This is due to a lack of general methodologies that can be applied and the inability to capture the morphologies at the nanometer scale. Here, we present a methodology that can accurately control the shape of polymersomes via the addition of polyethylene glycol (PEG) under nonequilibrium conditions. Various shapes including spheres, ellipsoids, tubes, discs, stomatocytes, nests, stomatocyte-in-stomatocytes, disc-in-discs, and large compound vesicles (LCVs) can be uniformly captured by adjusting the water content and the PEG concentration. Moreover, these shapes undergo nonequilibrium changes in time, which is reflected in their phase diagram changes. This research provides a universal tool to fabricate all shapes of polymersomes by controlling three variables: water content, PEG concentration, and time. The use of the biofriendly polymer PEG enables the application of this methodology in the field of nanomedicine.
Collapse
|
214
|
Jia X, Gábris F, Jacobsen Ó, Bedics G, Botz B, Helyes Z, Kellermayer Z, Vojkovics D, Berta G, Nagy N, Jakus Z, Balogh P. Foliate Lymphoid Aggregates as Novel Forms of Serous Lymphocyte Entry Sites of Peritoneal B Cells and High-Grade B Cell Lymphomas. THE JOURNAL OF IMMUNOLOGY 2019; 204:23-36. [PMID: 31767783 DOI: 10.4049/jimmunol.1900851] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/25/2019] [Indexed: 11/19/2022]
Abstract
The cellular homeostasis of lymphoid tissues is determined by the continuous interactions of mobile hematopoietic cells within specialized microenvironments created by sessile stromal cells. In contrast to the lymph nodes and mucosal lymphoid tissues with well-defined entry and exit routes, the movement of leukocytes in the peritoneal cavity is largely unknown. In this study, we report that, in addition to the omental milky spots and fat-associated lymphoid clusters, in mice, the serous surface of the mesenteric adipose streaks contains lymphocyte-rich organoids comprised of a highly compacted leaf-like part connected to the adipose tissue that can also efficiently bind B cells and high-grade B cell lymphoma (diffuse large B cell lymphoma) cells. Denoted as foliate lymphoid aggregates (FLAgs), these structures show incomplete T/B segregation and a partially differentiated stromal architecture. LYVE-1-positive macrophages covering FLAgs efficiently bind i.p. injected normal B cells as well as different types of diffuse large B cell lymphoma cells. Within FLAgs, the lymphocytes compartmentalize according to their chemokine receptor pattern and subsequently migrate toward the mesenteric lymph nodes via the mesenteric lymphatic capillaries. The blood supply of FLAgs includes short vascular segments displaying peripheral lymph node addressin, and the extravasation of lymphocytes to the omental and mesenteric adipose tissues is partly mediated by L-selectin. The appearance of i.p. injected cells in mesenteric lymph nodes suggests that the mesentery-associated lymphatics may also collect leukocytes from the fat-associated lymphoid clusters and FLAgs, thus combining the mucosal and serous exit of mobile leukocytes and increasing the range of drainage sites for the peritoneal expansion of lymphoid malignancies.
Collapse
Affiliation(s)
- Xinkai Jia
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs, 7643 Pécs, Hungary
| | - Fanni Gábris
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs, 7643 Pécs, Hungary
| | - Óli Jacobsen
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs, 7643 Pécs, Hungary
| | - Gábor Bedics
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs, 7643 Pécs, Hungary
| | - Bálint Botz
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary.,Molecular Pharmacology Research Group, Szentágothai Research Center, University of Pécs, 7624 Pécs, Hungary.,Department of Radiology, Clinical Center, University of Pécs, 7624 Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary.,Molecular Pharmacology Research Group, Szentágothai Research Center, University of Pécs, 7624 Pécs, Hungary
| | - Zoltán Kellermayer
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs, 7643 Pécs, Hungary.,Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs, 7624 Pécs, Hungary
| | - Dóra Vojkovics
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs, 7643 Pécs, Hungary.,Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs, 7624 Pécs, Hungary
| | - Gergely Berta
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, 7643 Pécs, Hungary
| | - Nándor Nagy
- Department of Anatomy, Histology and Embryology, Semmelweis University, 1085 Budapest, Hungary
| | - Zoltán Jakus
- MTA-SE Lendulet Lymphatic Physiology Research Group of the Hungarian Academy of Sciences and Semmelweis University, 1094 Budapest, Hungary; and.,Department of Physiology, Semmelweis University, 1094 Budapest, Hungary
| | - Péter Balogh
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs, 7643 Pécs, Hungary; .,Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
215
|
Jin T, Mohammad M, Hu Z, Fei Y, Moore ERB, Pullerits R, Ali A. A novel mouse model for septic arthritis induced by Pseudomonas aeruginosa. Sci Rep 2019; 9:16868. [PMID: 31727989 PMCID: PMC6856550 DOI: 10.1038/s41598-019-53434-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/31/2019] [Indexed: 12/26/2022] Open
Abstract
Septic arthritis is one of the most aggressive joint diseases. Although caused predominantly by S. aureus, Gram-negative bacteria, Pseudomonas aeruginosa among them, account for a significant percentage of the causal agents of septic arthritis. However, septic arthritis caused by P. aeruginosa has not been studied thus far, due to lack of an animal model. NMRI mice were inoculated with different doses of P. aeruginosa. The clinical course of septic arthritis and radiological changes of joints were examined. Furthermore, the host molecular and cellular mechanisms involved in P. aeruginosa-induced septic arthritis were investigated. Inoculation of mice with P. aeruginosa caused septic arthritis in a dose-dependent manner. Neutrophil depletion led to higher mortality and more severe joint destruction (p < 0.01). In contrast, monocyte depletion resulted in higher mortality (p < 0.05) but similar arthritis severity compared to controls. Mice depleted of CD4+ T-cells inoculated with P. aeruginosa displayed less severe bone damage (p < 0.05). For the first time, a mouse model for P. aeruginosa septic arthritis is presented. Our data demonstrate that neutrophils play a protective role in P. aeruginosa septic arthritis. Monocytes/macrophages, on the other hand, are only essential in preventing P. aeruginosa-induced mortality. Finally, CD4+ T-cells are pathogenic in P. aeruginosa septic arthritis.
Collapse
Affiliation(s)
- Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden.,Department of Rheumatology, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Majd Mohammad
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| | - Zhicheng Hu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden.,Department of Microbiology and Immunology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ying Fei
- Department of Microbiology and Immunology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Edward R B Moore
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden.,Culture Collection University of Gothenburg (CCUG), Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden.,Centre for Antibiotic Resistance Research (CARe), University of Gothenburg, Göteborg, Sweden
| | - Rille Pullerits
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden.,Department of Rheumatology, Sahlgrenska University Hospital, Göteborg, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Abukar Ali
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden.
| |
Collapse
|
216
|
Carpentier KS, Davenport BJ, Haist KC, McCarthy MK, May NA, Robison A, Ruckert C, Ebel GD, Morrison TE. Discrete viral E2 lysine residues and scavenger receptor MARCO are required for clearance of circulating alphaviruses. eLife 2019; 8:e49163. [PMID: 31596239 PMCID: PMC6839921 DOI: 10.7554/elife.49163] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022] Open
Abstract
The magnitude and duration of vertebrate viremia is a critical determinant of arbovirus transmission, geographic spread, and disease severity. We find that multiple alphaviruses, including chikungunya (CHIKV), Ross River (RRV), and o'nyong 'nyong (ONNV) viruses, are cleared from the circulation of mice by liver Kupffer cells, impeding viral dissemination. Clearance from the circulation was independent of natural antibodies or complement factor C3, and instead relied on scavenger receptor SR-A6 (MARCO). Remarkably, lysine to arginine substitutions at distinct residues within the E2 glycoproteins of CHIKV and ONNV (E2 K200R) as well as RRV (E2 K251R) allowed for escape from clearance and enhanced viremia and dissemination. Mutational analysis revealed that viral clearance from the circulation is strictly dependent on the presence of lysine at these positions. These findings reveal a previously unrecognized innate immune pathway that controls alphavirus viremia and dissemination in vertebrate hosts, ultimately influencing disease severity and likely transmission efficiency.
Collapse
Affiliation(s)
- Kathryn S Carpentier
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Bennett J Davenport
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Kelsey C Haist
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Mary K McCarthy
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Nicholas A May
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Alexis Robison
- Department of Microbiology, Immunology, and PathologyColorado State UniversityFort CollinsUnited States
| | - Claudia Ruckert
- Department of Microbiology, Immunology, and PathologyColorado State UniversityFort CollinsUnited States
| | - Gregory D Ebel
- Department of Microbiology, Immunology, and PathologyColorado State UniversityFort CollinsUnited States
| | - Thomas E Morrison
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| |
Collapse
|
217
|
Ariyaratne A, Finney CAM. Eosinophils and Macrophages within the Th2-Induced Granuloma: Balancing Killing and Healing in a Tight Space. Infect Immun 2019; 87:e00127-19. [PMID: 31285249 PMCID: PMC6759305 DOI: 10.1128/iai.00127-19] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Granuloma formation is a key host immune response generated to confine invading pathogens and limit extensive host damage. It consists of an accumulation of host immune cells around a pathogen. This host response has been extensively studied in the context of inflammatory diseases. However, there is much less known about Th2-type granulomas generated in response to parasitic worms. Based on in vitro data, innate immune cells within the granuloma are thought to immobilize and kill parasites but also act to repair damaged tissue. Understanding this dual function is key. The two billion people and many livestock/wild animals infected with helminths demonstrate that granulomas are not effective at clearing infection. However, the lack of high mortality highlights their importance in ensuring that parasite migration/tissue damage is restricted and wound healing is effective. In this review, we define two key cellular players (macrophages and eosinophils) and their associated molecular players involved in Th2 granuloma function. To date, the underlying mechanisms remain poorly understood, which is in part due to a lack of conclusive studies. Most have been performed in vitro rather than in vivo, using cells that have not been obtained from granulomas. Experiments using genetically modified mouse strains and/or antibody/chemical-mediated cell depletion have also generated conflicting results depending on the model. We discuss the caveats of previous studies and the new tools available that will help fill the gaps in our knowledge and allow a better understanding of the balance between immune killing and healing.
Collapse
Affiliation(s)
- Anupama Ariyaratne
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Constance A M Finney
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
218
|
Abstract
BACKGROUND Sepsis is a major cause of acute kidney injury (AKI), with high rates of morbidity and mortality. M2 macrophages have been shown to play important roles in the secretion of anti-inflammatory and tissue repair mediators. In this study, we investigate the role of M2 macrophages in sepsis-induced AKI by depleting these cells in vivo through the systemic administration of liposomal clodronate (LC). METHODS Male Sprague-Dawley rats were subjected to cecal ligation and puncture (CLP) or sham surgery. Biochemical and histological renal damage was assessed. Macrophage infiltration and M2 macrophage depletion were assessed by immunohistochemistry. RT-PCR was used to investigate the expression of the inducible nitric oxide synthase (iNOS), arginase 1 (Arg-1), and found in inflammatory zone 1 (FIZZ1) mRNAs. Western blots were performed to assay the tissue levels of interleukin-10 (IL-10) and tumor necrosis factor alpha (TNF-α). RESULTS M2 macrophages were obviously detected 72 h after sepsis-induced AKI. Kidney injury was more severe, renal function was decreased, and blood creatinine and blood urea nitrogen (BUN) levels were higher after M2 macrophage depletion. M2 macrophage depletion significantly inhibited the proliferation of tubular cells. M2 macrophage depletion also downregulated IL-10 expression and increased TNF-α secretion during sepsis-induced AKI. CONCLUSIONS M2 macrophages attenuate sepsis-induced AKI, presumably by upregulating IL-10 expression and suppressing TNF-α secretion.
Collapse
|
219
|
Baaklini CS, Rawji KS, Duncan GJ, Ho MFS, Plemel JR. Central Nervous System Remyelination: Roles of Glia and Innate Immune Cells. Front Mol Neurosci 2019; 12:225. [PMID: 31616249 PMCID: PMC6764409 DOI: 10.3389/fnmol.2019.00225] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/04/2019] [Indexed: 12/31/2022] Open
Abstract
In diseases such as multiple sclerosis (MS), inflammation can injure the myelin sheath that surrounds axons, a process known as demyelination. The spontaneous regeneration of myelin, called remyelination, is associated with restoration of function and prevention of axonal degeneration. Boosting remyelination with therapeutic intervention is a promising new approach that is currently being tested in several clinical trials. The endogenous regulation of remyelination is highly dependent on the immune response. In this review article, we highlight the cell biology of remyelination and its regulation by innate immune cells. For the purpose of this review, we discuss the roles of microglia, and also astrocytes and oligodendrocyte progenitor cells (OPCs) as they are being increasingly recognized to have immune cell functions.
Collapse
Affiliation(s)
- Charbel S. Baaklini
- Department of Medicine, Division of Neurology, Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Khalil S. Rawji
- Wellcome Trust-Medical Research Council, Cambridge Stem Cell Institute, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Greg J. Duncan
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, United States
| | - Madelene F. S. Ho
- Department of Medicine, Division of Neurology, Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | | |
Collapse
|
220
|
Soysa R, Lampert S, Yuen S, Douglass AN, Li W, Pfeffer K, Crispe IN. Fetal origin confers radioresistance on liver macrophages via p21 cip1/WAF1. J Hepatol 2019; 71:553-562. [PMID: 31077791 DOI: 10.1016/j.jhep.2019.04.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND & AIMS Cells of hematopoietic origin, including macrophages, are generally radiation sensitive, but a subset of Kupffer cells (KCs) is relatively radioresistant. Here, we focused on the identity of the radioresistant KCs in unmanipulated mice and the mechanism of radioresistance. METHODS We employed Emr1- and inducible CX3Cr1-based fate-mapping strategies combined with the RiboTag reporter to identify the total KCs and the embryo-derived KCs, respectively. The KC compartment was reconstituted with adult bone-marrow-derived KCs (bm-KCs) using clodronate depletion. Mice were lethally irradiated and transplanted with donor bone marrow, and the radioresistance of bone-marrow- or embryo-derived KCs was studied. Gene expression was analyzed using in situ mRNA isolation via RiboTag reporter mice, and the translatomes were compared among subsets. RESULTS Here, we identified the radioresistant KCs as the long-lived subset that is derived from CX3CR1-expressing progenitor cells in fetal life, while adult bm-KCs do not resist irradiation. While both subsets upregulated the Cdkn1a gene, encoding p21-cip1/WAF1 protein, radioresistant embryo-derived KCs showed a greater increase in response to irradiation. In the absence of this molecule, the radioresistance of KCs was compromised. Replacement KCs, derived from adult hematopoietic stem cells, differed from radioresistant KCs in their expression of genes related to immunity and phagocytosis. CONCLUSIONS Here, we show that, in the murine liver, a subset of KCs of embryonic origin resists lethal irradiation through Cdkn1a upregulation and is maintained for a long period, while bm-KCs do not survive lethal irradiation. LAY SUMMARY Kupffer cells (KCs) are the tissue-resident macrophages of the liver. KCs can be originated from fetal precursors and from monocytes during the fetal stage and post-birth, respectively. Most immune cells in mice are sensitive to lethal-irradiation-induced death, while a subset of KCs resists radiation-induced death. These radioresistant KCs continue to live in the irradiated mice. We discovered that this relatively radioresistant KC subset are the fetal-derived KCs, and they achieve this through cell-cycle arrest. Understanding the radiobiology of KCs will provide valuable insights into the mechanisms that elicit radiation-induced liver disease.
Collapse
Affiliation(s)
- Radika Soysa
- Department of Global Health, University of Washington, Seattle, WA, USA.
| | - Sarah Lampert
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Sebastian Yuen
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Alyse N Douglass
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Wanyu Li
- Department of Hepatology, First Hospital of Jilin University, Changchun, China
| | - Klaus Pfeffer
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich Heine University, Düsseldorf, Germany
| | - Ian N Crispe
- Department of Pathology, University of Washington, Seattle, WA, USA; Department of Immunology, University of Washington, Seattle, WA, USA
| |
Collapse
|
221
|
Activation of microglia in acute hippocampal slices affects activity-dependent long-term potentiation and synaptic tagging and capture in area CA1. Neurobiol Learn Mem 2019; 163:107039. [DOI: 10.1016/j.nlm.2019.107039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/26/2019] [Accepted: 07/02/2019] [Indexed: 12/11/2022]
|
222
|
Song C, Phuengkham H, Kim YS, Dinh VV, Lee I, Shin IW, Shin HS, Jin SM, Um SH, Lee H, Hong KS, Jin SM, Lee E, Kang TH, Park YM, Lim YT. Syringeable immunotherapeutic nanogel reshapes tumor microenvironment and prevents tumor metastasis and recurrence. Nat Commun 2019; 10:3745. [PMID: 31431623 PMCID: PMC6702226 DOI: 10.1038/s41467-019-11730-8] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 08/01/2019] [Indexed: 12/21/2022] Open
Abstract
The low response rate of current cancer immunotherapy suggests the presence of few antigen-specific T cells and a high number of immunosuppressive factors in tumor microenvironment (TME). Here, we develop a syringeable immunomodulatory multidomain nanogel (iGel) that overcomes the limitation by reprogramming of the pro-tumoral TME to antitumoral immune niches. Local and extended release of immunomodulatory drugs from iGel deplete immunosuppressive cells, while inducing immunogenic cell death and increased immunogenicity. When iGel is applied as a local postsurgical treatment, both systemic antitumor immunity and a memory T cell response are generated, and the recurrence and metastasis of tumors to lungs and other organs are significantly inhibited. Reshaping of the TME using iGel also reverts non-responding groups to checkpoint blockade therapies into responding groups. The iGel is expected as an immunotherapeutic platform that can reshape immunosuppressive TMEs and synergize cancer immunotherapy with checkpoint therapies, with minimized systemic toxicity. The limited efficacy of current immunotherapy suggests low antigen-specific T cells and immunosuppressive factors in tumor microenvironment (TME). Here, the authors develop a syringeable immunomodulatory multi-domain nanogel that can reprogram the TME and induce enhanced cancer immunotherapy.
Collapse
|
223
|
Replenishment of Organotypic Hippocampal Slice Cultures with Neonatal or Adult Microglia. Methods Mol Biol 2019. [PMID: 31392682 DOI: 10.1007/978-1-4939-9658-2_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
This protocol describes a method to deplete and repopulate organotypic hippocampal slice cultures with ramified microglia. We describe the slice culture preparation from newborn mice, standard culturing of neonatal microglia, and the acute isolation of microglia from adult mouse brain. Furthermore, we outline the technique for the replenishment of microglia-depleted slice cultures with different microglia populations and subsequent morphological analysis. We show that neonatal and adult microglia acquire specific ramified morphologies, which in case of adult microglia are indistinguishable from the in vivo situation. This procedure not only allows the functional investigation of microglia with different degrees of ramification but also enables the construction of chimeric slice cultures with respect to the microglia phenotype. Preparation of slice cultures can be completed in 3.5 h, preparation of mixed-glial cultures in 4 h, isolation of adult microglia can be accomplished in 3.5 h, and replenishment in 30 min.
Collapse
|
224
|
The NDV-3A vaccine protects mice from multidrug resistant Candida auris infection. PLoS Pathog 2019; 15:e1007460. [PMID: 31381597 PMCID: PMC6695204 DOI: 10.1371/journal.ppat.1007460] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 08/15/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022] Open
Abstract
Candida auris is an emerging, multi-drug resistant, health care-associated fungal pathogen. Its predominant prevalence in hospitals and nursing homes indicates its ability to adhere to and colonize the skin, or persist in an environment outside the host—a trait unique from other Candida species. Besides being associated globally with life-threatening disseminated infections, C. auris also poses significant clinical challenges due to its ability to adhere to polymeric surfaces and form highly drug-resistant biofilms. Here, we performed bioinformatic studies to identify the presence of adhesin proteins in C. auris, with sequence as well as 3-D structural homologies to the major adhesin/invasin of C. albicans, Als3. Anti-Als3p antibodies generated by vaccinating mice with NDV-3A (a vaccine based on the N-terminus of Als3 protein formulated with alum) recognized C. auris in vitro, blocked its ability to form biofilms and enhanced macrophage-mediated killing of the fungus. Furthermore, NDV-3A vaccination induced significant levels of C. auris cross-reactive humoral and cellular immune responses, and protected immunosuppressed mice from lethal C. auris disseminated infection, compared to the control alum-vaccinated mice. The mechanism of protection is attributed to anti-Als3p antibodies and CD4+ T helper cells activating tissue macrophages. Finally, NDV-3A potentiated the protective efficacy of the antifungal drug micafungin, against C. auris candidemia. Identification of Als3-like adhesins in C. auris makes it a target for immunotherapeutic strategies using NDV-3A, a vaccine with known efficacy against other Candida species and safety as well as efficacy in clinical trials. Considering that C. auris can be resistant to almost all classes of antifungal drugs, such an approach has profound clinical relevance. Candida auris has emerged as a major health concern to hospitalized patients and nursing home subjects. C. auris strains display multidrug resistance to current antifungal therapy and cause lethal infections. We have determined that C. auris harbors homologs of C. albicans Als cell surface proteins. The C. albicans NDV-3A vaccine, harboring the N-terminus of Als3p formulated with alum, generates cross-reactive antibodies against C. auris clinical isolates and protects neutropenic mice from hematogenously disseminated C. auris infection. Importantly, the NDV-3A vaccine displays an additive protective effect in neutropenic mice when combined with micafungin. Due to its proven safety and efficacy in humans against C. albicans infection, our studies support the expedited testing of the NDV-3A vaccine against C. auris in future clinical trials.
Collapse
|
225
|
Phuengkham H, Ren L, Shin IW, Lim YT. Nanoengineered Immune Niches for Reprogramming the Immunosuppressive Tumor Microenvironment and Enhancing Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1803322. [PMID: 30773696 DOI: 10.1002/adma.201803322] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 01/03/2019] [Indexed: 06/09/2023]
Abstract
Cancer immunotherapies that harness the body's immune system to combat tumors have received extensive attention and become mainstream strategies for treating cancer. Despite promising results, some problems remain, such as the limited patient response rate and the emergence of severe immune-related adverse effects. For most patients, the therapeutic efficacy of cancer immunotherapy is mainly limited by the immunosuppressive tumor microenvironment (TME). To overcome such obstacles in the TME, the immunomodulation of immunosuppressive factors and therapeutic immune cells (e.g., T cells and antigen-presenting cells) should be carefully designed and evaluated. Nanoengineered synthetic immune niches have emerged as highly customizable platforms with a potent capability for reprogramming the immunosuppressive TME. Here, recent developments in nano-biomaterials that are rationally designed to modulate the immunosuppressive TME in a spatiotemporal manner for enhanced cancer immunotherapy which are rationally designed to modulate the immunosuppressive TME in a spatiotemporal manner for enhanced cancer immunotherapy are highlighted.
Collapse
Affiliation(s)
- Hathaichanok Phuengkham
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, and School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Long Ren
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, and School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Il Woo Shin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, and School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, and School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| |
Collapse
|
226
|
Accolla RS, Ramia E, Tedeschi A, Forlani G. CIITA-Driven MHC Class II Expressing Tumor Cells as Antigen Presenting Cell Performers: Toward the Construction of an Optimal Anti-tumor Vaccine. Front Immunol 2019; 10:1806. [PMID: 31417570 PMCID: PMC6682709 DOI: 10.3389/fimmu.2019.01806] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 07/17/2019] [Indexed: 12/11/2022] Open
Abstract
Construction of an optimal vaccine against tumors relies on the availability of appropriate tumor-specific antigens capable to stimulate CD4+ T helper cells (TH) and CD8+ cytolytic T cells (CTL). CTL are considered the major effectors of the anti-tumor adaptive immune response as they recognize antigens presented on MHC class I (MHC-I) molecules usually expressed in all cells and thus also in tumors. However, attempts to translate in clinics vaccination protocols based only on tumor-specific MHC-I-bound peptides have resulted in very limited, if any, success. We believe failure was mostly due to inadequate triggering of the TH arm of adaptive immunity, as TH cells are necessary to trigger and maintain the proliferation of all the immune effector cells required to eliminate tumor cells. In this review, we focus on a novel strategy of anti-tumor vaccination established in our laboratory and based on the persistent expression of MHC class II (MHC-II) molecules in tumor cells. MHC-II are the restricting elements of TH recognition. They are usually not expressed in solid tumors. By genetically modifying tumor cells of distinct histological origin with the MHC-II transactivator CIITA, the physiological controller of MHC-II gene expression discovered in our laboratory, stable expression of all MHC class II genes was obtained. This resulted in tumor rejection or strong retardation of tumor growth in vivo in mice, mediated primarily by tumor-specific TH cells as assessed by both depletion and adoptive cell transfer experiments. Importantly these findings led us to apply this methodology to human settings for the purification of MHC-II-bound tumor specific peptides directly from tumor cells, specifically from hepatocarcinomas, and the construction of a multi-peptide (MHC-II and MHC-I specific) immunotherapeutic vaccine. Additionally, our approach unveiled a noticeable exception to the dogma that dendritic cells are the sole professional antigen presenting cells (APC) capable to prime naïve TH cells, because CIITA-dependent MHC-II expressing tumor cells could also perform this function. Thus, our approach has served not only to select the most appropriate tumor specific peptides to activate the key lymphocytes triggering the anti-tumor effector functions but also to increase our knowledge of intimate mechanisms governing basic immunological processes.
Collapse
Affiliation(s)
- Roberto S Accolla
- Laboratories of General Pathology and Immunology "Giovanna Tosi", Department of Medicine and Surgery, School of Medicine, University of Insubria, Varese, Italy
| | - Elise Ramia
- Laboratories of General Pathology and Immunology "Giovanna Tosi", Department of Medicine and Surgery, School of Medicine, University of Insubria, Varese, Italy
| | - Alessandra Tedeschi
- Laboratories of General Pathology and Immunology "Giovanna Tosi", Department of Medicine and Surgery, School of Medicine, University of Insubria, Varese, Italy
| | - Greta Forlani
- Laboratories of General Pathology and Immunology "Giovanna Tosi", Department of Medicine and Surgery, School of Medicine, University of Insubria, Varese, Italy
| |
Collapse
|
227
|
Bai H, Wen J, Gong JP, Wu H, Yuan FC, Cao D, Wu YK, Lai X, Wang MH. Blockade of the Notch1/Jagged1 pathway in Kupffer cells aggravates ischemia-reperfusion injury of orthotopic liver transplantation in mice. Autoimmunity 2019; 52:176-184. [PMID: 31322442 DOI: 10.1080/08916934.2019.1637424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Liver ischemia-reperfusion injury (IRI) represents a risk factor for early graft dysfunction and an obstacle to expanding donor pool in orthotopic liver transplantation (OLT). Kupffer cells (KCs) are the largest antigen-presenting cell (APC) group and the primary modulators of inflammation in liver tissues. The vital role of Notch1/Jagged1 pathway in mouse OLT model has been reported, however, its potential therapeutic mechanism is unknown. Here, we made use of short hairpin RNA-Jagged1 and AAV-Jagged1 to explore the effects of Notch1/Jagged1 pathway in OLT. In vitro, blockade of Notch1/Jagged1 pathway downregulated the expression of Hairy and enhancer of split-1 (Hes1) gene, which in turn increased the proinflammatory effects of KCs. Moreover, the anti-inflammatory effects of Notch1/Jagged1 pathway were induced by inhibiting Hes1/gene of phosphate and tension/protein kinase B/Toll-like receptor 4/nuclear factor kappa B (Hes1/PTEN/AKT/TLR4/NF-κB) axis in KCs. In vivo, we used a well-established mouse model of OLT to mimic clinical transplantation. Mice were stochastically divided into 6 groups: Sham group (n = 15); Normal saline (NS) group (n = 15); Adeno-associated virus-green fluorescent protein (AAV-GFP) group (n = 15); AAV-Jagged1 group (n = 15); Clodronate liposome (CL) group (n = 15); CL+AAV-Jagged1 group (n = 15) . After OLT the liver damage in AAV-Jagged1 group were significantly accentuated compared to the AAV-GFP group. While blockade of Jagged1 aftet clearence of KCs by CL would not lead to further liver injuries. Taken together, our study demonstrated that blockade of Notch1/Jagged1 pathway aggravates inflammation induced by lipopolysaccharide (LPS) via Hes1/PTEN/AKT/TLR4/NF-κB in KCs, and the blockade of Notch1/Jagged1 pathway in donor liver increased neutrophil/macrophage infiltration and hepatocellular apoptosis, which suggested the function of Notch1/Jagged1 pathway in mouse OLT and highlighted the protective function of Notch1/Jagged1 pathway in liver transplantation.
Collapse
Affiliation(s)
- He Bai
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Jian Wen
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Jian-Ping Gong
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Hao Wu
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Fang-Chao Yuan
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Ding Cao
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Ya-Kun Wu
- Department of Hepatobiliary Surgery, Suining Central Hospital , Sichuan , People's Republic of China
| | - Xing Lai
- Department of Hepatobiliary and Thyroid Breast Surgery, Tongnan District People's Hospital , Chongqing , People's Republic of China
| | - Meng-Hao Wang
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| |
Collapse
|
228
|
Lai C, Li C, Liu M, Qiu Q, Luo X, Liu X, Hu L, Deng Y, Song Y. Effect of Kupffer cells depletion on ABC phenomenon induced by Kupffer cells-targeted liposomes. Asian J Pharm Sci 2019; 14:455-464. [PMID: 32104474 PMCID: PMC7032111 DOI: 10.1016/j.ajps.2018.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/02/2018] [Accepted: 07/20/2018] [Indexed: 12/15/2022] Open
Abstract
Accelerated blood clearance (ABC) phenomenon is common in many PEGylated nanocarriers, whose mechanism has not been completely elucidated yet. In this study, the correlation between Kupffer cells (KCs) and ABC phenomenon has been studied by KCs-targeted liposomes inducing ABC phenomenon and KCs depletion. In other words, the 4-aminophenyl-α-D-mannopyranoside (APM) lipid derivative DSPE-PEG2000-APM (DPM), and 4-aminophenyl-β-L-fucopyranoside (APF) lipid derivative DSPE-PEG2000-APF (DPF) were conjugated and modified on alendronate sodium (AD) liposomes to specifically target and deplete KCs. The dual-ligand modified PEGylated liposomes (MFPL) showed stronger ability to damage KCs in vitro and in vivo, which also could indirectly illustrate that dual-ligand modification could better target KCs. Besides, the hepatic biodistribution and pharmacokinetics could directly prove that MFPL had a stronger targeting ability to KCs. In addition, in depletion rats, plasma concentration and splenic biodistribution of MFPL and PEGylated liposomes (PL) were significantly elevated and hepatic biodistribution was significantly reduced, which demonstrated that KCs played an important role on elimination of nanoparticles. What's more, ABC phenomenon of the secondary injection of PL was stronger in KCs depletion rats than that in normal rats, which indicated that depletion of KCs prolonged the circulation of PL in the first injection repeatedly stimulating B-cells in the marginal region of the spleen and causing it to secrete more IgM antibodies. This could also illustrate that anti-PEG IgM takes up a major station compared with KCs. Most important of all, KCs-targeted liposomes could induce a stronger ABC phenomenon than PL in normal rats, which declared that based on the same IgM concentration, the more the KCs were stimulated, the stronger ABC phenomenon was induced. However, in depletion rats, this difference of ABC phenomenon between PL and MFPL could no more exist, further demonstrating that KCs could participate and play a certain role in the ABC phenomenon.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yihui Deng
- Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yanzhi Song
- Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
229
|
The YIN and YANG of lipoproteins in developing and preventing infectious arthritis by Staphylococcus aureus. PLoS Pathog 2019; 15:e1007877. [PMID: 31226163 PMCID: PMC6608979 DOI: 10.1371/journal.ppat.1007877] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 07/03/2019] [Accepted: 05/28/2019] [Indexed: 12/21/2022] Open
Abstract
Rapid bone destruction often leads to permanent joint dysfunction in patients with septic arthritis, which is mainly caused by Staphylococcus aureus (S. aureus). Staphylococcal cell wall components are known to induce joint inflammation and bone destruction. Here, we show that a single intra-articular injection of S. aureus lipoproteins (Lpps) into mouse knee joints induced chronic destructive macroscopic arthritis through TLR2. Arthritis was characterized by rapid infiltration of neutrophils and monocytes. The arthritogenic effect was mediated mainly by macrophages/monocytes and partially via TNF-α but not by neutrophils. Surprisingly, a S. aureus mutant lacking Lpp diacylglyceryl transferase (lgt) caused more severe joint inflammation, which coincided with higher bacterial loads of the lgt mutant in local joints than those of its parental strain. Coinjection of pathogenic S. aureus LS-1 with staphylococcal Lpps into mouse knee joints caused improved bacterial elimination and diminished bone erosion. The protective effect of the Lpps was mediated by their lipid moiety and was fully dependent on TLR2 and neutrophils. The blocking of CXCR2 on neutrophils resulted in total abrogation of the protective effect of the Lpps. Our data demonstrate that S. aureus Lpps elicit innate immune responses, resulting in a double-edged effect. On the one hand, staphylococcal Lpps boost septic arthritis. On the other hand, Lpps act as adjuvants and activate innate immunity, which could be useful for combating infections with multiple drug-resistant strains. Rapid bone destruction often leads to permanent joint dysfunction in septic arthritis, which is mainly caused by S. aureus. Despite advances in the use of antibiotics, permanent reductions in joint function occur in up to 50% of patients, who may also need joint replacement surgery. Additional challenge is posed by increasing antibiotic resistance of S. aureus, causing significant clinical and economic consequences. Although the outcome is poor, the current treatments for septic arthritis remain unchanged since many decades. It is largely unknown which bacterial factors cause aggressive joint damage. Here, we show that a single intra-articular injection of S. aureus lipoproteins (Lpps) into mouse knee joints induced chronic destructive macroscopic arthritis, and the monocytes/macrophages were the main culprit. However, coinjection of pathogenic S. aureus with Lpps into mouse knee joints attenuated the disease. The protective effect of Lpps was mediated by their lipid moiety, TLR2 on the host cells, neutrophil chemokine release, and consequent neutrophil recruitment. Our finding might be used as a novel concept in the treatment of multidrug-resistant bacterial infections.
Collapse
|
230
|
Evans TA, Barkauskas DS, Silver J. Intravital imaging of immune cells and their interactions with other cell types in the spinal cord: Experiments with multicolored moving cells. Exp Neurol 2019; 320:112972. [PMID: 31234058 DOI: 10.1016/j.expneurol.2019.112972] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/25/2019] [Accepted: 06/04/2019] [Indexed: 12/25/2022]
Abstract
Intravital imaging of the immune system is a powerful technique for studying biology of the immune response in the spinal cord using a variety of disease models ranging from traumatic injury to autoimmune disorders. Here, we will discuss specific technical aspects as well as many intriguing biological phenomena that have been revealed with the use of intravital imaging for investigation of the immune system in the spinal cord. We will discuss surgical techniques for exposing and stabilizing the spine that are critical for obtaining images, visualizing immune and CNS cells with genetically expressed fluorescent proteins, fluorescent labeling techniques and briefly discuss some of the challenges of image analysis.
Collapse
Affiliation(s)
- Teresa A Evans
- Department of Pediatrics, Stanford University, Stanford, CA, USA.
| | | | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
231
|
Baert T, Vankerckhoven A, Riva M, Van Hoylandt A, Thirion G, Holger G, Mathivet T, Vergote I, Coosemans A. Myeloid Derived Suppressor Cells: Key Drivers of Immunosuppression in Ovarian Cancer. Front Immunol 2019; 10:1273. [PMID: 31214202 PMCID: PMC6558014 DOI: 10.3389/fimmu.2019.01273] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 05/20/2019] [Indexed: 12/27/2022] Open
Abstract
The presence of tumor infiltrating lymphocytes (TILs) is associated with a longer overall survival in advanced stage epithelial ovarian cancer. Despite the prognostic impact of TILs, response to checkpoint-inhibitors and antigen-specific active immunotherapy is limited in ovarian cancer. The goal of our study was to investigate the interaction between ovarian cancer and the innate and adaptive immune system in the ID8-fLuc syngeneic ovarian cancer mouse model. For the in vivo experiments C57BL/6, B6.129S7-Rag1tm1Mom/J, and B6.129P2(SJL)-Myd88tm1.1Defr/J mice were inoculated with ID8-fLuc. In vivo depletion experiments were performed using clodronate liposomes (CL), anti-CD8a, anti-GR1, anti-colony stimulating factor 1 (anti-CSF1), and TMβ1 (anti-CD122). Immune read out was performed by fluorescent activated cell sorting analysis for effector T cells, regulatory T cells, natural killer cells, B cells, macrophages, and myeloid derived suppressor cells (MDSC), immunohistochemistry for MDSC and tumor-associated macrophages (TAM) and immunofluorescence for M1 and M2 TAM in the vascular context. The effect of MDSC on T cell proliferation and phenotype were studied in vitro. We discovered that the absence of T and B cells did not influence tumor growth or survival of B6.129S7-Rag1tm1Mom/J mice compared to immunocompetent C57BL/6 mice. CL-induced macrophage depletion promoted tumor proliferation and shortened survival in C57BL/6 mice (p = 0.004) and in B6.129S7-Rag1tm1Mom/J mice (p = 0.0005). During CL treatment, we observed a clear increase of pro-inflammatory cytokines (p ≤ 0.02) and monocytic MDSC (p ≤ 0.01). Selective depletion of MDSC by anti-GR1 improved survival, certainly in comparison to mice treated with anti-CSF1 (p = 0.01-median survival 91 vs. 67.5 days). B6.129P2(SJL)-Myd88tm1.1Defr/J mice displayed to a longer median survival compared to C57BL/6 mice (90 vs. 76 days). MDSC activated by ID8-fLuc conditioned medium or ascites of tumor-bearing mice showed T cell suppressive functions in vitro. Based on these findings, we conclude that the adaptive immune system does not efficiently control tumor growth in the ID8-fLuc model. In addition, we discovered a prominent role for MDSC as the driver of immunosuppression in the ID8-fLuc ovarian cancer mouse model.
Collapse
Affiliation(s)
- Thaïs Baert
- ImmunOvar Research Group, Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, KU Leuven, Leuven, Belgium.,Department of Gynecology and Gynecologic Oncology, Kliniken Essen Mitte, Essen, Germany
| | - Ann Vankerckhoven
- ImmunOvar Research Group, Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Matteo Riva
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Anaïs Van Hoylandt
- ImmunOvar Research Group, Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Gitte Thirion
- ImmunOvar Research Group, Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Gerhardt Holger
- Vascular Patterning Lab, Center for Cancer Biology, VIB, KU Leuven, Leuven, Belgium
| | - Thomas Mathivet
- Vascular Patterning Lab, Center for Cancer Biology, VIB, KU Leuven, Leuven, Belgium.,PARCC, HEGP Institute (team 9), INSERM U970, Université Paris Descartes, Paris, France
| | - Ignace Vergote
- ImmunOvar Research Group, Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, KU Leuven, Leuven, Belgium.,Laboratory of Gynecologic Oncology, Department of Oncology, KU Leuven, Leuven, Belgium.,Department of Gynaecology and Obstetrics, Leuven Cancer Institute, University Hospitals Leuven (UZ Leuven), Leuven, Belgium
| | - An Coosemans
- ImmunOvar Research Group, Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, KU Leuven, Leuven, Belgium.,Department of Gynaecology and Obstetrics, Leuven Cancer Institute, University Hospitals Leuven (UZ Leuven), Leuven, Belgium
| |
Collapse
|
232
|
Marinkovic M, Fuoco C, Sacco F, Cerquone Perpetuini A, Giuliani G, Micarelli E, Pavlidou T, Petrilli LL, Reggio A, Riccio F, Spada F, Vumbaca S, Zuccotti A, Castagnoli L, Mann M, Gargioli C, Cesareni G. Fibro-adipogenic progenitors of dystrophic mice are insensitive to NOTCH regulation of adipogenesis. Life Sci Alliance 2019; 2:e201900437. [PMID: 31239312 PMCID: PMC6599969 DOI: 10.26508/lsa.201900437] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 12/14/2022] Open
Abstract
Fibro-adipogenic progenitors (FAPs) promote satellite cell differentiation in adult skeletal muscle regeneration. However, in pathological conditions, FAPs are responsible for fibrosis and fatty infiltrations. Here we show that the NOTCH pathway negatively modulates FAP differentiation both in vitro and in vivo. However, FAPs isolated from young dystrophin-deficient mdx mice are insensitive to this control mechanism. An unbiased mass spectrometry-based proteomic analysis of FAPs from muscles of wild-type and mdx mice suggested that the synergistic cooperation between NOTCH and inflammatory signals controls FAP differentiation. Remarkably, we demonstrated that factors released by hematopoietic cells restore the sensitivity to NOTCH adipogenic inhibition in mdx FAPs. These results offer a basis for rationalizing pathological ectopic fat infiltrations in skeletal muscle and may suggest new therapeutic strategies to mitigate the detrimental effects of fat depositions in muscles of dystrophic patients.
Collapse
Affiliation(s)
| | - Claudia Fuoco
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Francesca Sacco
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | | | - Giulio Giuliani
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Elisa Micarelli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | | | - Alessio Reggio
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Federica Riccio
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Filomena Spada
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Simone Vumbaca
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | - Luisa Castagnoli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Cesare Gargioli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Gianni Cesareni
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
233
|
Poon W, Zhang YN, Ouyang B, Kingston BR, Wu JLY, Wilhelm S, Chan WCW. Elimination Pathways of Nanoparticles. ACS NANO 2019; 13:5785-5798. [PMID: 30990673 DOI: 10.1021/acsnano.9b01383] [Citation(s) in RCA: 295] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Understanding how nanoparticles are eliminated from the body is required for their successful clinical translation. Many promising nanoparticle formulations for in vivo medical applications are large (>5.5 nm) and nonbiodegradable, so they cannot be eliminated renally. A proposed pathway for these nanoparticles is hepatobiliary elimination, but their transport has not been well-studied. Here, we explored the barriers that determined the elimination of nanoparticles through the hepatobiliary route. The route of hepatobiliary elimination is usually through the following pathway: (1) liver sinusoid, (2) space of Disse, (3) hepatocytes, (4) bile ducts, (5) intestines, and (6) out of the body. We discovered that the interaction of nanoparticles with liver nonparenchymal cells ( e. g., Kupffer cells and liver sinusoidal endothelial cells) determines the elimination fate. Each step in the route contains cells that can sequester and chemically or physically alter the nanoparticles, which influences their fecal elimination. We showed that the removal of Kupffer cells increased fecal elimination by >10 times. Combining our results with those of prior studies, we can start to build a systematic view of nanoparticle elimination pathways as it relates to particle size and other design parameters. This is critical to engineering medically useful and translatable nanotechnologies.
Collapse
Affiliation(s)
- Wilson Poon
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto , Ontario M5S 3G9 , Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research , University of Toronto , Toronto , Ontario M5S 3E1 , Canada
| | - Yi-Nan Zhang
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto , Ontario M5S 3G9 , Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research , University of Toronto , Toronto , Ontario M5S 3E1 , Canada
| | - Ben Ouyang
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto , Ontario M5S 3G9 , Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research , University of Toronto , Toronto , Ontario M5S 3E1 , Canada
- MD/PhD Program, Faculty of Medicine , University of Toronto , Toronto , Ontario M5S 1A8 , Canada
| | - Benjamin R Kingston
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto , Ontario M5S 3G9 , Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research , University of Toronto , Toronto , Ontario M5S 3E1 , Canada
| | - Jamie L Y Wu
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto , Ontario M5S 3G9 , Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research , University of Toronto , Toronto , Ontario M5S 3E1 , Canada
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering , University of Oklahoma , Norman , Oklahoma 73019 , United States
- Stephenson Cancer Center , Oklahoma City , Oklahoma 73104 , United States
| | - Warren C W Chan
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto , Ontario M5S 3G9 , Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research , University of Toronto , Toronto , Ontario M5S 3E1 , Canada
- Department of Chemical Engineering and Applied Chemistry , University of Toronto , Toronto , Ontario M5S 3E5 , Canada
- Department of Materials Science and Engineering , University of Toronto , Toronto , Ontario M5S 1A1 , Canada
- Department of Chemistry , University of Toronto , Toronto , Ontario M5S 3H6 , Canada
| |
Collapse
|
234
|
de Kruijff RM, Raavé R, Kip A, Molkenboer-Kuenen J, Roobol SJ, Essers J, Heskamp S, Denkova AG. Elucidating the Influence of Tumor Presence on the Polymersome Circulation Time in Mice. Pharmaceutics 2019; 11:E241. [PMID: 31137479 PMCID: PMC6572275 DOI: 10.3390/pharmaceutics11050241] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/14/2019] [Accepted: 05/16/2019] [Indexed: 11/16/2022] Open
Abstract
The use of nanoparticles as tumor-targeting agents is steadily increasing, and the influence of nanoparticle characteristics such as size and stealthiness have been established for a large number of nanocarrier systems. However, not much is known about the impact of tumor presence on nanocarrier circulation times. This paper reports on the influence of tumor presence on the in vivo circulation time and biodistribution of polybutadiene-polyethylene oxide (PBd-PEO) polymersomes. For this purpose, polymersomes were loaded with the gamma-emitter 111In and administered intravenously, followed by timed ex vivo biodistribution. A large reduction in circulation time was observed for tumor-bearing mice, with a circulation half-life of merely 5 min (R2 = 0.98) vs 117 min (R2 = 0.95) in healthy mice. To determine whether the rapid polymersome clearance observed in tumor-bearing mice was mediated by macrophages, chlodronate liposomes were administered to both healthy and tumor-bearing mice prior to the intravenous injection of radiolabeled polymersomes to deplete their macrophages. Pretreatment with chlodronate liposomes depleted macrophages in the spleen and liver and restored the circulation time of the polymersomes with no significant difference in circulation time between healthy mice and tumor-bearing mice pretreated with clodronate liposomes (15.2 ± 1.2% ID/g and 13.6 ± 2.7% ID/g, respectively, at 4 h p.i. with p = 0.3). This indicates that activation of macrophages due to tumor presence indeed affected polymersome clearance rate. Thus, next to particle design, the presence of a tumor can also greatly impact circulation times and should be taken into account when designing studies to evaluate the distribution of polymersomes.
Collapse
Affiliation(s)
- Robin M de Kruijff
- Radiation Science and Technology, Delft University of Technology, 2629 JB Delft, The Netherlands.
| | - René Raavé
- Radiology and Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.
| | - Annemarie Kip
- Radiology and Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.
| | - Janneke Molkenboer-Kuenen
- Radiology and Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.
| | - Stefan J Roobol
- Molecular Genetics, Oncode Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands.
- Radiology and Nuclear Medicine, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands.
| | - Jeroen Essers
- Molecular Genetics, Oncode Institute, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands.
| | - Sandra Heskamp
- Radiology and Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands.
| | - Antonia G Denkova
- Radiation Science and Technology, Delft University of Technology, 2629 JB Delft, The Netherlands.
| |
Collapse
|
235
|
Modifying Graft-versus-Host Disease in a Humanized Mouse Model by Targeting Macrophages or B-Cells. J Immunol Res 2019; 2019:3538963. [PMID: 31205954 PMCID: PMC6530157 DOI: 10.1155/2019/3538963] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/19/2019] [Accepted: 04/02/2019] [Indexed: 01/03/2023] Open
Abstract
Humanized mouse models can well be modified to study specific aspects of Graft-versus-Host Disease (GvHD). This paper shows the results of both macrophage depletion and (early) B-cell depletion in a humanized mouse model using RAG2-/- γc-/- mice injected with HuPBMCs. Macrophage depletion showed a significant decrease in survival and also lead to a change in the histomorphology of the xenogeneic reaction. Higher levels of infiltrating B-cells were observed in various organs of mice depleted for macrophages. With (early) B-cell depletion using Rituximab, a clear improvement on clinical symptoms was observed, even when probably only inactivated B-cells were deleted. However, the histological examinations only showed a significant morphological effect on liver fibrosis. This may be related to a difference in the mRNA levels of TGF-β. Also, lower mRNA levels of Tregs in some organs were observed after Rituximab treatment, which contradicts that a higher number of Tregs would always be related to less severe GvHD. Our data show that both macrophage depletion and (early) B-cell depletion in a xenogeneic mouse model can influence the clinical, histological, and cytokine production of a GvHD response.
Collapse
|
236
|
Pedragosa M, Riera G, Casella V, Esteve-Codina A, Steuerman Y, Seth C, Bocharov G, Heath S, Gat-Viks I, Argilaguet J, Meyerhans A. Linking Cell Dynamics With Gene Coexpression Networks to Characterize Key Events in Chronic Virus Infections. Front Immunol 2019; 10:1002. [PMID: 31130969 PMCID: PMC6509617 DOI: 10.3389/fimmu.2019.01002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/18/2019] [Indexed: 01/29/2023] Open
Abstract
The host immune response against infection requires the coordinated action of many diverse cell subsets that dynamically adapt to a pathogen threat. Due to the complexity of such a response, most immunological studies have focused on a few genes, proteins, or cell types. With the development of “omic”-technologies and computational analysis methods, attempts to analyze and understand complex system dynamics are now feasible. However, the decomposition of transcriptomic data sets generated from complete organs remains a major challenge. Here, we combined Weighted Gene Coexpression Network Analysis (WGCNA) and Digital Cell Quantifier (DCQ) to analyze time-resolved mouse splenic transcriptomes in acute and chronic Lymphocytic Choriomeningitis Virus (LCMV) infections. This enabled us to generate hypotheses about complex immune functioning after a virus-induced perturbation. This strategy was validated by successfully predicting several known immune phenomena, such as effector cytotoxic T lymphocyte (CTL) expansion and exhaustion. Furthermore, we predicted and subsequently verified experimentally macrophage-CD8 T cell cooperativity and the participation of virus-specific CD8+ T cells with an early effector transcriptome profile in the host adaptation to chronic infection. Thus, the linking of gene expression changes with immune cell kinetics provides novel insights into the complex immune processes within infected tissues.
Collapse
Affiliation(s)
- Mireia Pedragosa
- Infection Biology Laboratory, Department of Experimental and Health Sciences (DCEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Graciela Riera
- Infection Biology Laboratory, Department of Experimental and Health Sciences (DCEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Valentina Casella
- Infection Biology Laboratory, Department of Experimental and Health Sciences (DCEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Center for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Yael Steuerman
- Cell Research and Immunology Department, Tel Aviv University, Tel Aviv, Israel
| | - Celina Seth
- Infection Biology Laboratory, Department of Experimental and Health Sciences (DCEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Gennady Bocharov
- Marchuk Institute of Numerical Mathematics, Russian Academy of Sciences, Moscow, Russia.,Institute for Personalized Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Simon Heath
- CNAG-CRG, Center for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Irit Gat-Viks
- Cell Research and Immunology Department, Tel Aviv University, Tel Aviv, Israel
| | - Jordi Argilaguet
- Infection Biology Laboratory, Department of Experimental and Health Sciences (DCEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Andreas Meyerhans
- Infection Biology Laboratory, Department of Experimental and Health Sciences (DCEXS), Universitat Pompeu Fabra, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
237
|
Santamaria-Barria JA, Zeng S, Greer JB, Beckman MJ, Seifert AM, Cohen NA, Zhang JQ, Crawley MH, Green BL, Loo JK, Maltbaek JH, DeMatteo RP. Csf1r or Mer inhibition delays liver regeneration via suppression of Kupffer cells. PLoS One 2019; 14:e0216275. [PMID: 31042769 PMCID: PMC6493758 DOI: 10.1371/journal.pone.0216275] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/17/2019] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Murine Kupffer cells (KCs) comprise CD11bhi and F4/80hi subsets. Tissue-resident macrophages are known to express the tyrosine kinase receptors colony-stimulating factor 1 receptor (Csf1r) and Mer. However, the expression of Csf1r and Mer on KC subsets and the importance of these tyrosine kinases during liver regeneration (LR) are unknown. METHODS KCs from wild-type and Csf1r-GFP mice were characterized by flow cytometry. Partial hepatectomy (PH) was performed in mice treated with clodronate liposomes, a Csf1r small molecule inhibitor or depleting antibody, or a small molecule Mer inhibitor. Sera and livers were analyzed. The function of sorted KC subsets was tested in vitro. RESULTS Mer was specifically expressed on tissue-resident F4/80hi KCs, 55% of which also expressed Csf1r. Mer+Csf1r+ and Mer+Csf1r- KCs had distinct expression of macrophage markers. Csf1r inhibition in mice reduced F4/80hi KCs by approximately 50%, but did not affect CD11bhi KCs. Clodronate liposomes depleted F4/80hi KCs, but also altered levels of other intrahepatic leukocytes. Csf1r inhibition delayed LR, as demonstrated by a 20% reduction in liver-to-body weight ratios 7 days after PH. At 36h after PH, Csf1r inhibition increased serum ALT and histological liver injury, and decreased liver cell proliferation. A small molecule inhibitor of Mer did not alter the percentage of KCs or their proliferation and just modestly delayed LR. In vitro, Csf1r or Mer inhibition did not decrease KC viability, but did attenuate their cytokine response to stimulation. CONCLUSIONS F4/80hi KCs are Mer+ and can be subdivided based on Csf1r expression. Csf1r or Mer inhibition each reduces KC cytokine production and delays LR.
Collapse
Affiliation(s)
- Juan A. Santamaria-Barria
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Shan Zeng
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Jonathan B. Greer
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Michael J. Beckman
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Adrian M. Seifert
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Noah A. Cohen
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Jennifer Q. Zhang
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Megan H. Crawley
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Benjamin L. Green
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Jennifer K. Loo
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Joanna H. Maltbaek
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Ronald P. DeMatteo
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| |
Collapse
|
238
|
Challa DK, Wang X, Montoyo HP, Velmurugan R, Ober RJ, Ward ES. Neonatal Fc receptor expression in macrophages is indispensable for IgG homeostasis. MAbs 2019; 11:848-860. [PMID: 30964743 PMCID: PMC6601554 DOI: 10.1080/19420862.2019.1602459] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The maintenance of the homeostasis of immunoglobulin G (IgG) represents a fundamental aspect of humoral immunity that has direct relevance to the successful delivery of antibody-based therapeutics. The ubiquitously expressed neonatal Fc receptor (FcRn) salvages IgG from cellular degradation following pinocytic uptake into cells, conferring prolonged in vivo persistence on IgG. However, the cellular sites of FcRn function are poorly defined. Pinocytic uptake is a prerequisite for FcRn-mediated IgG salvage, prompting us to investigate the consequences of IgG uptake and catabolism by macrophages, which represent both abundant and highly pinocytic cells in the body. Site-specific deletion of FcRn to generate mice harboring FcRn-deficient macrophages results in IgG hypercatabolism and ~threefold reductions in serum IgG levels, whereas these effects were not observed in mice that lack functional FcRn in B cells and dendritic cells. Consistent with the degradative activity of FcRn-deficient macrophages, depletion of these cells in FcRn-deficient mice leads to increased persistence and serum levels of IgG. These studies demonstrate a pivotal role for FcRn-mediated salvage in compensating for the high pinocytic and degradative activities of macrophages to maintain IgG homeostasis.
Collapse
Affiliation(s)
- Dilip K Challa
- a Department of Molecular and Cellular Medicine , Texas A&M University Health Science Center , College Station , TX , USA
| | - Xiaoli Wang
- a Department of Molecular and Cellular Medicine , Texas A&M University Health Science Center , College Station , TX , USA
| | - Héctor Pérez Montoyo
- b Department of Immunology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Ramraj Velmurugan
- a Department of Molecular and Cellular Medicine , Texas A&M University Health Science Center , College Station , TX , USA
| | - Raimund J Ober
- a Department of Molecular and Cellular Medicine , Texas A&M University Health Science Center , College Station , TX , USA.,c Department of Biomedical Engineering , Texas A&M University , College Station , TX , USA.,d Cancer Sciences Unit, Centre for Cancer Immunology, Faculty of Medicine , University of Southampton , Southampton , UK
| | - E Sally Ward
- a Department of Molecular and Cellular Medicine , Texas A&M University Health Science Center , College Station , TX , USA.,d Cancer Sciences Unit, Centre for Cancer Immunology, Faculty of Medicine , University of Southampton , Southampton , UK.,e Department of Microbial Pathogenesis and Immunology , Texas A&M University Health Science Center , Bryan , TX , USA
| |
Collapse
|
239
|
Venosa A, Katzen J, Tomer Y, Kopp M, Jamil S, Russo SJ, Mulugeta S, Beers MF. Epithelial Expression of an Interstitial Lung Disease-Associated Mutation in Surfactant Protein-C Modulates Recruitment and Activation of Key Myeloid Cell Populations in Mice. THE JOURNAL OF IMMUNOLOGY 2019; 202:2760-2771. [PMID: 30910861 DOI: 10.4049/jimmunol.1900039] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/04/2019] [Indexed: 12/24/2022]
Abstract
Patients with idiopathic pulmonary fibrosis (IPF) often experience precipitous deteriorations, termed "acute exacerbations" (AE), marked by diffuse alveolitis and altered gas exchange, resulting in a significant loss of lung function or mortality. The missense isoleucine to threonine substitution at position 73 (I73T) in the alveolar type 2 cell-restricted surfactant protein-C (SP-C) gene (SFTPC) has been linked to clinical IPF. To better understand the sequence of events that impact AE-IPF, we leveraged a murine model of inducible SP-CI73T (SP-CI73T/I73TFlp+/- ) expression. Following administration of tamoxifen to 8-12-wk-old mice, an upregulation of SftpcI73T initiated a diffuse lung injury marked by increases in bronchoalveolar lavage fluid (BALF) protein and histochemical evidence of CD45+ and CD11b+ cell infiltrates. Flow cytometry of collagenase-digested lung cells revealed a transient, early reduction in SiglecFhiCD11blowCD64hiCD11chi macrophages, countered by the sequential accumulation of SiglecFloCD11b+CD64-CD11c-CCR2+Ly6C+ immature macrophages (3 d), Ly6G+ neutrophils (7 d), and SiglecFhiCD11bhiCD11clo eosinophils (2 wk). By mRNA analysis, BALF cells demonstrated a time-dependent phenotypic shift from a proinflammatory (3 d) to an anti-inflammatory/profibrotic activation state, along with serial elaboration of monocyte and eosinophil recruitment factors. The i.v. administration of clodronate effectively reduced total BALF cell numbers, CCR2+ immature macrophages, and eosinophil influx while improving survival. In contrast, resident macrophage depletion from the intratracheal delivery of clodronate liposomes enhanced SftpcI73T -induced mortality. These results using SftpcI73T mice provide a detailed ontogeny for AE-IPF driven by alveolar epithelial dysfunction that induces a polycellular inflammation initiated by the early influx of proinflammatory CCR2+Ly6Chi immature macrophages.
Collapse
Affiliation(s)
- Alessandro Venosa
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Jeremy Katzen
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Yaniv Tomer
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Meghan Kopp
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Sarita Jamil
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Scott J Russo
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Surafel Mulugeta
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and.,Penn Center for Pulmonary Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104
| | - Michael F Beers
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and .,Penn Center for Pulmonary Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104
| |
Collapse
|
240
|
Bader JE, Enos RT, Velázquez KT, Carson MS, Sougiannis AT, McGuinness OP, Robinson CM, Murphy EA. Repeated clodronate-liposome treatment results in neutrophilia and is not effective in limiting obesity-linked metabolic impairments. Am J Physiol Endocrinol Metab 2019; 316:E358-E372. [PMID: 30576244 PMCID: PMC6415716 DOI: 10.1152/ajpendo.00438.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Depletion of macrophages is thought to be a therapeutic option for obesity-induced inflammation and metabolic dysfunction. However, whether the therapeutic effect is a direct result of reduced macrophage-derived inflammation or secondary to decreases in fat mass is controversial, as macrophage depletion has been shown to disrupt energy homeostasis. This study was designed to determine if macrophage depletion via clodronate-liposome (CLD) treatment could serve as an effective intervention to reduce obesity-driven inflammatory and metabolic impairments independent of changes in energy intake. After 16 wk on a high-fat diet (HFD) or the AIN-76A control (low-fat) diet (LFD) ( n = 30/diet treatment), male C57BL/6J mice were assigned to a CLD- or PBS-liposome treatment ( n = 15/group) for 4 wk. Liposomes were administered biweekly via intraperitoneal injections (8 administrations in total). PBS-liposome-treated groups were pair-fed to their CLD-treated dietary counterparts. Metabolic function was assessed before and after liposome treatment. Adipose tissue, as well as the liver, was investigated for macrophage infiltration and the presence of inflammatory mediators. Additionally, a complete blood count was performed. CLD treatment reduced energy intake. When controlling for energy intake, CLD treatment was unable to regress metabolic dysfunction or nonalcoholic fatty liver disease and impaired adipose tissue insulin action. Moreover, repeated CLD treatment induced neutrophilia and anemia, increased adipose tissue mRNA expression of the proinflammatory cytokines IL-6 and IL-1β, and augmented circulating IL-6 and monocyte chemoattractant protein-1 concentrations ( P < 0.05). This study suggests that repeated intraperitoneal administration of CLD to deplete macrophages attenuates obesity by limiting energy intake. Moreover, after controlling for the benefits of weight loss, the accompanying detrimental side effects limit regular CLD treatment as an effective therapeutic strategy.
Collapse
Affiliation(s)
- Jackie E Bader
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Reilly T Enos
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Kandy T Velázquez
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Meredith S Carson
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Alex T Sougiannis
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| | - Owen P McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine , Nashville, Tennessee
| | - Cory M Robinson
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University , Morgantown, West Virginia
| | - E Angela Murphy
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina , Columbia, South Carolina
| |
Collapse
|
241
|
Sima C, Viniegra A, Glogauer M. Macrophage immunomodulation in chronic osteolytic diseases-the case of periodontitis. J Leukoc Biol 2019; 105:473-487. [PMID: 30452781 PMCID: PMC6386606 DOI: 10.1002/jlb.1ru0818-310r] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/26/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022] Open
Abstract
Periodontitis (PD) is a chronic osteolytic disease that shares pathogenic inflammatory features with other conditions associated with nonresolving inflammation. A hallmark of PD is inflammation-mediated alveolar bone loss. Myeloid cells, in particular polymorphonuclear neutrophils (PMN) and macrophages (Mac), are essential players in PD by control of gingival biofilm pathogenicity, activation of adaptive immunity, as well as nonresolving inflammation and collateral tissue damage. Despite mounting evidence of significant innate immune implications to PD progression and healing after therapy, myeloid cell markers and targets for immune modulation have not been validated for clinical use. The remarkable plasticity of monocytes/Mac in response to local activation factors enables these cells to play central roles in inflammation and restoration of tissue homeostasis and provides opportunities for biomarker and therapeutic target discovery for management of chronic inflammatory conditions, including osteolytic diseases such as PD and arthritis. Along a wide spectrum of activation states ranging from proinflammatory to pro-resolving, Macs respond to environmental changes in a site-specific manner in virtually all tissues. This review summarizes the existing evidence on Mac immunomodulation therapies for osteolytic diseases in the broader context of conditions associated with nonresolving inflammation, and discusses osteoimmune implications of Macs in PD.
Collapse
Affiliation(s)
- Corneliu Sima
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Ana Viniegra
- Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Michael Glogauer
- Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
242
|
Conedera FM, Pousa AMQ, Mercader N, Tschopp M, Enzmann V. Retinal microglia signaling affects Müller cell behavior in the zebrafish following laser injury induction. Glia 2019; 67:1150-1166. [PMID: 30794326 DOI: 10.1002/glia.23601] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 12/21/2022]
Abstract
Microglia are the resident tissue macrophages of the central nervous system including the retina. Under pathophysiological conditions, microglia can signal to Müller cells, the major glial component of the retina, affecting their morphological, molecular, and functional responses. Microglia-Müller cell interactions appear to be bidirectional shaping the overall injury response in the retina. Hence, microglia and Müller cell responses to disease and injury have been ascribed both positive and negative outcomes. However, Müller cell reactivity and survival in the absence of immune cells after injury have not been investigated in detail in adult zebrafish. Here, we develop a model of focal retinal injury combined with pharmacological treatments for immune cell depletion in zebrafish. The retinal injury was induced by a diode laser to damage photoreceptors. Two pharmacological treatments were used to deplete either macrophage-microglia (PLX3397) or selectively eliminate peripheral macrophages (clodronate liposomes). We show that PLX3397 treatment hinders retinal regeneration in zebrafish, which is reversed by microglial repopulation. On the other hand, selective macrophage elimination did not affect the kinetics of retinal regeneration. The absence of retinal microglia and macrophages leads to dysregulated Müller cell behavior. In the untreated fish, Müller cells react after injury induction showing glial fibrillary acidic protein (GFAP), Phospho-p44/42 MAPK (Erk1/2), and PCNA upregulation. However, in the immunosuppressed animals, GFAP and phospho-p44/42 MAPK (Erk1/2) expression was not upregulated overtime and the reentry in the cell cycle was not affected. Thus, microglia and Müller cell signaling is pivotal to unlock the regenerative potential of Müller cells in order to repair the damaged retina.
Collapse
Affiliation(s)
- Federica Maria Conedera
- Department of Ophthalmology, University Hospital of Bern, University of Bern, Bern, Switzerland.,Department of BioMedical Research, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Ana Maria Quintela Pousa
- Department of Ophthalmology, University Hospital of Bern, University of Bern, Bern, Switzerland.,Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Nadia Mercader
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Markus Tschopp
- Department of Ophthalmology, University Hospital of Bern, University of Bern, Bern, Switzerland.,Department of Ophthalmology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Volker Enzmann
- Department of Ophthalmology, University Hospital of Bern, University of Bern, Bern, Switzerland.,Department of BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
243
|
Huang K, Liu Y, Tang H, Qiu M, Li C, Duan C, Wang C, Yang J, Zhou X. Glabridin Prevents Doxorubicin-Induced Cardiotoxicity Through Gut Microbiota Modulation and Colonic Macrophage Polarization in Mice. Front Pharmacol 2019; 10:107. [PMID: 30833897 PMCID: PMC6387923 DOI: 10.3389/fphar.2019.00107] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/28/2019] [Indexed: 01/12/2023] Open
Abstract
The chemotherapeutic drug doxorubicin (DOX) provokes a dose-related cardiotoxicity. Thus, there is an urgent need to identify the underlying mechanisms and develop strategies to overcome them. Here we demonstrated that glabridin (GLA), an isoflavone from licorice root, prevents DOX-induced cardiotoxicity through gut microbiota modulation and colonic macrophage polarization in mice. GLA reduced DOX-induced leakage of myocardial enzymes including aminotransferase, creatine kinase, lactate dehydrogenase, and creatine kinase-MB. GLA downregulated pro-apoptotic proteins (Bax, cleaved-caspase 9 and cleaved-caspase 3) and upregulated anti-apoptotic proteins (HAX-1 and Bcl-2) in the cardiac tissues. In addition, GLA modulated DOX-induced dysbiosis of gut microbiota and thereby decreased the ratio of M1/M2 colonic macrophage, accompanied by the downregulated lipopolysaccharide (LPS) and upregulated butyrate in the feces and peripheral blood. The leakage of myocardial enzymes induced by the DOX was decreased by antibiotics treatment, but not altered by co-treatment with the GLA and antibiotics. The ratio of M1/M2 colonic macrophage and leakage of myocardial enzymes reduced by the GLA were greatly increased by the Desulfovibrio vulgaris or LPS but decreased by the butyrate. Depletion of the macrophage attenuated DOX-induced cardiotoxicity but failed to further affect the effects of GLA. Importantly, GLA decreased production of M1 cytokines (IL-1β and TNF-α) but increased production of M2 cytokines (IL-10 and TGF-β) in the colonic macrophage with the downregulation of NF-κB and the upregulation of STAT6. In summary, GLA prevents DOX-induced cardiotoxicity through gut microbiota modulation and colonic macrophage polarization, and may serve as a potential therapeutic strategy for the DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Keqing Huang
- Department of Cardiology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Yanzhuo Liu
- Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Medical Information Analysis & Tumor Diagnosis and Treatment, Key Laboratory of Cognitive Science, College of Biomedical Engineering, South Central University for Nationalities, Wuhan, China
| | - Honglin Tang
- Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Miao Qiu
- Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China.,Shenzhen Stomatological Hospital of Southern Medical University, Shenzhen, China
| | - Chenhong Li
- Laboratory of Membrane Ion Channels and Medicine, Key Laboratory of Cognitive Science, State Ethnic Affairs Commission, College of Biomedical Engineering, South Central University for Nationalities, Wuhan, China
| | - Chenfan Duan
- Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Chenlong Wang
- Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jing Yang
- Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Xiaoyang Zhou
- Department of Cardiology, Renmin Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
244
|
Aertker BM, Kumar A, Prabhakara KS, Smith P, Furman NET, Hasen X, Cox CS, Bedi SS. Pre-injury monocyte/macrophage depletion results in increased blood-brain barrier permeability after traumatic brain injury. J Neurosci Res 2019; 97:698-707. [PMID: 30746753 DOI: 10.1002/jnr.24395] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 02/04/2023]
Abstract
Traumatic brain injury (TBI) effects both the brain and the immune system. Circulating monocytes/macrophages (Mo /Ma ) after a TBI may play an important role in preserving the blood-brain barrier (BBB), reducing brain edema, and interacting with resident microglia. To elucidate the role of circulating Mo /Ma , we utilized a monocyte/macrophage depletion model in response to TBI in male rats. Clodronate liposomes (CL) were used to deplete circulating Mo /Ma . A controlled cortical impact (CCI) injury model was used to create a TBI. All animals received either CL or PBS liposomes (PL), 48 and 24 hr prior to the procedure, and were sacrificed 72 hr post-injury for analysis of BBB permeability, brain edema, whole blood (Mo /Ma and granulocytes), and/or microglial analysis. Animals undergoing Mo /Ma depletion with CL prior to CCI (CCI-CL) were found to have increased BBB permeability when compared to non-depleted CCI (CCI-PL) animals. At 72 hr following injury, Sham-CL maintained on average an 82% reduction in the whole blood monocytes when compared to Sham-PL (p < 0.001). Monocytes in the whole blood remained significantly lower in CCI-CL animals when compared to CCI-PL (p < 0.001). The number of granulocytes in the whole blood of CCI-CL animals was higher at 3 days when compared to CCI-PL (p < 0.022). Surprisingly, the depletion of Mo /Ma did not affect brain edema. However, the depletion of Mo /Ma did result in a significant decrease in microglia (CCI-CL vs. CCI-PL, p < 0.012). In conclusion, an intact Mo /Ma population is required to repair BBB integrity and microglial response following injury.
Collapse
Affiliation(s)
- Benjamin M Aertker
- Departments of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas
| | - Akshita Kumar
- Departments of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas
| | - Karthik S Prabhakara
- Departments of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas
| | - Philippa Smith
- Departments of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas
| | - Naama E Toledano Furman
- Departments of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas
| | - Xue Hasen
- Departments of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas
| | - Charles S Cox
- Departments of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas
| | - Supinder S Bedi
- Departments of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas
| |
Collapse
|
245
|
Prior Exposure to Immunosuppressors Sensitizes Retinal Microglia and Accelerates Optic Nerve Regeneration in Zebrafish. Mediators Inflamm 2019; 2019:6135795. [PMID: 30881223 PMCID: PMC6387731 DOI: 10.1155/2019/6135795] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 11/15/2018] [Accepted: 11/28/2018] [Indexed: 12/21/2022] Open
Abstract
As adult mammals lack the capacity to replace or repair damaged neurons, degeneration and trauma (and subsequent dysfunction) of the central nervous system (CNS) seriously constrains the patient's life quality. Recent work has shown that appropriate modulation of acute neuroinflammation upon CNS injury can trigger a regenerative response; yet, the underlying cellular and molecular mechanisms remain largely elusive. In contrast to mammals, zebrafish retain high regenerative capacities into adulthood and thus form a powerful model to study the contribution of neuroinflammation to successful regeneration. Here, we used pharmacological immunosuppression methods to study the role of microglia/macrophages during optic nerve regeneration in adult zebrafish. We first demonstrated that systemic immunosuppression with dexamethasone (dex) impedes regeneration after optic nerve injury. Secondly, and strikingly, local intravitreal application of dex or clodronate liposomes prior to injury was found to sensitize retinal microglia. Consequently, we observed an exaggerated inflammatory response to subsequent optic nerve damage, along with enhanced tectal reinnervation. In conclusion, we found a strong positive correlation between the acute inflammatory response in the retina and the regenerative capacity of the optic nerve in adult zebrafish subjected to nerve injury.
Collapse
|
246
|
Microglial Depletion with Clodronate Liposomes Increases Proinflammatory Cytokine Levels, Induces Astrocyte Activation, and Damages Blood Vessel Integrity. Mol Neurobiol 2019; 56:6184-6196. [PMID: 30734229 DOI: 10.1007/s12035-019-1502-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 01/18/2019] [Indexed: 01/22/2023]
Abstract
Investigators are increasingly interested in using microglial depletion to study the role of microglia under pathologic conditions. Liposome-encapsulated clodronate is commonly used to eliminate macrophage populations because it causes functionally irreversible inhibition and apoptosis once phagocytized by macrophages. Recent studies have shown that microglia can be depleted in disease models by injecting clodronate liposomes into the brain parenchyma. However, it is unclear whether intracerebral administration of clodronate liposomes is a practical method of eliminating microglia under physiologic conditions or whether microglial depletion induces damage to other brain cells. In this study, injecting 1 μL of clodronate liposomes (7 μg/μL) into the striatum of mice caused ablation of microglia at 1 day that persisted for 3 days. Microglia reappeared in the boundary regions of microglia elimination after 5 days. Importantly, we observed an increase in proinflammatory cytokine levels and an increase in neural/glial antigen 2 and glial fibrillary acidic protein expression in the perilesional region. In contrast, expression levels of myelin basic protein, microtubule-associated protein 2, and postsynaptic protein-95 decreased in the periphery of regions where microglia were depleted. Moreover, clodronate liposome administration decreased the density and integrity of blood vessels in the perilesional regions. In cultured primary neurons, clodronate liposome exposure also attenuated ATP synthesis. Together, these findings suggest that intracerebral administration of clodronate liposomes into brain parenchyma can deplete microglia, but can also damage other brain cells and blood vessel integrity.
Collapse
|
247
|
Marro BS, Legrain S, Ware BC, Oldstone MB. Macrophage IFN-I signaling promotes autoreactive T cell infiltration into islets in type 1 diabetes model. JCI Insight 2019; 4:125067. [PMID: 30674713 DOI: 10.1172/jci.insight.125067] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/11/2018] [Indexed: 01/05/2023] Open
Abstract
Here, we report a pathogenic role for type I IFN (IFN-I) signaling in macrophages, and not β cells in the islets, for the development of type 1 diabetes (T1D). Following lymphocytic choriomeningitis (LCMV) infection in the Rip-LCMV-GP T1D model, macrophages accumulated near islets and in close contact to islet-infiltrating GP-specific (autoimmune) CD8+ T cells. Depletion of macrophages with clodronate liposomes or genetic ablation of Ifnar in macrophages aborted T1D, despite proliferation of GP-specific (autoimmune) CD8+ T cells. Histopathologically, disrupted IFNα/β receptor (IFNAR) signaling in macrophages resulted in restriction of CD8+ T cells entering into the islets with significant lymphoid accumulation around the islet. Collectively, these results provide evidence that macrophages via IFN-I signaling, while not entering the islets, are directly involved in interacting, directing, or restricting trafficking of autoreactive-specific T cells into the islets as an important component in causing T1D.
Collapse
|
248
|
Aires ID, Boia R, Rodrigues-Neves AC, Madeira MH, Marques C, Ambrósio AF, Santiago AR. Blockade of microglial adenosine A 2A receptor suppresses elevated pressure-induced inflammation, oxidative stress, and cell death in retinal cells. Glia 2019; 67:896-914. [PMID: 30667095 PMCID: PMC6590475 DOI: 10.1002/glia.23579] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 11/08/2018] [Accepted: 11/28/2018] [Indexed: 12/20/2022]
Abstract
Glaucoma is a retinal degenerative disease characterized by the loss of retinal ganglion cells and damage of the optic nerve. Recently, we demonstrated that antagonists of adenosine A2A receptor (A2A R) control retinal inflammation and afford protection to rat retinal cells in glaucoma models. However, the precise contribution of microglia to retinal injury was not addressed, as well as the effect of A2A R blockade directly in microglia. Here we show that blocking microglial A2A R prevents microglial cell response to elevated pressure and it is sufficient to protect retinal cells from elevated pressure-induced death. The A2A R antagonist SCH 58261 or the knockdown of A2A R expression with siRNA in microglial cells prevented the increase in microglia response to elevated hydrostatic pressure. Furthermore, in retinal neural cell cultures, the A2A R antagonist decreased microglia proliferation, as well as the expression and release of pro-inflammatory mediators. Microglia ablation prevented neural cell death triggered by elevated pressure. The A2A R blockade recapitulated the effects of microglia depletion, suggesting that blocking A2A R in microglia is able to control neurodegeneration in glaucoma-like conditions. Importantly, in human organotypic retinal cultures, A2A R blockade prevented the increase in reactive oxygen species and the morphological alterations in microglia triggered by elevated pressure. These findings place microglia as the main contributors for retinal cell death during elevated pressure and identify microglial A2A R as a therapeutic target to control retinal neuroinflammation and prevent neural apoptosis elicited by elevated pressure.
Collapse
Affiliation(s)
- Inês Dinis Aires
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Raquel Boia
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Ana Catarina Rodrigues-Neves
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Maria Helena Madeira
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Carla Marques
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - António Francisco Ambrósio
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Ana Raquel Santiago
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal.,Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, Portugal
| |
Collapse
|
249
|
Depletion of RIPK1 in hepatocytes exacerbates liver damage in fulminant viral hepatitis. Cell Death Dis 2019; 10:12. [PMID: 30622241 PMCID: PMC6325114 DOI: 10.1038/s41419-018-1277-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/21/2018] [Accepted: 12/03/2018] [Indexed: 12/19/2022]
Abstract
The protein kinase RIPK1 plays a crucial role at the crossroad of stress-induced signaling pathways that affects cell’s decision to live or die. The present study aimed to define the role of RIPK1 in hepatocytes during fulminant viral hepatitis, a worldwide syndrome mainly observed in hepatitis B virus (HBV) infected patients. Mice deficient for RIPK1, specifically in liver parenchymal cells (Ripk1LPC-KO) and their wild-type littermates (Ripk1fl/fl), were challenged by either the murine hepatitis virus type 3 (MHV3) or poly I:C, a synthetic analog of double-stranded RNA mimicking viral pathogen-associated molecular pattern. Ripk1LPC-KO mice developed more severe symptoms at early stage of the MHV3-induced fulminant hepatitis. Similarly, administration of poly I:C only triggered increase of systemic transaminases in Ripk1LPC-KO mice, reflecting liver damage through induced apoptosis as illustrated by cleaved-caspase 3 labeling of liver tissue sections. Neutralization of TNF-α or prior depletion of macrophages were able to prevent the appearance of apoptosis of hepatocytes in poly I:C-challenged Ripk1LPC-KO mice. Moreover, poly I:C never induced direct hepatocyte death in primary culture whatever the murine genotype, while it always stimulated an anti-viral response. Our investigations demonstrated that RIPK1 protects hepatocytes from TNF-α secreted from macrophages during viral induced fulminant hepatitis. These data emphasize the potential worsening risks of an HBV infection in people with polymorphism or homozygous amorphic mutations already described for the RIPK1 gene.
Collapse
|
250
|
Abstract
Macrophages are innate immune cells, which have important roles in the inflammatory response to infections or tissue injury, and have an equally important role in the resolution of inflammation. Macrophages play a key part in directing the innate immune response and subsequent adaptive immune response. They can acquire a variety of distinct but also overlapping activation states, depending on the local microenvironment, in order to perform these functions. Stimuli, such as IFNγ and LPS, can promote an inflammatory activation state, which is associated with the production of reactive oxygen species, and pro-inflammatory cytokines and chemokines. Immune complexes and LPS can promote an anti-inflammatory activation state to prevent damage to the host, which is associated with the production of high levels of the anti-inflammatory cytokine IL-10 and low levels of pro-inflammatory cytokines. Wound-healing macrophages can be activated by IL-4 or IL-13 and have roles in tissue remodeling and the resolution of inflammation. Macrophages are present in nearly every tissue of the body and are important for maintaining homeostasis, but their dysfunction can also lead to diseases, such as inflammatory bowel disease. To study the role macrophages play in a complex in vivo environment, depletion and reconstitution experiments can be utilized. Clodronate liposomes are an effective and versatile way to deplete macrophages in vivo; they can allow selective depletion from tissues of interest and can be used on transgenic mice. However, clodronate liposomes deplete all types of macrophages as well as dendritic cells, so other strategies are required in parallel to determine whether macrophages or macrophages of a particular activation state are required. Reconstitution of macrophages by adoptive transfer can be performed, with or without prior depletion, to further suggest that the observed effect is macrophage dependent. Macrophages activated ex vivo or macrophages from transgenic mice can be adoptively transferred during disease models to determine whether a specific protein or activation state affects disease outcome. Macrophage contribution to health and disease can be effectively studied using depletion with clodronate liposomes and by macrophage reconstitution, as demonstrated in this chapter.
Collapse
|