1
|
Herrero-Fernández B, Ortega-Zapero M, Gómez-Bris R, Sáez A, Iborra S, Zorita V, Quintas A, Vázquez E, Dopazo A, Sánchez-Madrid F, Arribas SM, González-Granado JM. Role of lamin A/C on dendritic cell function in antiviral immunity. Cell Mol Life Sci 2024; 81:400. [PMID: 39264480 PMCID: PMC11393282 DOI: 10.1007/s00018-024-05423-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/29/2024] [Accepted: 08/23/2024] [Indexed: 09/13/2024]
Abstract
Dendritic cells (DCs) play a crucial role in orchestrating immune responses, particularly in promoting IFNγ-producing-CD8 cytotoxic T lymphocytes (CTLs) and IFNγ-producing-CD4 T helper 1 (Th1) cells, which are essential for defending against viral infections. Additionally, the nuclear envelope protein lamin A/C has been implicated in T cell immunity. Nevertheless, the intricate interplay between innate and adaptive immunity in response to viral infections, particularly the role of lamin A/C in DC functions within this context, remains poorly understood. In this study, we demonstrate that mice lacking lamin A/C in myeloid LysM promoter-expressing cells exhibit a reduced capacity to induce Th1 and CD8 CTL responses, leading to impaired clearance of acute primary Vaccinia virus (VACV) infection. Remarkably, in vitro-generated granulocyte macrophage colony-stimulating factor bone marrow-derived DCs (GM-CSF BMDCs) show high levels of lamin A/C. Lamin A/C absence on GM-CSF BMDCs does not affect the expression of costimulatory molecules on the cell membrane but it reduces the cellular ability to form immunological synapses with naïve CD4 T cells. Lamin A/C deletion induces alterations in NFκB nuclear localization, thereby influencing NF-κB-dependent transcription. Furthermore, lamin A/C ablation modifies the gene accessibility of BMDCs, predisposing these cells to mount a less effective antiviral response upon TLR stimulation. This study highlights the critical role of DCs in interacting with CD4 T cells during antiviral responses and proposes some mechanisms through which lamin A/C may modulate DC function via gene accessibility and transcriptional regulation.
Collapse
Affiliation(s)
- Beatriz Herrero-Fernández
- LamImSys Lab, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, 28041, Spain
- Department of Physiology, Faculty of Medicine, Universidad Autonoma de Madrid, Madrid, 28029, Spain
| | - Marina Ortega-Zapero
- LamImSys Lab, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, 28041, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Raquel Gómez-Bris
- LamImSys Lab, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, 28041, Spain
- Department of Physiology, Faculty of Medicine, Universidad Autonoma de Madrid, Madrid, 28029, Spain
| | - Angela Sáez
- LamImSys Lab, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, 28041, Spain
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria (UFV), Pozuelo de Alarcón, 28223, Spain
| | - Salvador Iborra
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain
- Fundacion Inmunotek, Alcalá de Henares, 28805, Spain
| | - Virginia Zorita
- Centro Nacional de Investigaciones, Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Ana Quintas
- Centro Nacional de Investigaciones, Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Enrique Vázquez
- Centro Nacional de Investigaciones, Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Ana Dopazo
- Centro Nacional de Investigaciones, Cardiovasculares (CNIC), Madrid, 28029, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Centro Nacional de Investigaciones, Cardiovasculares (CNIC), Madrid, 28029, Spain
- Immunology Unit, Medicine Department, Hospital Universitario La Princesa, Universidad Autónoma de Madrid, Instituto Investigacion Sanitaria-Princesa IIS-IP, Madrid, Spain, Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Silvia Magdalena Arribas
- Department of Physiology, Faculty of Medicine, Universidad Autonoma de Madrid, Madrid, 28029, Spain.
| | - Jose Maria González-Granado
- LamImSys Lab, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, 28041, Spain.
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain.
- Centro de Investigacion Biomedica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| |
Collapse
|
2
|
Zhang F, Cui Y, Zhang T, Yin W. Epigenetic regulation of macrophage activation in chronic obstructive pulmonary disease. Front Immunol 2024; 15:1445372. [PMID: 39206196 PMCID: PMC11349576 DOI: 10.3389/fimmu.2024.1445372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Macrophages in the innate immune system play a vital role in various lung diseases such as asthma, chronic obstructive pulmonary disease (COPD), acute lung injury and pulmonary fibrosis. Macrophages involved in the process of immunity need to go through a process of activation, including changes in gene expression and cell metabolism. Epigenetic modifications are key factors of macrophage activation including DNA methylation, histone modification and non-coding RNA regulation. Understanding the role and mechanisms of epigenetic regulation of macrophage activation can provide insights into the function of macrophages in lung diseases and help identification of potential therapeutic targets. This review summarizes the latest progress in the epigenetic changes and regulation of macrophages in their development process and in normal physiological states, and the epigenetic regulation of macrophages in COPD as well as the influence of macrophage activation on COPD development.
Collapse
Affiliation(s)
- Feng Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GMU) - Guangzhou Institutes of Biomedicine and Health (GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Yachao Cui
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GMU) - Guangzhou Institutes of Biomedicine and Health (GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Tiejun Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GMU) - Guangzhou Institutes of Biomedicine and Health (GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Medical University Affiliated Qingyuan Hospital, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenguang Yin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GMU) - Guangzhou Institutes of Biomedicine and Health (GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Kim HY, Moon JO, Kim SW. Development and application of a multi-step porcine in vitro system to evaluate feedstuffs and feed additives for their efficacy in nutrient digestion, digesta characteristics, and intestinal immune responses. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 17:265-282. [PMID: 38800740 PMCID: PMC11127235 DOI: 10.1016/j.aninu.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/09/2024] [Accepted: 01/17/2024] [Indexed: 05/29/2024]
Abstract
In vitro model provides alternatives to the use of live animals in research. In pig nutrition, there has been a tremendous increase in in vivo research over the decades. Proper utilization of in vitro models could provide a screening tool to reduce the needs of in vivo studies, research duration, cost, and the use of animals and feeds. This study aimed to develop a multi-step porcine in vitro system to simulate nutrient digestion and intestinal epithelial immune responses affected by feedstuffs and feed additives. Seven feedstuffs (corn, corn distillers dried grains with solubles [corn DDGS], barley, wheat, soybean meal, soy protein concentrates, and Corynebacterium glutamicum cell mass [CGCM]), feed enzymes (xylanase and phytase), and supplemental amino acids (arginine, methionine, and tryptophan), were used in this in vitro evaluation for their efficacy on digestibility, digesta characteristics, and intestinal health compared with the results from previously published in vivo studies. All in vitro evaluations were triplicated. Data were analyzed using Mixed procedure of SAS9.4. Evaluations included (1) nutrient digestibility of feedstuffs, (2) the effects of feed enzymes, xylanase and phytase, on digestibility of feedstuffs and specific substrates, and (3) the effects of amino acids, arginine, tryptophan, and methionine, on anti-inflammatory, anti-oxidative, and anti-heat stress statuses showing their effects (P < 0.05) on the measured items. Differences in dry matter and crude protein digestibility among the feedstuffs as well as effects of xylanase and phytase were detected (P < 0.05), including xylo-oligosaccharide profiles and phosphorus release from phytate. Supplementation of arginine, tryptophan, and methionine modulated (P < 0.05) cellular inflammatory and oxidative stress responses. The use of this in vitro model allowed the use of 3 experimental replications providing sufficient statistical power at P < 0.05. This indicates in vitro models can have increased precision and consistency compared with in vivo animal studies.
Collapse
Affiliation(s)
- Hee Yeon Kim
- Application Center, CJ Blossom Park, Suwon, South Korea
| | - Jun-Ok Moon
- Application Center, CJ Blossom Park, Suwon, South Korea
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
4
|
Castellano M, Blanco V, Calzi ML, Costa B, Witwer K, Hill M, Cayota A, Segovia M, Tosar JP. Ribonuclease activity undermines immune sensing of naked extracellular RNA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.23.590771. [PMID: 38712104 PMCID: PMC11071435 DOI: 10.1101/2024.04.23.590771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The plasma membrane and the membrane of endosomal vesicles are considered physical barriers preventing extracellular RNA uptake. While naked RNA can be spontaneously internalized by certain cells types, functional delivery of naked RNA into the cytosol has been rarely observed. Here we show that extracellular ribonucleases, mainly derived from cell culture supplements, have so far hindered the study of extracellular RNA functionality. In the presence of active ribonuclease inhibitors (RI), naked bacterial RNA is pro-inflammatory when spiked in the media of dendritic cells and macrophages. In murine cells, this response mainly depends on the action of endosomal Toll-like receptors. However, we also show that naked RNA can perform endosomal escape and engage with cytosolic RNA sensors and ribosomes. For example, naked mRNAs encoding reporter proteins can be spontaneously internalized and translated by a variety of cell types, in an RI-dependent manner. In vivo, RI co-injection enhances the activation induced by naked extracellular RNA on splenic lymphocytes and myeloid-derived leukocytes. Furthermore, naked extracellular RNA is inherently pro-inflammatory in ribonuclease-poor compartments such as the peritoneal cavity. Overall, these results demonstrate that naked RNA is bioactive and does not need encapsulation inside synthetic or biological lipid vesicles for functional uptake, making a case for nonvesicular extracellular RNA-mediated intercellular communication.
Collapse
Affiliation(s)
- Mauricio Castellano
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
- Immunoregulation and Inflammation Laboratory, Institut Pasteur Montevideo, Uruguay
| | - Valentina Blanco
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
| | - Marco Li Calzi
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
| | - Bruno Costa
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
- Analytical Biochemistry Unit, School of Science, Universidad de la República, Uruguay
| | - Kenneth Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- EV Core Facility “EXCEL”, Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marcelo Hill
- Immunoregulation and Inflammation Laboratory, Institut Pasteur Montevideo, Uruguay
- Academic Unit of Immunobiology, School of Medicine, Universidad de la República, Uruguay
| | - Alfonso Cayota
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
- Hospital de Clínicas, Universidad de la República, Uruguay
| | - Mercedes Segovia
- Immunoregulation and Inflammation Laboratory, Institut Pasteur Montevideo, Uruguay
- Academic Unit of Immunobiology, School of Medicine, Universidad de la República, Uruguay
| | - Juan Pablo Tosar
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
- Analytical Biochemistry Unit, School of Science, Universidad de la República, Uruguay
| |
Collapse
|
5
|
Liu X, Wang X, Ma X, Li H, Miao C, Tian Z, Hu Y. Genetic disruption of Ano5 leads to impaired osteoclastogenesis for gnathodiaphyseal dysplasia. Oral Dis 2024; 30:1403-1415. [PMID: 36989132 DOI: 10.1111/odi.14562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/19/2023]
Abstract
OBJECTIVES Gnathodiaphyseal dysplasia (GDD; OMIM#166260) is a rare skeletal genetic disorder characterized by sclerosis of tubular bones and cemento-osseous lesions in mandibles. TMEM16E/ANO5 gene mutations have been identified in patients with GDD. Here, Ano5 knockout (Ano5-/-) mice with enhanced osteoblastogenesis were used to investigate whether Ano5 disruption affects osteoclastogenesis. SUBJECTS AND METHODS The maturation of osteoclasts, formation of F-actin ring and bone resorption were detected by immunohistochemistry, TRAP, phalloidin staining and Coming Osteo assays. The expression of osteoclast-related factors was measured by qRT-PCR. Early signaling pathways were verified by western blot. RESULTS Ano5-/- mice exhibited inhibitory formation of multinucleated osteoclasts with a reduction of TRAP activity. The expression of Nfatc1, c-Fos, Trap, Ctsk, Mmp9, Rank and Dc-stamp was significantly decreased in bone tissues and bone marrow-derived macrophages (BMMs) of Ano5-/- mice. Ano5-/- osteoclasts manifested disrupted actin ring and less mineral resorption. RANKL-induced early signaling pathways were suppressed in Ano5-/- osteoclasts and Ano5 knockdown RAW264.7 cells. Moreover, the inhibitory effects of NF-κB signalling pathway on osteoclastogenesis were partially attenuated with NF-κB signalling activator. CONCLUSIONS Ano5 deficiency impairs osteoclastogenesis, which leads to enhanced osteogenic phenotypes mediated by bone homeostasis dysregulation.
Collapse
Affiliation(s)
- Xiu Liu
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
- Department of Oral Medicine, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Xiaoyu Wang
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Xinrong Ma
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Hongyu Li
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Congcong Miao
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Zhenchuan Tian
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Ying Hu
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| |
Collapse
|
6
|
Lazarus HM, Pitts K, Wang T, Lee E, Buchbinder E, Dougan M, Armstrong DG, Paine R, Ragsdale CE, Boyd T, Rock EP, Gale RP. Recombinant GM-CSF for diseases of GM-CSF insufficiency: Correcting dysfunctional mononuclear phagocyte disorders. Front Immunol 2023; 13:1069444. [PMID: 36685591 PMCID: PMC9850113 DOI: 10.3389/fimmu.2022.1069444] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction Endogenous granulocyte-macrophage colony-stimulating factor (GM-CSF), identified by its ability to support differentiation of hematopoietic cells into several types of myeloid cells, is now known to support maturation and maintain the metabolic capacity of mononuclear phagocytes including monocytes, macrophages, and dendritic cells. These cells sense and attack potential pathogens, present antigens to adaptive immune cells, and recruit other immune cells. Recombinant human (rhu) GM-CSF (e.g., sargramostim [glycosylated, yeast-derived rhu GM-CSF]) has immune modulating properties and can restore the normal function of mononuclear phagocytes rendered dysfunctional by deficient or insufficient endogenous GM-CSF. Methods We reviewed the emerging biologic and cellular effects of GM-CSF. Experts in clinical disease areas caused by deficient or insufficient endogenous GM-CSF examined the role of GM-CSF in mononuclear phagocyte disorders including autoimmune pulmonary alveolar proteinosis (aPAP), diverse infections (including COVID-19), wound healing, and anti-cancer immune checkpoint inhibitor therapy. Results We discuss emerging data for GM-CSF biology including the positive effects on mitochondrial function and cell metabolism, augmentation of phagocytosis and efferocytosis, and immune cell modulation. We further address how giving exogenous rhu GM-CSF may control or treat mononuclear phagocyte dysfunction disorders caused or exacerbated by GM-CSF deficiency or insufficiency. We discuss how rhu GM-CSF may augment the anti-cancer effects of immune checkpoint inhibitor immunotherapy as well as ameliorate immune-related adverse events. Discussion We identify research gaps, opportunities, and the concept that rhu GM-CSF, by supporting and restoring the metabolic capacity and function of mononuclear phagocytes, can have significant therapeutic effects. rhu GM-CSF (e.g., sargramostim) might ameliorate multiple diseases of GM-CSF deficiency or insufficiency and address a high unmet medical need.
Collapse
Affiliation(s)
- Hillard M. Lazarus
- Department of Medicine, Division of Hematology and Oncology, Case Western Reserve University, Cleveland, OH, United States
| | - Katherine Pitts
- Medical Affairs, Partner Therapeutics, Inc., Lexington, MA, United States
| | - Tisha Wang
- Division of Pulmonary, Critical Care, and Sleep Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Elinor Lee
- Division of Pulmonary, Critical Care, and Sleep Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Elizabeth Buchbinder
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Michael Dougan
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - David G. Armstrong
- Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Robert Paine
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT, United States
| | | | - Timothy Boyd
- Clinical Development, Partner Therapeutics, Inc., Lexington, MA, United States
| | - Edwin P. Rock
- Clinical Development, Partner Therapeutics, Inc., Lexington, MA, United States
| | - Robert Peter Gale
- Hematology Centre, Department of Immunology and Inflammation, Imperial College, London, United Kingdom
| |
Collapse
|
7
|
Maisonneuve L, Manoury B. In Vitro and In Vivo Assays to Evaluate Dendritic Cell Phagocytic Capacity. Methods Mol Biol 2023; 2618:279-288. [PMID: 36905524 DOI: 10.1007/978-1-0716-2938-3_20] [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: 03/12/2023]
Abstract
Phagocytosis is a process by which specific immune cells such as macrophages or dendritic cells engulf large particles. It is an important innate immune defense mechanism for removing a wide variety of pathogens and apoptotic cells. Following phagocytosis, nascent phagosomes are formed which, when fused to lysosome to become phagolysosome containing acidic proteases, will allow the degradation of ingested material. This chapter describes in vitro and in vivo assays to measure phagocytosis by murine dendritic cells using amine beads coupled with streptavidin Alexa 488. This protocol can also be applied to monitor phagocytosis in human dendritic cells.
Collapse
Affiliation(s)
- Lucie Maisonneuve
- Institut Necker Enfants Malades, INSERM U1151-CNRS UMR 8253, Université de Paris, Faculté de Médecine Necker, Paris, France
| | - Bénédicte Manoury
- Institut Necker Enfants Malades, INSERM U1151-CNRS UMR 8253, Université de Paris, Faculté de Médecine Necker, Paris, France.
| |
Collapse
|
8
|
Manthrirathna MATP, Dangerfield EM, Ishizuka S, Woods A, Luong BS, Yamasaki S, Timmer MSM, Stocker BL. Water-soluble trehalose glycolipids show superior Mincle binding and signaling but impaired phagocytosis and IL-1β production. Front Mol Biosci 2022; 9:1015210. [PMID: 36504717 PMCID: PMC9729344 DOI: 10.3389/fmolb.2022.1015210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/25/2022] [Indexed: 11/25/2022] Open
Abstract
The tremendous potential of trehalose glycolipids as vaccine adjuvants has incentivized the study of how the structures of these ligands relate to their Mincle-mediated agonist activities. Despite this, structure-activity work in the field has been largely empirical, and less is known about how Mincle-independent pathways might be affected by different trehalose glycolipids, and whether Mincle binding by itself can serve as a proxy for adjuvanticity. There is also much demand for more water-soluble Mincle ligands. To address this need, we prepared polyethylene glycol modified trehalose glycolipids (PEG-TGLs) with enhanced water solubility and strong murine Mincle (mMincle) binding and signaling. However, only modest cytokine and chemokine responses were observed upon the treatment of GM-CSF treated bone-marrow cells with the PEG-TGLs. Notability, no IL-1β was observed. Using RNA-Seq analysis and a representative PEG-TGL, we determined that the more water-soluble adducts were less able to activate phagocytic pathways, and hence, failed to induce IL-1β production. Taken together, our data suggests that in addition to strong Mincle binding, which is a pre-requisite for Mincle-mediated cellular responses, the physical presentation of trehalose glycolipids in colloidal form is required for inflammasome activation, and hence, a strong inflammatory immune response.
Collapse
Affiliation(s)
| | - Emma M. Dangerfield
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand,Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Shigenari Ishizuka
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan,Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Aodhamair Woods
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Brenda S. Luong
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand,Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan,Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan,Division of Molecular Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan,Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan
| | - Mattie S. M. Timmer
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand,Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand,*Correspondence: Bridget L. Stocker, ; Mattie S. M. Timmer,
| | - Bridget L. Stocker
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand,Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand,*Correspondence: Bridget L. Stocker, ; Mattie S. M. Timmer,
| |
Collapse
|
9
|
Zimmermann J, Goretzki A, Meier C, Wolfheimer S, Lin YJ, Rainer H, Krause M, Wedel S, Spies G, Führer F, Vieths S, Scheurer S, Schülke S. Modulation of dendritic cell metabolism by an MPLA-adjuvanted allergen product for specific immunotherapy. Front Immunol 2022; 13:916491. [PMID: 36059475 PMCID: PMC9430023 DOI: 10.3389/fimmu.2022.916491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 07/18/2022] [Indexed: 01/20/2023] Open
Abstract
Background Recently, bacterial components were shown to enhance immune responses by shifting immune cell metabolism towards glycolysis and lactic acid production, also known as the Warburg Effect. Currently, the effect of allergen products for immunotherapy (AIT) and commercial vaccines on immune cell metabolism is mostly unknown. Objective To investigate the effect of AIT products (adjuvanted with either MPLA or Alum) on myeloid dendritic cell (mDC) metabolism and activation. Methods Bone marrow-derived mDCs were stimulated with five allergoid-based AIT products (one adjuvanted with MPLA, four adjuvanted with Alum) and two MPLA-adjuvanted vaccines and analyzed for their metabolic activation, expression of cell surface markers, and cytokine secretion by ELISA. mDCs were pre-incubated with either immunological or metabolic inhibitors or cultured in glucose- or glutamine-free culture media and subsequently stimulated with the MPLA-containing AIT product (AIT product 1). mDCs were co-cultured with allergen-specific CD4+ T cells to investigate the contribution of metabolic pathways to the T cell priming capacity of mDCs stimulated with AIT product 1. Results Both the MPLA-containing AIT product 1 and commercial vaccines, but not the Alum-adjuvanted AIT products, activated Warburg metabolism and TNF-α secretion in mDCs. Further experiments focused on AIT product 1. Metabolic analysis showed that AIT product 1 increased glycolytic activity while also inducing the secretion of IL-1β, IL-10, IL-12, and TNF-α. Both rapamycin (mTOR-inhibitor) and SP600125 (SAP/JNK MAPK-inhibitor) dose-dependently suppressed the AIT product 1-induced Warburg Effect, glucose consumption, IL-10-, and TNF-α secretion. Moreover, both glucose- and glutamine deficiency suppressed secretion of all investigated cytokines (IL-1β, IL-10, and TNF-α). Glucose metabolism in mDCs was also critical for the (Th1-biased) T cell priming capacity of AIT product 1-stimulated mDCs, as inhibition of mTOR signaling abrogated their ability to induce Th1-responses. Conclusion The AIT product and commercial vaccines containing the adjuvant MPLA were shown to modulate the induction of immune responses by changing the metabolic state of mDCs. Better understanding the mechanisms underlying the interactions between cell metabolism and immune responses will allow us to further improve vaccine development and AIT.
Collapse
Affiliation(s)
- Jennifer Zimmermann
- Vice President´s Research Group 1: Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Alexandra Goretzki
- Vice President´s Research Group 1: Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Clara Meier
- Vice President´s Research Group 1: Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Sonja Wolfheimer
- Vice President´s Research Group 1: Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Yen-Ju Lin
- Vice President´s Research Group 1: Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Hannah Rainer
- Vice President´s Research Group 1: Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Maren Krause
- Vice President´s Research Group 1: Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Saskia Wedel
- Vice President´s Research Group 1: Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Gerd Spies
- Z6 Occupational Safety, Paul-Ehrlich-Institut, Langen, Germany
| | - Frank Führer
- Division of Allergology, Batch Control and Allergen Analytics, Paul-Ehrlich-Institut, Langen, Germany
| | - Stefan Vieths
- Vice President´s Research Group 1: Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Stephan Scheurer
- Vice President´s Research Group 1: Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Stefan Schülke
- Vice President´s Research Group 1: Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
- *Correspondence: Stefan Schülke,
| |
Collapse
|
10
|
Borowska D, Sives S, Vervelde L, Sutton KM. Chicken CSF2 and IL-4-, and CSF2-dependent bone marrow cultures differentiate into macrophages over time. Front Immunol 2022; 13:1064084. [PMID: 36618373 PMCID: PMC9812659 DOI: 10.3389/fimmu.2022.1064084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Chicken bone marrow-derived macrophages (BMMΦ) and dendritic cells (BMDC) are utilized as models to study the mononuclear phagocytic system (MPS). A widely used method to generate macrophages and DC in vitro is to culture bone marrow cells in the presence of colony-stimulating factor-1 (CSF1) to differentiate BMMΦ and granulocyte-macrophage-CSF (GM-CSF, CSF2) and interleukin-4 (IL-4) to differentiate BMDC, while CSF2 alone can lead to the development of granulocyte-macrophage-CSF-derived DC (GMDC). However, in chickens, the MPS cell lineages and their functions represented by these cultures are poorly understood. Here, we decipher the phenotypical, functional and transcriptional differences between chicken BMMΦ and BMDC along with examining differences in DC cultures grown in the absence of IL-4 on days 2, 4, 6 and 8 of culture. BMMΦ cultures develop into a morphologically homogenous cell population in contrast to the BMDC and GMDC cultures, which produce morphologically heterogeneous cell cultures. At a phenotypical level, all cultures contained similar cell percentages and expression levels of MHCII, CD11c and CSF1R-transgene, whilst MRC1L-B expression decreased over time in BMMΦ. All cultures were efficiently able to uptake 0.5 µm beads, but poorly phagocytosed 1 µm beads. Little difference was observed in the kinetics of phagosomal acidification across the cultures on each day of analysis. Temporal transcriptomic analysis indicated that all cultures expressed high levels of CSF3R, MERTK, SEPP1, SPI1 and TLR4, genes associated with macrophages in mammals. In contrast, low levels of FLT3, XCR1 and CAMD1, genes associated with DC, were expressed at day 2 in BMDC and GMDC after which expression levels decreased. Collectively, chicken CSF2 + IL-4- and CSF2-dependent BM cultures represent cells of the macrophage lineage rather than inducing conventional DC.
Collapse
Affiliation(s)
- Dominika Borowska
- The Division of Infection and Immunity, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Samantha Sives
- The Division of Infection and Immunity, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Lonneke Vervelde
- The Division of Infection and Immunity, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Kate M Sutton
- The Division of Infection and Immunity, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
11
|
Das A, Chauhan KS, Kumar H, Tailor P. Mutation in Irf8 Gene ( Irf8R294C ) Impairs Type I IFN-Mediated Antiviral Immune Response by Murine pDCs. Front Immunol 2021; 12:758190. [PMID: 34867997 PMCID: PMC8635750 DOI: 10.3389/fimmu.2021.758190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/25/2021] [Indexed: 12/01/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs) are the key producers of type I interferons (IFNs), thus playing a central role in initiating antiviral immune response. Besides robust type I IFN production, pDCs also act as antigen presenting cells post immunogenic stimulation. Transcription factor Irf8 is indispensable for the development of both pDC and cDC1 subset. However, the mechanism underlying the differential regulation by IRF8 in cDC1- and pDC-specific genomic architecture of developmental pathways still remains to be fully elucidated. Previous studies indicated that the Irf8R294C mutation specifically abrogates development of cDC1 without affecting that of pDC. In the present study using RNA-seq based approach, we have found that though the point mutation Irf8R294C did not affect pDC development, it led to defective type I IFN production, thus resulting in inefficient antiviral response. This observation unraveled the distinctive roles of IRF8 in these two subpopulations—regulating the development of cDC1 whereas modulating the functionality of pDCs without affecting development. We have reported here that Irf8R294C mutation also caused defect in production of ISGs as well as defective upregulation of costimulatory molecules in pDCs in response to NDV infection (or CpG stimulation). Through in vivo studies, we demonstrated that abrogation of type I IFN production was concomitant with reduced upregulation of costimulatory molecules in pDCs and increased NDV burden in IRF8R294C mice in comparison with wild type, indicating inefficient viral clearance. Further, we have also shown that Irf8R294C mutation abolished the activation of type I IFN promoter by IRF8, justifying the low level of type I IFN production. Taken together, our study signifies that the single point mutation in Irf8, Irf8R294C severely compromised type I IFN-mediated immune response by murine pDCs, thereby causing impairment in antiviral immunity.
Collapse
Affiliation(s)
- Annesa Das
- Laboratory of Innate Immunity, National Institute of Immunology, New Delhi, India
| | | | - Himanshu Kumar
- Department of Biological Sciences, Laboratory of Immunology and Infectious Disease Biology, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, India
| | - Prafullakumar Tailor
- Laboratory of Innate Immunity, National Institute of Immunology, New Delhi, India.,Special Centre for Systems Medicine (SCSM), Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
12
|
Hinkle L, Liu Y, Meng C, Chen Z, Mai J, Zhang L, Xu Y, Pan PY, Chen SH, Shen H. The Sympathetic Nervous System Modulates Cancer Vaccine Activity through Monocyte-Derived Cells. THE JOURNAL OF IMMUNOLOGY 2021; 207:3131-3140. [PMID: 34772699 DOI: 10.4049/jimmunol.2100719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/05/2021] [Indexed: 11/19/2022]
Abstract
The sympathetic nervous system (SNS) is an important regulator of immune cell function during homeostasis and states of inflammation. Recently, the SNS has been found to bolster tumor growth and impair the development of antitumor immunity. However, it is unclear whether the SNS can modulate APC function. Here, we investigated the effects of SNS signaling in murine monocyte-derived macrophages (moMФ) and dendritic cells (DCs) and further combined the nonspecific β-blocker propranolol with a peptide cancer vaccine for the treatment of melanoma in mice. We report that norepinephrine treatment dramatically altered moMФ cytokine production, whereas DCs were unresponsive to norepinephrine and critically lack β2-adrenergic receptor expression. In addition, we show that propranolol plus cancer vaccine enhanced peripheral DC maturation, increased the intratumor proportion of effector CD8+ T cells, and decreased the presence of intratumor PD-L1+ myeloid-derived suppressor cells. Furthermore, this combination dramatically reduced tumor growth compared with vaccination alone. Taken together, these results offer insights into the cell-specific manner by which the SNS regulates the APC immune compartment and provide strong support for the use of propranolol in combination with cancer vaccines to improve patient response rates and survival.
Collapse
Affiliation(s)
- Louis Hinkle
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX.,Texas A&M Health Science Center, Bryan, TX
| | - Yongbin Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX
| | - Chaoyang Meng
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX.,Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhe Chen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX.,Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Junhua Mai
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX
| | - Licheng Zhang
- Center for Cancer Immunotherapy, Houston Methodist Research Institute, Houston, TX; and
| | - Yitian Xu
- Center for Cancer Immunotherapy, Houston Methodist Research Institute, Houston, TX; and
| | - Ping-Ying Pan
- Center for Cancer Immunotherapy, Houston Methodist Research Institute, Houston, TX; and.,Weill Cornell Medical College, New York, NY
| | - Shu-Hsia Chen
- Center for Cancer Immunotherapy, Houston Methodist Research Institute, Houston, TX; and.,Weill Cornell Medical College, New York, NY
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX; .,Weill Cornell Medical College, New York, NY
| |
Collapse
|
13
|
Characterization of M116.1p, a murine cytomegalovirus protein required for efficient infection of mononuclear phagocytes. J Virol 2021; 96:e0087621. [PMID: 34705561 DOI: 10.1128/jvi.00876-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Broad tissue tropism of cytomegaloviruses (CMVs) is facilitated by different glycoprotein entry complexes, which are conserved between human CMV (HCMV) and murine CMV (MCMV). Among the wide array of cell types susceptible to the infection, mononuclear phagocytes (MNPs) play a unique role in the pathogenesis of the infection as they contribute both to the virus spread and immune control. CMVs have dedicated numerous genes for the efficient infection and evasion of macrophages and dendritic cells. In this study, we have characterized the properties and function of M116, a previously poorly described but highly transcribed MCMV gene region which encodes M116.1p, a novel protein necessary for the efficient infection of MNPs and viral spread in vivo. Our study further revealed that M116.1p shares similarities with its positional homologs in HCMV and RCMV, UL116 and R116, respectively, such as late kinetics of expression, N-glycosylation, localization to the virion assembly compartment, and interaction with gH - a member of the CMVs fusion complex. This study, therefore, expands our knowledge about virally encoded glycoproteins that play important roles in viral infectivity and tropism. Importance Human cytomegalovirus (HCMV) is a species-specific herpesvirus that causes severe disease in immunocompromised individuals and immunologically immature neonates. Murine cytomegalovirus (MCMV) is biologically similar to HCMV, and it serves as a widely used model for studying the infection, pathogenesis, and immune responses to HCMV. In our previous work, we have identified the M116 ORF as one of the most extensively transcribed regions of the MCMV genome without an assigned function. This study shows that the M116 locus codes for a novel protein, M116.1p, which shares similarities with UL116 and R116 in HCMV and RCMV, respectively, and is required for the efficient infection of mononuclear phagocytes and virus spread in vivo. Furthermore, this study establishes the α-M116 monoclonal antibody and MCMV mutants lacking M116, generated in this work, as valuable tools for studying the role of macrophages and dendritic cells in limiting CMV infection following different MCMV administration routes.
Collapse
|
14
|
Bang YJ, Hu Z, Li Y, Gattu S, Ruhn KA, Raj P, Herz J, Hooper LV. Serum amyloid A delivers retinol to intestinal myeloid cells to promote adaptive immunity. Science 2021; 373:eabf9232. [PMID: 34529485 DOI: 10.1126/science.abf9232] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Ye-Ji Bang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zehan Hu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yun Li
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sureka Gattu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kelly A Ruhn
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Prithvi Raj
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lora V Hooper
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
15
|
Saitoh S, Van Wijk K, Nakajima O. Crosstalk between Metabolic Disorders and Immune Cells. Int J Mol Sci 2021; 22:ijms221810017. [PMID: 34576181 PMCID: PMC8469678 DOI: 10.3390/ijms221810017] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/08/2021] [Accepted: 09/15/2021] [Indexed: 12/22/2022] Open
Abstract
Metabolic syndrome results from multiple risk factors that arise from insulin resistance induced by abnormal fat deposition. Chronic inflammation owing to obesity primarily results from the recruitment of pro-inflammatory M1 macrophages into the adipose tissue stroma, as the adipocytes within become hypertrophied. During obesity-induced inflammation in adipose tissue, pro-inflammatory cytokines are produced by macrophages and recruit further pro-inflammatory immune cells into the adipose tissue to boost the immune response. Here, we provide an overview of the biology of macrophages in adipose tissue and the relationship between other immune cells, such as CD4+ T cells, natural killer cells, and innate lymphoid cells, and obesity and type 2 diabetes. Finally, we discuss the link between the human pathology and immune response and metabolism and further highlight potential therapeutic targets for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Shinichi Saitoh
- Department of Immunology, Yamagata University Faculty of Medicine, Yamagata 990-9585, Japan;
| | - Koen Van Wijk
- Research Center for Molecular Genetics, Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine, Yamagata 990-9585, Japan;
| | - Osamu Nakajima
- Research Center for Molecular Genetics, Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine, Yamagata 990-9585, Japan;
- Correspondence:
| |
Collapse
|
16
|
Pointner L, Kraiem A, Thaler M, Richter F, Wenger M, Bethanis A, Klotz M, Traidl-Hoffmann C, Gilles S, Aglas L. Birch Pollen Induces Toll-Like Receptor 4-Dependent Dendritic Cell Activation Favoring T Cell Responses. FRONTIERS IN ALLERGY 2021; 2:680937. [PMID: 35386993 PMCID: PMC8974861 DOI: 10.3389/falgy.2021.680937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
Seasonal exposure to birch pollen (BP) is a major cause of pollinosis. The specific role of Toll-like receptor 4 (TLR4) in BP-induced allergic inflammation and the identification of key factors in birch pollen extracts (BPE) initiating this process remain to be explored. This study aimed to examine (i) the importance of TLR4 for dendritic cell (DC) activation by BPE, (ii) the extent of the contribution of BPE-derived lipopolysaccharide (LPS) and other potential TLR4 adjuvant(s) in BPE, and (iii) the relevance of the TLR4-dependent activation of BPE-stimulated DCs in the initiation of an adaptive immune response. In vitro, activation of murine bone marrow-derived DCs (BMDCs) and human monocyte-derived DCs by BPE or the equivalent LPS (nLPS) was analyzed by flow cytometry. Polymyxin B (PMB), a TLR4 antagonist and TLR4-deficient BMDCs were used to investigate the TLR4 signaling in DC activation. The immunostimulatory activity of BPE was compared to protein-/lipid-depleted BPE-fractions. In co-cultures of BPE-pulsed BMDCs and Bet v 1-specific hybridoma T cells, the influence of the TLR4-dependent DC activation on T cell activation was analyzed. In vivo immunization of IL-4 reporter mice was conducted to study BPE-induced Th2 polarization upon PMB pre-treatment. Murine and human DC activation induced by either BPE or nLPS was inhibited by the TLR4 antagonist or by PMB, and abrogated in TLR4-deficient BMDCs compared to wild-type BMDCs. The lipid-free but not the protein-free fraction showed a reduced capacity to activate the TLR4 signaling and murine DCs. In human DCs, nLPS only partially reproduced the BPE-induced activation intensity. BPE-primed BMDCs efficiently stimulated T cell activation, which was repressed by the TLR4 antagonist or PMB, and the addition of nLPS to Bet v 1 did not reproduce the effect of BPE. In vivo, immunization with BPE induced a significant Th2 polarization, whereas administration of BPE pre-incubated with PMB showed a decreased tendency. These findings suggest that TLR4 is a major pathway by which BPE triggers DC activation that is involved in the initiation of adaptive immune responses. Further characterization of these BP-derived TLR4 adjuvants could provide new candidates for therapeutic strategies targeting specific mechanisms in BP-induced allergic inflammation.
Collapse
Affiliation(s)
- Lisa Pointner
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Amin Kraiem
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Michael Thaler
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Fabian Richter
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Mario Wenger
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | | | - Markus Klotz
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Claudia Traidl-Hoffmann
- Chair of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, Neuherberg, Germany
- Christine Kühne 96 Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Stefanie Gilles
- Chair of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, Neuherberg, Germany
| | - Lorenz Aglas
- Department of Biosciences, University of Salzburg, Salzburg, Austria
- *Correspondence: Lorenz Aglas
| |
Collapse
|
17
|
Combination Adjuvants Affect the Magnitude of Effector-Like Memory CD8 T Cells and Protection against Listeriosis. Infect Immun 2021; 89:e0076820. [PMID: 33782151 DOI: 10.1128/iai.00768-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The development of T cell-based subunit protein vaccines against diseases such as tuberculosis and malaria remains a challenge for immunologists. Here, we have identified a nanoemulsion adjuvant, Adjuplex (ADJ), which enhanced dendritic cell (DC) cross-presentation and elicited effective memory T cell-based immunity to Listeria monocytogenes. We further evaluated whether cross-presentation induced by ADJ can be combined with the immunomodulatory effects of Toll-like receptor (TLR) agonists (CpG or glucopyranosyl lipid adjuvant [GLA]) to evoke systemic CD8 T cell-based immunity to L. monocytogenes. Mechanistically, vaccination with ADJ, alone or in combination with CpG or GLA, augmented activation and antigen uptake by CD103+ migratory and CD8α+ resident DCs and upregulated CD69 expression on B and T lymphocytes in vaccine-draining lymph nodes. By engaging basic leucine zipper ATF-like transcription factor 3-dependent cross-presenting DCs, ADJ potently elicited effector CD8 T cells that differentiated into granzyme B-expressing CD27LO effector-like memory CD8 T cells, which provided effective immunity to L. monocytogenes in the spleen and liver. CpG or GLA alone did not elicit effector-like memory CD8 T cells and induced moderate protection in the spleen but not in the liver. Surprisingly, combining CpG or GLA with ADJ reduced the number of ADJ-induced memory CD8 T cells and compromised protective immunity to L. monocytogenes, especially in the liver. Taken together, the data presented in this study provide a glimpse of protective CD8 T cell memory differentiation induced by a nanoemulsion adjuvant and demonstrate the unexpected negative effects of TLR signaling on the magnitude of CD8 T cell memory and protective immunity to L. monocytogenes, a model intracellular pathogen.
Collapse
|
18
|
Robinson EK, Covarrubias S, Zhou S, Carpenter S. Generation and utilization of a HEK-293T murine GM-CSF expressing cell line. PLoS One 2021; 16:e0249117. [PMID: 33836009 PMCID: PMC8034741 DOI: 10.1371/journal.pone.0249117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 03/11/2021] [Indexed: 12/13/2022] Open
Abstract
Macrophages and dendritic cells (DCs) are innate immune cells that play a key role in defense against pathogens. In vitro cultures of bone marrow-derived macrophages (BMDMs) and dendritic cells (BMDCs) are well-established and valuable methods for immunological studies. Typically, commercially available recombinant GM-CSF is utilized to generate BMDCs and is also used to culture alveolar macrophages. We have generated a new HEK-293T cell line expressing murine GM-CSF that secretes high levels of GM-CSF (~180 ng/ml) into complete media as an alternative to commercial GM-CSF. Differentiation of dendritic cells and expression of various markers were kinetically assessed using the GM-CSF HEK293T cell line, termed supGM-CSF and compared directly to purified commercial GMCSF. After 7–9 days of cell culture the supGM-CSF yielded twice as many viable cells compared to the commercial purified GM-CSF. In addition to differentiating BMDCs, the supGM-CSF can be utilized to culture functionally active alveolar macrophages. Collectively, our results show that supernatant from our GM-CSF HEK293T cell line supports the differentiation of mouse BMDCs or alveolar macrophage culturing, providing an economical alternative to purified GM-CSF.
Collapse
Affiliation(s)
- Elektra Kantzari Robinson
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, United States of America
| | - Sergio Covarrubias
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, United States of America
| | - Simon Zhou
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, United States of America
| | - Susan Carpenter
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, United States of America
- * E-mail:
| |
Collapse
|
19
|
Bednarczyk M, Medina-Montano C, Fittler FJ, Stege H, Roskamp M, Kuske M, Langer C, Vahldieck M, Montermann E, Tubbe I, Röhrig N, Dzionek A, Grabbe S, Bros M. Complement-Opsonized Nano-Carriers Are Bound by Dendritic Cells (DC) via Complement Receptor (CR)3, and by B Cell Subpopulations via CR-1/2, and Affect the Activation of DC and B-1 Cells. Int J Mol Sci 2021; 22:2869. [PMID: 33799879 PMCID: PMC8001596 DOI: 10.3390/ijms22062869] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/22/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
The development of nanocarriers (NC) for biomedical applications has gained large interest due to their potential to co-deliver drugs in a cell-type-targeting manner. However, depending on their surface characteristics, NC accumulate serum factors, termed protein corona, which may affect their cellular binding. We have previously shown that NC coated with carbohydrates to enable biocompatibility triggered the lectin-dependent complement pathway, resulting in enhanced binding to B cells via complement receptor (CR)1/2. Here we show that such NC also engaged all types of splenic leukocytes known to express CR3 at a high rate when NC were pre-incubated with native mouse serum resulting in complement opsonization. By focusing on dendritic cells (DC) as an important antigen-presenting cell type, we show that CR3 was essential for binding/uptake of complement-opsonized NC, whereas CR4, which in mouse is specifically expressed by DC, played no role. Further, a minor B cell subpopulation (B-1), which is important for first-line pathogen responses, and co-expressed CR1/2 and CR3, in general, engaged NC to a much higher extent than normal B cells. Here, we identified CR-1/2 as necessary for binding of complement-opsonized NC, whereas CR3 was dispensable. Interestingly, the binding of complement-opsonized NC to both DC and B-1 cells affected the expression of activation markers. Our findings may have important implications for the design of nano-vaccines against infectious diseases, which codeliver pathogen-specific protein antigen and adjuvant, aimed to induce a broad adaptive cellular and humoral immune response by inducing cytotoxic T lymphocytes that kill infected cells and pathogen-neutralizing antibodies, respectively. Decoration of nano-vaccines either with carbohydrates to trigger complement activation in vivo or with active complement may result in concomitant targeting of DC and B cells and thereby may strongly enhance the extent of dual cellular/humoral immune responses.
Collapse
Affiliation(s)
- Monika Bednarczyk
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Carolina Medina-Montano
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Frederic Julien Fittler
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Henner Stege
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Meike Roskamp
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany; (M.R.); (C.L.); (M.V.); (A.D.)
| | - Michael Kuske
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Christian Langer
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany; (M.R.); (C.L.); (M.V.); (A.D.)
| | - Marco Vahldieck
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany; (M.R.); (C.L.); (M.V.); (A.D.)
| | - Evelyn Montermann
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Ingrid Tubbe
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Nadine Röhrig
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Andrzej Dzionek
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany; (M.R.); (C.L.); (M.V.); (A.D.)
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| |
Collapse
|
20
|
Du F, Liu B, Zhang S. COVID-19: the role of excessive cytokine release and potential ACE2 down-regulation in promoting hypercoagulable state associated with severe illness. J Thromb Thrombolysis 2021; 51:313-329. [PMID: 32676883 PMCID: PMC7365308 DOI: 10.1007/s11239-020-02224-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The novel coronavirus disease (COVID-19) has become a universally prevalent infectious disease. The causative virus of COVID-19 is severe acute respiratory syndrome coronavirus type 2. Recent retrospective clinical studies have established a significant association between the incidence of vascular thrombotic events and the severity of COVID-19. The enhancement in serum levels of markers that reflect a hypercoagulable state has been suggested to indicate a poor prognosis. Therefore, at present, it is crucial to understand the mechanisms that foster the hypercoagulable state in COVID-19. Over-activated inflammatory response, which is manifested as excessive cytokine release in COVID-19 patients, is also associated with COVID-19 severity. This review discusses the immuno-pathological basis of the excessive cytokine release in COVID-19. Besides, this article reviews the role of pro-inflammatory or anti-inflammatory cytokines, whose significant elevations in their serum levels have been consistently detected in multiple different clinical studies, in promoting the hypercoagulable state. Since the expression of angiotensin-converting enzyme 2 (ACE2) is potentially down-regulated in COVID-19, as proposed by a recent bio-informatic analysis, mechanisms through which reduced ACE2 expressions promote vascular thrombosis are summarized. In addition, the reciprocal-enhancing effects of the excessive cytokine release and the downregulated ACE2 expression on their pro-thrombotic activities are further discussed. Here, based on currently available evidence, we review the pathogenic mechanisms of the hypercoagulable state associated with severe cases of COVID-19 to give insights into prevention and treatment of the vascular thrombotic events in COVID-19.
Collapse
Affiliation(s)
- Fenghe Du
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1st, Dongcheng District, Beijing, 100730, China.,Four-Year Program of Clinical Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Bao Liu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1st, Dongcheng District, Beijing, 100730, China.
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Shuaifuyuan 1st, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
21
|
Duhan V, Khairnar V, Kitanovski S, Hamdan TA, Klein AD, Lang J, Ali M, Adomati T, Bhat H, Friedrich SK, Li F, Krebs P, Futerman AH, Addo MM, Hardt C, Hoffmann D, Lang PA, Lang KS. Integrin Alpha E (CD103) Limits Virus-Induced IFN-I Production in Conventional Dendritic Cells. Front Immunol 2021; 11:607889. [PMID: 33584680 PMCID: PMC7873973 DOI: 10.3389/fimmu.2020.607889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/14/2020] [Indexed: 11/17/2022] Open
Abstract
Early and strong production of IFN-I by dendritic cells is important to control vesicular stomatitis virus (VSV), however mechanisms which explain this cell-type specific innate immune activation remain to be defined. Here, using a genome wide association study (GWAS), we identified Integrin alpha-E (Itgae, CD103) as a new regulator of antiviral IFN-I production in a mouse model of vesicular stomatitis virus (VSV) infection. CD103 was specifically expressed by splenic conventional dendritic cells (cDCs) and limited IFN-I production in these cells during VSV infection. Mechanistically, CD103 suppressed AKT phosphorylation and mTOR activation in DCs. Deficiency in CD103 accelerated early IFN-I in cDCs and prevented death in VSV infected animals. In conclusion, CD103 participates in regulation of cDC specific IFN-I induction and thereby influences immune activation after VSV infection.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Cells, Cultured
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/virology
- Disease Models, Animal
- Genome-Wide Association Study
- Host-Pathogen Interactions
- Immunity, Innate
- Integrin alpha Chains/genetics
- Integrin alpha Chains/metabolism
- Interferon Type I/metabolism
- Mice, 129 Strain
- Mice, Inbred AKR
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Inbred NOD
- Mice, Knockout
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor, Interferon alpha-beta/genetics
- Receptor, Interferon alpha-beta/metabolism
- Signal Transduction
- TOR Serine-Threonine Kinases/metabolism
- Vesicular Stomatitis/genetics
- Vesicular Stomatitis/immunology
- Vesicular Stomatitis/metabolism
- Vesicular Stomatitis/virology
- Vesiculovirus/growth & development
- Vesiculovirus/pathogenicity
- Virus Replication
- Mice
Collapse
Affiliation(s)
- Vikas Duhan
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Essen, Germany
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Vishal Khairnar
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Essen, Germany
- Dana-Farber Cancer Institute, Harvard University, Boston, MA, United States
| | - Simo Kitanovski
- Bioinformatics and Computational Biophysics, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Thamer A. Hamdan
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Essen, Germany
- Department of Medical Laboratories, Faculty of Health Sciences, American University of Madaba, Amman, Jordan
| | - Andrés D. Klein
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
- Centro de Genética y Genómica, Universidad Del Desarrollo Clínica Alemana de Santiago, Santiago, Chile
| | - Judith Lang
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Murtaza Ali
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Tom Adomati
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Hilal Bhat
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Essen, Germany
- Center for Molecular Medicine Cologne, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Sarah-Kim Friedrich
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Fanghui Li
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Philippe Krebs
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Anthony H. Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Marylyn M. Addo
- University Medical Center Hamburg-Eppendorf, Division of Infectious Diseases, 1st Department of Medicine, Hamburg, Germany
- German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riemse, Hamburg, Germany
- Department of Clinical Immunology of Infectious Diseases, Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, Germany
| | - Cornelia Hardt
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Daniel Hoffmann
- Bioinformatics and Computational Biophysics, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Philipp A. Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Karl S. Lang
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
22
|
Lee W, Kingstad-Bakke B, Paulson B, Larsen A, Overmyer K, Marinaik CB, Dulli K, Toy R, Vogel G, Mueller KP, Tweed K, Walsh AJ, Russell J, Saha K, Reyes L, Skala MC, Sauer JD, Shayakhmetov DM, Coon J, Roy K, Suresh M. Carbomer-based adjuvant elicits CD8 T-cell immunity by inducing a distinct metabolic state in cross-presenting dendritic cells. PLoS Pathog 2021; 17:e1009168. [PMID: 33444400 PMCID: PMC7840022 DOI: 10.1371/journal.ppat.1009168] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 01/27/2021] [Accepted: 11/16/2020] [Indexed: 01/25/2023] Open
Abstract
There is a critical need for adjuvants that can safely elicit potent and durable T cell-based immunity to intracellular pathogens. Here, we report that parenteral vaccination with a carbomer-based adjuvant, Adjuplex (ADJ), stimulated robust CD8 T-cell responses to subunit antigens and afforded effective immunity against respiratory challenge with a virus and a systemic intracellular bacterial infection. Studies to understand the metabolic and molecular basis for ADJ's effect on antigen cross-presentation by dendritic cells (DCs) revealed several unique and distinctive mechanisms. ADJ-stimulated DCs produced IL-1β and IL-18, suggestive of inflammasome activation, but in vivo activation of CD8 T cells was unaffected in caspase 1-deficient mice. Cross-presentation induced by TLR agonists requires a critical switch to anabolic metabolism, but ADJ enhanced cross presentation without this metabolic switch in DCs. Instead, ADJ induced in DCs, an unique metabolic state, typified by dampened oxidative phosphorylation and basal levels of glycolysis. In the absence of increased glycolytic flux, ADJ modulated multiple steps in the cytosolic pathway of cross-presentation by enabling accumulation of degraded antigen, reducing endosomal acidity and promoting antigen localization to early endosomes. Further, by increasing ROS production and lipid peroxidation, ADJ promoted antigen escape from endosomes to the cytosol for degradation by proteasomes into peptides for MHC I loading by TAP-dependent pathways. Furthermore, we found that induction of lipid bodies (LBs) and alterations in LB composition mediated by ADJ were also critical for DC cross-presentation. Collectively, our model challenges the prevailing metabolic paradigm by suggesting that DCs can perform effective DC cross-presentation, independent of glycolysis to induce robust T cell-dependent protective immunity to intracellular pathogens. These findings have strong implications in the rational development of safe and effective immune adjuvants to potentiate robust T-cell based immunity.
Collapse
Affiliation(s)
- Woojong Lee
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Brock Kingstad-Bakke
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Brett Paulson
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Autumn Larsen
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Katherine Overmyer
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Chandranaik B. Marinaik
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kelly Dulli
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Randall Toy
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University and The Parker H. Petit Institute for Bioengineering and Biosciences, Center for ImmunoEngineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Gabriela Vogel
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University and The Parker H. Petit Institute for Bioengineering and Biosciences, Center for ImmunoEngineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Katherine P. Mueller
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kelsey Tweed
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Alex J. Walsh
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jason Russell
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Leticia Reyes
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Melissa C. Skala
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Dmitry M. Shayakhmetov
- Lowance Center for Human Immunology, Emory Vaccine Center, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Joshua Coon
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Krishnendu Roy
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University and The Parker H. Petit Institute for Bioengineering and Biosciences, Center for ImmunoEngineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - M. Suresh
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
23
|
Pradhan P, Toy R, Jhita N, Atalis A, Pandey B, Beach A, Blanchard EL, Moore SG, Gaul DA, Santangelo PJ, Shayakhmetov DM, Roy K. TRAF6-IRF5 kinetics, TRIF, and biophysical factors drive synergistic innate responses to particle-mediated MPLA-CpG co-presentation. SCIENCE ADVANCES 2021; 7:eabd4235. [PMID: 33523878 PMCID: PMC7806213 DOI: 10.1126/sciadv.abd4235] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 11/18/2020] [Indexed: 05/21/2023]
Abstract
Innate immune responses to pathogens are driven by co-presentation of multiple pathogen-associated molecular patterns (PAMPs). Combinations of PAMPs can trigger synergistic immune responses, but the underlying molecular mechanisms of synergy are poorly understood. Here, we used synthetic particulate carriers co-loaded with monophosphoryl lipid A (MPLA) and CpG as pathogen-like particles (PLPs) to dissect the signaling pathways responsible for dual adjuvant immune responses. PLP-based co-delivery of MPLA and CpG to GM-CSF-driven mouse bone marrow-derived antigen-presenting cells (BM-APCs) elicited synergistic interferon-β (IFN-β) and interleukin-12p70 (IL-12p70) responses, which were strongly influenced by the biophysical properties of PLPs. Mechanistically, we found that MyD88 and interferon regulatory factor 5 (IRF5) were necessary for IFN-β and IL-12p70 production, while TRIF signaling was required for the synergistic response. Both the kinetics and magnitude of downstream TRAF6 and IRF5 signaling drove the synergy. These results identify the key mechanisms of synergistic Toll-like receptor 4 (TLR4)-TLR9 co-signaling in mouse BM-APCs and underscore the critical role of signaling kinetics and biophysical properties on the integrated response to combination adjuvants.
Collapse
Affiliation(s)
- P Pradhan
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Georgia Institute of Technology, Atlanta, GA, USA
| | - R Toy
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - N Jhita
- Lowance Center of Human Immunology, Department of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - A Atalis
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - B Pandey
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - A Beach
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - E L Blanchard
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - S G Moore
- The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - D A Gaul
- The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - P J Santangelo
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - D M Shayakhmetov
- Lowance Center of Human Immunology, Department of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - K Roy
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
24
|
Janke LJ, Imai DM, Tillman H, Doty R, Hoenerhoff MJ, Xu JJ, Freeman Z, Allen P, Fowlkes NW, Iacobucci I, Dickerson K, Mullighan CG, Vogel P, Rehg JE. Development of Mast Cell and Eosinophil Hyperplasia and HLH/MAS-Like Disease in NSG-SGM3 Mice Receiving Human CD34+ Hematopoietic Stem Cells or Patient-Derived Leukemia Xenografts. Vet Pathol 2021; 58:181-204. [PMID: 33208054 PMCID: PMC8414369 DOI: 10.1177/0300985820970144] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Immunocompromised mouse strains expressing human transgenes are being increasingly used in biomedical research. The genetic modifications in these mice cause various cellular responses, resulting in histologic features unique to each strain. The NSG-SGM3 mouse strain is similar to the commonly used NSG (NOD scid gamma) strain but expresses human transgenes encoding stem cell factor (also known as KIT ligand), granulocyte-macrophage colony-stimulating factor, and interleukin 3. This report describes 3 histopathologic features seen in these mice when they are unmanipulated or after transplantation with human CD34+ hematopoietic stem cells (HSCs), virally transduced hCD34+ HSCs, or a leukemia patient-derived xenograft. The first feature is mast cell hyperplasia: unmanipulated, naïve mice develop periductular pancreatic aggregates of murine mast cells, whereas mice given the aforementioned human cells develop a proliferative infiltrative interstitial pancreatic mast cell hyperplasia but with human mast cells. The second feature is the predisposition of NSG-SGM3 mice given these human cells to develop eosinophil hyperplasia. The third feature, secondary hemophagocytic lymphohistiocytosis/macrophage activation syndrome (HLH/MAS)-like disease, is the most pronounced in both its clinical and histopathologic presentations. As part of this disease, a small number of mice also have histiocytic infiltration of the brain and spinal cord with subsequent neurologic or vestibular signs. The presence of any of these features can confound accurate histopathologic interpretation; therefore, it is important to recognize them as strain characteristics and to differentiate them from what may be experimentally induced in the model being studied.
Collapse
Affiliation(s)
- Laura J. Janke
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Denise M. Imai
- Comparative Pathology Laboratory, University of California, Davis, California, USA
| | - Heather Tillman
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | | | - Mark J. Hoenerhoff
- In Vivo Animal Core, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jiajie J. Xu
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Zach Freeman
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Portia Allen
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Natalie Wall Fowlkes
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ilaria Iacobucci
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Kirsten Dickerson
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Charles G. Mullighan
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Hematological Malignancies Program, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Peter Vogel
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jerold E. Rehg
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
25
|
Delbridge ARD, Huh D, Brickelmaier M, Burns JC, Roberts C, Challa R, Raymond N, Cullen P, Carlile TM, Ennis KA, Liu M, Sun C, Allaire NE, Foos M, Tsai HH, Franchimont N, Ransohoff RM, Butts C, Mingueneau M. Organotypic Brain Slice Culture Microglia Exhibit Molecular Similarity to Acutely-Isolated Adult Microglia and Provide a Platform to Study Neuroinflammation. Front Cell Neurosci 2020; 14:592005. [PMID: 33473245 PMCID: PMC7812919 DOI: 10.3389/fncel.2020.592005] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
Microglia are central nervous system (CNS) resident immune cells that have been implicated in neuroinflammatory pathogenesis of a variety of neurological conditions. Their manifold context-dependent contributions to neuroinflammation are only beginning to be elucidated, which can be attributed in part to the challenges of studying microglia in vivo and the lack of tractable in vitro systems to study microglia function. Organotypic brain slice cultures offer a tissue-relevant context that enables the study of CNS resident cells and the analysis of brain slice microglial phenotypes has provided important insights, in particular into neuroprotective functions. Here we use RNA sequencing, direct digital quantification of gene expression with nCounter® technology and targeted analysis of individual microglial signature genes, to characterize brain slice microglia relative to acutely-isolated counterparts and 2-dimensional (2D) primary microglia cultures, a widely used in vitro surrogate. Analysis using single cell and population-based methods found brain slice microglia exhibited better preservation of canonical microglia markers and overall gene expression with stronger fidelity to acutely-isolated adult microglia, relative to in vitro cells. We characterized the dynamic phenotypic changes of brain slice microglia over time, after plating in culture. Mechanical damage associated with slice preparation prompted an initial period of inflammation, which resolved over time. Based on flow cytometry and gene expression profiling we identified the 2-week timepoint as optimal for investigation of microglia responses to exogenously-applied stimuli as exemplified by treatment-induced neuroinflammatory changes observed in microglia following LPS, TNF and GM-CSF addition to the culture medium. Altogether these findings indicate that brain slice cultures provide an experimental system superior to in vitro culture of microglia as a surrogate to investigate microglia functions, and the impact of soluble factors and cellular context on their physiology.
Collapse
Affiliation(s)
- Alex R D Delbridge
- Multiple Sclerosis and Neuroimmunology Research Unit, Biogen, Cambridge, MA, United States.,Biogen Postdoctoral Scientist Program, Biogen, Cambridge, MA, United States
| | - Dann Huh
- Translational Biology, Biogen, Cambridge, MA, United States
| | - Margot Brickelmaier
- Multiple Sclerosis and Neuroimmunology Research Unit, Biogen, Cambridge, MA, United States
| | - Jeremy C Burns
- Multiple Sclerosis and Neuroimmunology Research Unit, Biogen, Cambridge, MA, United States
| | - Chris Roberts
- Translational Biology, Biogen, Cambridge, MA, United States
| | - Ravi Challa
- Translational Biology, Biogen, Cambridge, MA, United States
| | - Naideline Raymond
- Multiple Sclerosis and Neuroimmunology Research Unit, Biogen, Cambridge, MA, United States
| | - Patrick Cullen
- Translational Biology, Biogen, Cambridge, MA, United States
| | | | - Katelin A Ennis
- Genetic and Neurodevelopmental Disorders, Biogen, Cambridge, MA, United States
| | - Mei Liu
- Biogen Postdoctoral Scientist Program, Biogen, Cambridge, MA, United States
| | - Chao Sun
- Biogen Postdoctoral Scientist Program, Biogen, Cambridge, MA, United States
| | - Normand E Allaire
- Biogen Postdoctoral Scientist Program, Biogen, Cambridge, MA, United States
| | - Marianna Foos
- Biogen Postdoctoral Scientist Program, Biogen, Cambridge, MA, United States
| | - Hui-Hsin Tsai
- Multiple Sclerosis and Neuroimmunology Research Unit, Biogen, Cambridge, MA, United States
| | | | - Richard M Ransohoff
- Multiple Sclerosis and Neuroimmunology Research Unit, Biogen, Cambridge, MA, United States
| | - Cherie Butts
- Digital & Quantitative Medicine, Biogen, Cambridge, MA, United States
| | - Michael Mingueneau
- Multiple Sclerosis and Neuroimmunology Research Unit, Biogen, Cambridge, MA, United States
| |
Collapse
|
26
|
Alothaimeen T, Trus E, Basta S, Gee K. Differential TLR7-mediated cytokine expression by R848 in M-CSF- versus GM-CSF-derived macrophages after LCMV infection. J Gen Virol 2020; 102. [PMID: 33331816 PMCID: PMC8515861 DOI: 10.1099/jgv.0.001541] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) and macrophage colony-stimulating factor (M-CSF) play an important role in macrophage (MФ) development by influencing their differentiation and polarization. Our goal was to explore the difference between M-CSF- and GM-CSF-derived bone marrow MФ responsiveness to TLR7-mediated signalling pathways that influence cytokine production early after infection in a model of acute virus infection. To do so, we examined cytokine production and TLR7-mediated signalling at 1 h post-lymphocytic choriomeningitis virus (LCMV) Armstrong (ARM) infection. We found that R848-induced cytokine expression was enhanced in these cells, with GM-CSF cells exhibiting higher proinflammatory cytokine expression and M-CSF cells exhibiting higher anti-inflammatory cytokine expression. However, R848-mediated signalling molecule activation was diminished in LCMV-infected M-CSF and GM-CSF macrophages. Interestingly, we observed that TLR7 expression was maintained during LCMV infection of M-CSF and GM-CSF cells. Moreover, TLR7 expression was significantly higher in M-CSF cells compared to GM-CSF cells. Taken together, our data demonstrate that although LCMV restrains early TLR7-mediated signalling, it primes differentiated MФ to enhance expression of their respective cytokine profiles and maintains levels of TLR7 expression early after infection.
Collapse
Affiliation(s)
- Torki Alothaimeen
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Evan Trus
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Sameh Basta
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Katrina Gee
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| |
Collapse
|
27
|
Toll-Like Receptor 2-Tpl2-Dependent ERK Signaling Drives Inverse Interleukin 12 Regulation in Dendritic Cells and Macrophages. Infect Immun 2020; 89:IAI.00323-20. [PMID: 33077627 DOI: 10.1128/iai.00323-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 10/14/2020] [Indexed: 01/22/2023] Open
Abstract
This study investigated responses to Toll-like receptor 2 (TLR2)-driven extracellular signal-related kinase (ERK) signaling in dendritic cells (DCs) versus macrophages. TLR2 signaling was induced with Pam3Cys-Ser-Lys4, and the role of ERK signaling was interrogated pharmacologically with MEK1/2 inhibitor U0126 or genetically with bone marrow-derived macrophages or DCs from Tpl2-/- mice. We assessed cytokine production via enzyme-linked immunosorbent assay (ELISA) or V-Plex, and mRNA levels were assessed via reverse transcriptase quantitative PCR (qRT-PCR). In macrophages, blockade of ERK signaling by pharmacologic or genetic approaches inhibited interleukin 10 (IL-10) expression and increased expression of the p40 subunit shared by IL-12 and IL-23 (IL-12/23p40). In DCs, blockade of ERK signaling similarly inhibited IL-10 expression but decreased IL-12/23p40 expression, which is opposite to the effect of ERK signaling blockade on IL-12/23p40 in macrophages. This difference in IL-12/23p40 regulation correlated with the differential expression of transcription factors cFos and IRF1, which are known to regulate IL-12 family members, including IL-12 and IL-23. Thus, the impact of ERK signaling in response to TLR2 stimulation differs between macrophages and DCs, potentially regulating their distinctive functions in the immune system. ERK-mediated suppression of IL-12/23p40 in macrophages may prevent excessive inflammation and associated tissue damage following TLR2-stimulation, while ERK-mediated induction of IL-12/23p40 in DCs may promote priming of T helper 1 (Th1) responses. A greater understanding of the role that ERK signaling plays in different immune cell types may inform the development of host-directed therapy and optimal adjuvanticity for a number of infectious pathogens.
Collapse
|
28
|
Tremblay ML, O’Brien-Moran Z, Rioux JA, Nuschke A, Davis C, Kast WM, Weir G, Stanford M, Brewer KD. Quantitative MRI cell tracking of immune cell recruitment to tumors and draining lymph nodes in response to anti-PD-1 and a DPX-based immunotherapy. Oncoimmunology 2020; 9:1851539. [PMID: 33299663 PMCID: PMC7714509 DOI: 10.1080/2162402x.2020.1851539] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 11/01/2022] Open
Abstract
DPX is a unique T cell activating formulation that generates robust immune responses (both clinically and preclinically) which can be tailored to various cancers via the use of tumor-specific antigens and adjuvants. While DPX-based immunotherapies may act complementary with checkpoint inhibitors, combination therapy is not always easily predictable based on individual therapeutic responses. Optimizing these combinations can be improved by understanding the mechanism of action underlying the individual therapies. Magnetic Resonance Imaging (MRI) allows tracking of cells labeled with superparamagnetic iron oxide (SPIO), which can yield valuable information about the localization of crucial immune cell subsets. In this work, we evaluated the use of a multi-echo, single point MRI pulse sequence, TurboSPI, for tracking and quantifying cytotoxic T lymphocytes (CTLs) and myeloid lineage cells (MLCs). In a subcutaneous cervical cancer model (C3) we compared untreated mice to mice treated with either a single therapy (anti-PD-1 or DPX-R9F) or a combination of both therapies. We were able to detect, using TurboSPI, significant increases in CTL recruitment dynamics in response to combination therapy. We also observed differences in MLC recruitment to therapy-draining (DPX-R9F) lymph nodes in response to treatment with DPX-R9F (alone or in combination with anti-PD-1). We demonstrated that the therapies presented herein induced time-varying changes in cell recruitment. This work establishes that these quantitative molecular MRI techniques can be expanded to study a number of cancer and immunotherapy combinations to improve our understanding of longitudinal immunological changes and mechanisms of action.
Collapse
Affiliation(s)
- Marie-Laurence Tremblay
- Biomedical Translational Imaging Centre (BIOTIC, IWK Health Centre, Halifax, NS, Canada
- Dalhousie University, Halifax, NS, Canada
- IMV Inc, Halifax, NS, Canada
| | - Zoe O’Brien-Moran
- Biomedical Translational Imaging Centre (BIOTIC, IWK Health Centre, Halifax, NS, Canada
- Department of Physics, Dalhousie University, Halifax, NS, Canada
| | - James A. Rioux
- Biomedical Translational Imaging Centre (BIOTIC, IWK Health Centre, Halifax, NS, Canada
- Department of Physics, Dalhousie University, Halifax, NS, Canada
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS, Canada
| | - Andrea Nuschke
- Biomedical Translational Imaging Centre (BIOTIC, IWK Health Centre, Halifax, NS, Canada
| | - Christa Davis
- Biomedical Translational Imaging Centre (BIOTIC, IWK Health Centre, Halifax, NS, Canada
| | - W. Martin Kast
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Marianne Stanford
- Dalhousie University, Halifax, NS, Canada
- IMV Inc, Halifax, NS, Canada
| | - Kimberly D. Brewer
- Biomedical Translational Imaging Centre (BIOTIC, IWK Health Centre, Halifax, NS, Canada
- Dalhousie University, Halifax, NS, Canada
- Department of Physics, Dalhousie University, Halifax, NS, Canada
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS, Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
29
|
Cappelletti M, Doll JR, Stankiewicz TE, Lawson MJ, Sauer V, Wen B, Kalinichenko VV, Sun X, Tilburgs T, Divanovic S. Maternal regulation of inflammatory cues is required for induction of preterm birth. JCI Insight 2020; 5:138812. [PMID: 33208552 PMCID: PMC7710297 DOI: 10.1172/jci.insight.138812] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
Infection-driven inflammation in pregnancy is a major cause of spontaneous preterm birth (PTB). Both systemic infection and bacterial ascension through the vagina/cervix to the amniotic cavity are strongly associated with PTB. However, the contribution of maternal or fetal inflammatory responses in the context of systemic or localized models of infection-driven PTB is not well defined. Here, using intraperitoneal or intraamniotic LPS challenge, we examined the necessity and sufficiency of maternal and fetal Toll-like receptor (TLR) 4 signaling in induction of inflammatory vigor and PTB. Both systemic and local LPS challenge promoted induction of inflammatory pathways in uteroplacental tissues and induced PTB. Restriction of TLR4 expression to the maternal compartment was sufficient for induction of LPS-driven PTB in either systemic or intraamniotic challenge models. In contrast, restriction of TLR4 expression to the fetal compartment failed to induce LPS-driven PTB. Vav1-Cre-mediated genetic deletion of TLR4 suggested a critical role for maternal immune cells in inflammation-driven PTB. Further, passive transfer of WT in vitro-derived macrophages and dendritic cells to TLR4-null gravid females was sufficient to induce an inflammatory response and drive PTB. Cumulatively, these findings highlight the critical role for maternal regulation of inflammatory cues in induction of inflammation-driven parturition.
Collapse
Affiliation(s)
- Monica Cappelletti
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jessica R. Doll
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Traci E. Stankiewicz
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Matthew J. Lawson
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Vivien Sauer
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Bingqiang Wen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Center for Lung Regenerative Medicine
| | - Vladimir V. Kalinichenko
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Center for Lung Regenerative Medicine
| | | | - Tamara Tilburgs
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
30
|
Popović J, Wellstein I, Pernis A, Jessberger R, Ocaña-Morgner C. Control of GM-CSF-Dependent Dendritic Cell Differentiation and Maturation by DEF6 and SWAP-70. THE JOURNAL OF IMMUNOLOGY 2020; 205:1306-1317. [PMID: 32709659 DOI: 10.4049/jimmunol.2000020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 06/30/2020] [Indexed: 11/19/2022]
Abstract
Although GM-CSF has been widely used in dendritic cell (DC) research, the mechanisms, factors, and signals regulating steady-state differentiation and maturation of GM-CSF-dependent DCs are insufficiently known. We found that the absence, individually or combined, of the related proteins DEF6 and SWAP-70 strongly enhances differentiation of murine GM-CSF-derived DCs. Contrasting SWAP-70, control through DEF6 does not depend on RHOA activation. DEF6 deficiency leads to expression of the DC-specific transcription factor ZBTB46 and prolonged STAT5 activation in GM-CSF cultures. SWAP-70 and DEF6-mediated restriction of DC differentiation converges mechanistically at the NF-κB pathway. DEF6 acts at early stages of DC differentiation in CD115-cKIT+ myeloid DC progenitors, whereas SWAP-70 acts subsequently. SWAP-70 and DEF6 regulate steady-state DC cytokine expression as well as in vivo accumulation in lymphatic tissue of migratory DCs. Our studies thus elucidate previously unknown roles of two closely related factors with distinct and complementary activities in DC differentiation and steady-state DC function.
Collapse
Affiliation(s)
- Jelena Popović
- Institut für Physiologische Chemie, Medizinische Fakultät Carl Gustav Carus, Technische Unversität Dresden, D-01307 Dresden, Germany; and
| | - Inga Wellstein
- Institut für Physiologische Chemie, Medizinische Fakultät Carl Gustav Carus, Technische Unversität Dresden, D-01307 Dresden, Germany; and
| | - Alessandra Pernis
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021
| | - Rolf Jessberger
- Institut für Physiologische Chemie, Medizinische Fakultät Carl Gustav Carus, Technische Unversität Dresden, D-01307 Dresden, Germany; and
| | - Carlos Ocaña-Morgner
- Institut für Physiologische Chemie, Medizinische Fakultät Carl Gustav Carus, Technische Unversität Dresden, D-01307 Dresden, Germany; and
| |
Collapse
|
31
|
Bailey JD, Shaw A, McNeill E, Nicol T, Diotallevi M, Chuaiphichai S, Patel J, Hale A, Channon KM, Crabtree MJ. Isolation and culture of murine bone marrow-derived macrophages for nitric oxide and redox biology. Nitric Oxide 2020; 100-101:17-29. [PMID: 32339668 PMCID: PMC7284309 DOI: 10.1016/j.niox.2020.04.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/10/2020] [Accepted: 04/20/2020] [Indexed: 12/16/2022]
Abstract
Macrophages are mononuclear phagocytes derived from haematopoietic progenitors that are widely distributed throughout the body. These cells participate in both innate and adaptive immune responses and lie central to the processes of inflammation, development, and homeostasis. Macrophage physiology varies depending on the environment in which they reside and they exhibit rapid functional adaption in response to external stimuli. To study macrophages in vitro, cells are typically cultured ex vivo from the peritoneum or alveoli, or differentiated from myeloid bone marrow progenitor cells to form bone marrow-derived macrophages (BMDMs). BMDMs represent an efficient and cost-effective means of studying macrophage biology. However, the inherent sensitivity of macrophages to biochemical stimuli (such as cytokines, metabolic intermediates, and RNS/ROS) makes it imperative to control experimental conditions rigorously. Therefore, the aim of this study was to establish an optimised and standardised method for the isolation and culture of BMDMs. We used classically activated macrophages isolated from WT and nitric oxide (NO)-deficient mice to develop a standardised culture method, whereby the constituents of the culture media are defined. We then methodically compared our standardised protocol to the most commonly used method of BMDM culture to establish an optimal protocol for the study of nitric oxide (NO)-redox biology and immunometabolism in vitro.
Collapse
Affiliation(s)
- Jade D Bailey
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Andrew Shaw
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; School of Pharmacy & Biomedical Sciences, Faculty of Clinical & Biomedical Sciences, University of Central Lancashire, Preston, PR1 2HE, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Eileen McNeill
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Thomas Nicol
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Marina Diotallevi
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Surawee Chuaiphichai
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Jyoti Patel
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Ashley Hale
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Keith M Channon
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Mark J Crabtree
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK.
| |
Collapse
|
32
|
Blumenthal D, Chandra V, Avery L, Burkhardt JK. Mouse T cell priming is enhanced by maturation-dependent stiffening of the dendritic cell cortex. eLife 2020; 9:e55995. [PMID: 32720892 PMCID: PMC7417170 DOI: 10.7554/elife.55995] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 07/27/2020] [Indexed: 12/28/2022] Open
Abstract
T cell activation by dendritic cells (DCs) involves forces exerted by the T cell actin cytoskeleton, which are opposed by the cortical cytoskeleton of the interacting antigen-presenting cell. During an immune response, DCs undergo a maturation process that optimizes their ability to efficiently prime naïve T cells. Using atomic force microscopy, we find that during maturation, DC cortical stiffness increases via a process that involves actin polymerization. Using stimulatory hydrogels and DCs expressing mutant cytoskeletal proteins, we find that increasing stiffness lowers the agonist dose needed for T cell activation. CD4+ T cells exhibit much more profound stiffness dependency than CD8+ T cells. Finally, stiffness responses are most robust when T cells are stimulated with pMHC rather than anti-CD3ε, consistent with a mechanosensing mechanism involving receptor deformation. Taken together, our data reveal that maturation-associated cytoskeletal changes alter the biophysical properties of DCs, providing mechanical cues that costimulate T cell activation.
Collapse
Affiliation(s)
- Daniel Blumenthal
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Vidhi Chandra
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Lyndsay Avery
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute and Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
33
|
Hamilton JA. GM-CSF in inflammation. J Exp Med 2020; 217:jem.20190945. [PMID: 31611249 PMCID: PMC7037240 DOI: 10.1084/jem.20190945] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/09/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023] Open
Abstract
GM-CSF is a potential therapeutic target in inflammation and autoimmunity. This study reviews the literature on the biology of GM-CSF, in particular that describing the research leading to clinical trials targeting GM-CSF and its receptor in numerous inflammatory/autoimmune conditions, such as rheumatoid arthritis. Granulocyte–macrophage colony-stimulating factor (GM-CSF) has many more functions than its original in vitro identification as an inducer of granulocyte and macrophage development from progenitor cells. Key features of GM-CSF biology need to be defined better, such as the responding and producing cell types, its links with other mediators, its prosurvival versus activation/differentiation functions, and when it is relevant in pathology. Significant preclinical data have emerged from GM-CSF deletion/depletion approaches indicating that GM-CSF is a potential target in many inflammatory/autoimmune conditions. Clinical trials targeting GM-CSF or its receptor have shown encouraging efficacy and safety profiles, particularly in rheumatoid arthritis. This review provides an update on the above topics and current issues/questions surrounding GM-CSF biology.
Collapse
Affiliation(s)
- John A Hamilton
- The University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Australian Institute for Musculoskeletal Science, The University of Melbourne and Western Health, St Albans, Victoria, Australia
| |
Collapse
|
34
|
TonEBP in dendritic cells mediates pro-inflammatory maturation and Th1/Th17 responses. Cell Death Dis 2020; 11:421. [PMID: 32499518 PMCID: PMC7272407 DOI: 10.1038/s41419-020-2632-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 12/29/2022]
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells that link the innate and adaptive immune responses; as such they play pivotal roles in initiation and progression of rheumatoid arthritis (RA). Here, we report that the tonicity-responsive enhancer-binding protein (TonEBP or NFAT5), a Rel family protein involved in the pathogenesis of autoimmune disease and inflammation, is required for maturation and function of DCs. Myeloid cell-specific TonEBP deletion reduces disease severity in a murine model of collagen-induced arthritis; it also inhibits maturation of DCs and differentiation of pathogenic Th1 and Th17 cells in vivo. Upon stimulation by TLR4, TonEBP promotes surface expression of major histocompatibility complex class II and co-stimulatory molecules via p38 mitogen-activated protein kinase. This is followed by DC-mediated differentiation of pro-inflammatory Th1 and Th17 cells. Taken together, these findings provide mechanistic basis for the pathogenic role of TonEBP in RA and possibly other autoimmune diseases.
Collapse
|
35
|
TLR2 and Dectin-1 Signaling in Mouse Hematopoietic Stem and Progenitor Cells Impacts the Ability of the Antigen Presenting Cells They Produce to Activate CD4 T Cells. Cells 2020; 9:cells9051317. [PMID: 32466296 PMCID: PMC7290964 DOI: 10.3390/cells9051317] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/19/2020] [Accepted: 05/22/2020] [Indexed: 12/25/2022] Open
Abstract
Microbial recognition by pattern recognition receptors (PRRs) expressed on hematopoietic stem and progenitor cells (HSPCs) not only activates myelopoiesis but also programs the function of the monocytes and macrophages they produce. For instance, changes in HSPC programming modify the ability of macrophages derived from them to produce inflammatory cytokines. While HSPCs exposed to a TLR2 agonist give rise to tolerized macrophages (lower proinflammatory cytokine production), HSPCs treated with Dectin-1 ligands produce trained macrophages (higher proinflammatory cytokine production). However, nothing is known about the impact of HSPC exposure to microbes on the function of antigen presenting cells (APCs). In this study we evaluated whether treatment of murine bone marrow HSPCs with a TLR2 or Dectin-1 ligand impacts the antigen presenting capacity of APCs derived from them in vitro. Following activation with microbial ligands or Candida albicans yeasts, APCs derived from TLR2/Dectin-1-programed HSPCs exhibit altered expression of MHCII (signal 1), co-stimulatory molecules (CD40, CD80 and CD86; signal 2) and cytokines (TNF-α, IL-6, IL-12 p40 and IL-2; signal 3). Moreover, APCs derived from TLR2/Dectin-1-programed HSPCs prime enhanced Th1 and Th17 responses, which are important for antifungal defense, in CD4 T cell cocultures. Overall, these results demonstrate for the first time that microbial detection by bone marrow HSPCs can modulate the adaptive immune response by inducing the production of APCs with an altered phenotype.
Collapse
|
36
|
van den Biggelaar RHGA, Arkesteijn GJA, Rutten VPMG, van Eden W, Jansen CA. In vitro Chicken Bone Marrow-Derived Dendritic Cells Comprise Subsets at Different States of Maturation. Front Immunol 2020; 11:141. [PMID: 32174908 PMCID: PMC7054383 DOI: 10.3389/fimmu.2020.00141] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/20/2020] [Indexed: 11/13/2022] Open
Abstract
Research in chickens has been fundamental for the discovery of basic aspects of the immune system and has led to an interest in the in-depth characterization of avian immune cell types including dendritic cells (DCs). The in vitro generation and expansion of chicken bone marrow-derived DCs (chBMDCs) in the presence of granulocyte-macrophage colony-stimulating factor (GM-CSF) has provided a way to study chicken DCs, which are only present at limited cell numbers in vivo. This method has been employed to study the interactions between chicken DCs and pathogens or vaccines. However, a detailed characterization of the chBMDC culture is still lacking. In the present study, we performed an elaborate phenotypical and functional analysis of the chBMDC culture and addressed its heterogeneity. After 8 days of culture, chBMDCs comprised major histocompatibility complex class II (MHC-II)low and MHC-IIhigh subsets with different morphologies. Compared with MHC-IIlow chBMDCs, the MHC-IIhigh subset showed a more mature phenotype, with higher expressions of CD1.1, CD40, CD80, CCR7, and CD83, and a relatively low opsonophagocytic capacity. Nevertheless, MHC-IIhigh chBMDCs did not show an increased capacity to induce T-cell proliferation. Therefore, MHC-IIhigh chBMDCs were found to be semi-mature. Interestingly, the presence of the semi-mature MHC-IIhigh chBMDC subset reduced when cells were cultured in the presence of IL-4. Finally, prolonged cell culture after fluorescence-activated cell sorting (FACS) converted the semi-mature MHC-IIhigh subset back into the immature phenotype of the MHC-IIlow subset, demonstrating plasticity of their maturation state. This detailed characterization explained the heterogeneity of the chBMDC culture by the simultaneous presence of immature and semi-mature chBMDC subsets, in addition to cells without features of antigen-presenting cells. Our findings are instrumental for the interpretation of experiments using the chBMDC culture in past and future research by providing insights into its phenotypically and functionally distinct cell types.
Collapse
Affiliation(s)
- Robin H G A van den Biggelaar
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Ger J A Arkesteijn
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Victor P M G Rutten
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Willem van Eden
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Christine A Jansen
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
37
|
Zhou Y, Leng X, Luo X, Mo C, Zou Q, Liu Y, Wang Y. Regulatory Dendritic Cells Induced by K313 Display Anti-Inflammatory Properties and Ameliorate Experimental Autoimmune Encephalitis in Mice. Front Pharmacol 2020; 10:1579. [PMID: 32063843 PMCID: PMC6997778 DOI: 10.3389/fphar.2019.01579] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/05/2019] [Indexed: 12/31/2022] Open
Abstract
As a GSK-3β inhibitor reported by our group, K313 is a novel benzoxazole derivative and displays anti-inflammatory properties in RAW264.7 macrophages without cytotoxicity. The activity of GSK-3β affects the differentiation and maturation of bone marrow-derived dendritic cells (DCs). This study aims to investigate whether K313 can be used to induce regulatory/tolerogenic dendritic cells (DCregs), and the therapeutic effects of DCregs induced by K313 in the autoimmune model of experimental autoimmune encephalitis (EAE). The results show that compared with LPS stimulated mature DCs, K313-treated bone marrow-derived DCs display obvious tolerogenic characteristics with decreased expression of co-stimulatory molecules, downregulated secretions of pro-inflammatory cytokines and unregulated secretion of anti-inflammatory cytokine IL-10. The above characteristics conform to the typical phenotypes of DCregs. Moreover, K313-modified DCregs inhibit antigen-specific T cell responses in vitro. Furthermore, by adoptive transfer, K313 modified DCregs to the EAE mice, and the development of disease was ameliorated to some extent. In addition, treatment with K313-modified DCregs also significantly reduced the percentages of splenetic Th1 and Th17 cells and increased the percentage of regulatory T cells in EAE mice. In conclusion, K313-modified DCregs show anti-inflammatory properties in vitro and have a significant positive effect on the EAE disease in vivo. Our data indicate that K313-induced DCregs pulsed with auto-antigen might have potential use as a therapeutic approach for autoimmune inflammation of the central nervous system.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Emergency, West China Second University Hospital and Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Xiao Leng
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Xingyan Luo
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Chunfen Mo
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Qiang Zou
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Yang Liu
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Yantang Wang
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| |
Collapse
|
38
|
Wahi K, Kodar K, McConnell MJ, Harper JL, Timmer MSM, Stocker BL. MSU Crystals Enhance TDB-Mediated Inflammatory Macrophage IL-1β Secretion. Inflammation 2019; 42:1129-1136. [PMID: 30806957 DOI: 10.1007/s10753-019-00976-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The tumour microenvironment predominantly consists of macrophages with phenotypes ranging from pro-inflammatory (M1-like) to anti-inflammatory (M2-like). Trehalose-6,6'-dibehenate (TDB) displays moderate anti-tumour activity and stimulates M1-like macrophages via the macrophage inducible C-type lectin (Mincle) resulting in IL-1β production. In this study, we examined if monosodium urate (MSU), a known vaccine adjuvant, can boost IL-1β production by TDB-stimulated macrophages. We investigated the effect of MSU/TDB co-treatment on IL-1β production by GM-CSF (M1-like) and M-CSF/IL-4 (M2-like) differentiated mouse bone marrow macrophages (BMMs) and found that MSU/TDB co-treatment of GM-CSF BMMs significantly enhanced IL-1β production in a Mincle-dependent manner. Western blot analysis showed that increased IL-1β production by GM-CSF BMMs was associated with the induction of pro-IL-1β expression by TDB rather than MSU. Flow cytometry analysis showed that MSU/TDB co-stimulation of GM-CSF BMMs led to greater expansion of CD86high/MHC IIhigh and CD86low/MHC IIlow subpopulations; however, only the latter showed increased production of IL-1β. Together, these findings provide evidence of the potential to use MSU/TDB co-treatment to boost IL-1β-mediated anti-tumour activity in M1-like tumour-associated macrophages.
Collapse
Affiliation(s)
- Kanu Wahi
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.,Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Kristel Kodar
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.,Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Melanie J McConnell
- Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.,School of Biological Sciences, PO Box 600, Wellington, New Zealand
| | - Jacquie L Harper
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Mattie S M Timmer
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand. .,Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.
| | - Bridget L Stocker
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand. .,Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.
| |
Collapse
|
39
|
Zhan Y, Lew AM, Chopin M. The Pleiotropic Effects of the GM-CSF Rheostat on Myeloid Cell Differentiation and Function: More Than a Numbers Game. Front Immunol 2019; 10:2679. [PMID: 31803190 PMCID: PMC6873328 DOI: 10.3389/fimmu.2019.02679] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 10/30/2019] [Indexed: 12/27/2022] Open
Abstract
Granulocyte Macrophage-Colony Stimulating Factor (GM-CSF) is a myelopoietic growth factor that has pleiotropic effects not only in promoting the differentiation of immature precursors into polymorphonuclear neutrophils (PMNs), monocytes/macrophages (MØs) and dendritic cells (DCs), but also in controlling the function of fully mature myeloid cells. This broad spectrum of GM-CSF action may elicit paradoxical outcomes-both immunostimulation and immunosuppression-in infection, inflammation, and cancer. The complexity of GM-CSF action remains to be fully unraveled. Several aspects of GM-CSF action could contribute to its diverse biological consequences. Firstly, GM-CSF as a single cytokine affects development of most myeloid cells from progenitors to mature immune cells. Secondly, GM-CSF activates JAK2/STAT5 and also activate multiple signaling modules and transcriptional factors that direct different biological processes. Thirdly, GM-CSF can be produced by different cell types including tumor cells in response to different environmental cues; thus, GM-CSF quantity can vary greatly under different pathophysiological settings. Finally, GM-CSF signaling is also fine-tuned by other less defined feedback mechanisms. In this review, we will discuss the role of GM-CSF in orchestrating the differentiation, survival, and proliferation during the generation of multiple lineages of myeloid cells (PMNs, MØs, and DCs). We will also discuss the role of GM-CSF in regulating the function of DCs and the functional polarization of MØs. We highlight how the dose of GM-CSF and corresponding signal strength acts as a rheostat to fine-tune cell fate, and thus the way GM-CSF may best be targeted for immuno-intervention in infection, inflammation and cancer.
Collapse
Affiliation(s)
- Yifan Zhan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Andrew M Lew
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,Department of Immunology and Microbiology, University of Melbourne, Parkville, VIC, Australia
| | - Michael Chopin
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
40
|
Kim SE, Hwang JH, Kim YK, Lee HT. Heterogeneity of porcine bone marrow-derived dendritic cells induced by GM-CSF. PLoS One 2019; 14:e0223590. [PMID: 31689334 PMCID: PMC6830806 DOI: 10.1371/journal.pone.0223590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 09/24/2019] [Indexed: 11/24/2022] Open
Abstract
In vitro generation of dendritic cells (DCs) is advantageous for overcoming the low frequency of primary DCs and the difficulty of applying isolation techniques for studying DC immunobiology. The culture of bone marrow cells with granulocyte-macrophage colony-stimulating factor (GM-CSF) has been used extensively to generate bone marrow-derived dendritic cells (BMDCs). Studies have reported the heterogeneity of cells grown in murine GM-CSF culture based on the levels of MHCII expression. Although porcine DCs are generated by this classical method, the exact characteristics of the BMDC population have not yet been defined. In this study, we discriminated GM-CSF-grown BMDCs from gnotobiotic miniature pigs according to several criteria including morphology, phenotype, gene expression pattern and function. We showed that porcine BMDCs were heterogeneous cells that differentially expressed MHCII. MHCIIhigh cells displayed more representative of DC-like morphology and phenotype, including costimulatory molecules, as well as they showed a superior T cell priming capacity as compared to MHCIIlow cell. Our data showed that the difference in MHCIIhigh and MHCIIlow cell populations involved distinct maturation states rather than the presence of different cell types. Overall, characterization of porcine BMDC cultures provides important information about this widely used cellular model.
Collapse
Affiliation(s)
- Sang Eun Kim
- Department of Animal Biotechnology, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
| | - Jeong Ho Hwang
- Department of Animal Biotechnology, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
- Animal Model Research Group, Jeonbuk Department of Inhalation Research, Korea Institute of Toxicology, Jeongeup, Jeollabuk-do, Republic of Korea
| | - Young Kyu Kim
- Department of Animal Biotechnology, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
| | - Hoon Taek Lee
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
41
|
Majdoubi A, Lee JS, Balood M, Sabourin A, DeMontigny A, Kishta OA, Moulefera MA, Galbas T, Yun TJ, Talbot S, Ishido S, Cheong C, Thibodeau J. Downregulation of MHC Class II by Ubiquitination Is Required for the Migration of CD206 + Dendritic Cells to Skin-Draining Lymph Nodes. THE JOURNAL OF IMMUNOLOGY 2019; 203:2887-2898. [PMID: 31659013 DOI: 10.4049/jimmunol.1900593] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/20/2019] [Indexed: 12/20/2022]
Abstract
Dendritic cells (DCs) are critical players in skin homeostasis. A subset of mannose receptor (CD206)-expressing monocyte-derived DCs was found in skin, and their migratory counterpart is present in skin-draining lymph nodes (sdLNs). Skin CD206+ DCs were shown to upregulate MHC class II (MHCII) progressively, raising the question of whether this feature affects their biology. In this study, we assessed the role of MHCII regulation in the development and migration of these cells in mouse models expressing differential MHCII levels. Using CD206 as a surrogate marker, we found that skin CD206+ DCs develop in an MHCII-independent manner. However, their migration to sdLNs was affected by overexpression rather than absence or lower expression of MHCII. Accordingly, B16 tumor growth was exacerbated in mice overexpressing MHCII in the absence of ubiquitination. Mechanistically, CD206+ DCs from these mice showed decreased IRF4 and CCR7 expression. LPS, which is known to promote monocyte-derived DC recruitment to sdLNs, partially improved these defects. However, GM-CSF delivery restored CD206+ DC migration by promoting IRF4 expression. Collectively, these data show that MHCII downregulation is crucial for IRF4-dependent migration of CD206+ DCs to sdLNs in health and disease.
Collapse
Affiliation(s)
- Abdelilah Majdoubi
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal H3T 1J4, Quebec, Canada
| | - Jun Seong Lee
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal H3T 1J4, Quebec, Canada
| | - Mohammad Balood
- Département de Pharmacologie, Université de Montréal, Montreal H3T 1J4, Quebec, Canada
| | - Antoine Sabourin
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal H3T 1J4, Quebec, Canada
| | - Auriane DeMontigny
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal H3T 1J4, Quebec, Canada
| | - Osama A Kishta
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal H3T 1J4, Quebec, Canada
| | - Mohamed Abdelwafi Moulefera
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal H3T 1J4, Quebec, Canada
| | - Tristan Galbas
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal H3T 1J4, Quebec, Canada
| | - Tae Jin Yun
- Institut de Recherches Cliniques de Montréal, Montreal H2W 1R7, Quebec, Canada; and
| | - Sébastien Talbot
- Département de Pharmacologie, Université de Montréal, Montreal H3T 1J4, Quebec, Canada
| | - Satoshi Ishido
- Department of Microbiology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Cheolho Cheong
- Institut de Recherches Cliniques de Montréal, Montreal H2W 1R7, Quebec, Canada; and
| | - Jacques Thibodeau
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal H3T 1J4, Quebec, Canada;
| |
Collapse
|
42
|
Abstract
Pre-clinical models and clinical trials demonstrate that targeting the action of the cytokine, granulocyte macrophage-colony stimulating factor (GM-CSF), can be efficacious in inflammation/autoimmunity reinforcing the importance of understanding how GM-CSF functions; a significant GM-CSF-responding cell in this context is likely to be the monocyte. This article summarizes critically the literature on the downstream cellular pathways regulating GM-CSF interaction with monocytes (and macrophages), highlighting some contentious issues, and conclusions surrounding this biology. It also suggests future directions which could be undertaken so as to more fully understand this aspect of GM-CSF biology. Given the focus of this collection of articles on monocytes, the following discussion in general will be limited to this population or to its more mature progeny, the macrophage, even though GM-CSF biology is broader than this.
Collapse
Affiliation(s)
- John A Hamilton
- The University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, VIC, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St. Albans, VIC, Australia
| |
Collapse
|
43
|
Becerra-Díaz M, Strickland AB, Keselman A, Heller NM. Androgen and Androgen Receptor as Enhancers of M2 Macrophage Polarization in Allergic Lung Inflammation. THE JOURNAL OF IMMUNOLOGY 2018; 201:2923-2933. [PMID: 30305328 DOI: 10.4049/jimmunol.1800352] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 09/10/2018] [Indexed: 02/06/2023]
Abstract
Allergic asthma is a disease initiated by a breach of the lung mucosal barrier and an inappropriate Th2 inflammatory immune response that results in M2 polarization of alveolar macrophages (AM). The number of M2 macrophages in the airway correlates with asthma severity in humans. Sex differences in asthma suggest that sex hormones modify lung inflammation and macrophage polarization. Asthmatic women have more M2 macrophages than asthmatic men and androgens have been used as an experimental asthma treatment. In this study, we demonstrate that although androgen (dihydrotestosterone) reconstitution of castrated mice reduced lung inflammation in a mouse model of allergic lung inflammation, it enhanced M2 polarization of AM. This indicates a cell-specific role for androgens. Dihydrotestosterone also enhanced IL-4-stimulated M2 macrophage polarization in vitro. Using mice lacking androgen receptor (AR) in monocytes/macrophages (ARfloxLysMCre), we found that male but not female mice exhibited less eosinophil recruitment and lung inflammation due to impaired M2 polarization. There was a reduction in eosinophil-recruiting chemokines and IL-5 in AR-deficient AM. These data reveal an unexpected and novel role for androgen/AR in promoting M2 macrophage polarization. Our findings are also important for understanding pathology in diseases promoted by M2 macrophages and androgens, such as asthma, eosinophilic esophagitis, and prostate cancer, and for designing new approaches to treatment.
Collapse
Affiliation(s)
- Mireya Becerra-Díaz
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21205; and
| | - Ashley B Strickland
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21205; and
| | - Aleksander Keselman
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21205; and
| | - Nicola M Heller
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21205; and .,Allergy and Clinical Immunology, Johns Hopkins University, Baltimore, MD 21205
| |
Collapse
|
44
|
So EY, Sun C, Reginato AM, Dubielecka PM, Ouchi T, Liang OD. Loss of lipid phosphatase SHIP1 promotes macrophage differentiation through suppression of dendritic cell differentiation. Cancer Biol Ther 2018; 20:201-211. [PMID: 30277839 DOI: 10.1080/15384047.2018.1523846] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
SH2-containing inositol 5'-phosphatase-1 (SHIP1) deficiency in mice results in abnormal myeloid expansion, and proinflammatory conditions in the lung. However, the mechanisms involved in SHIP1-mediated regulation of myeloid differentiation remain unclear. Here we show that SHIP1 is a key regulator of early differentiation for dendritic cells (DCs). We also provide critical evidence to modify the function of SHIP1 in in vitro development of BMDCs using the recent framework of defining DCs. We found that loss of SHIP1 suppresses GM-CSF-induced formation of bone marrow-derived DC (BMDC) colonies, leading to reduced BMDC number in BM cell culture. The number of maturated BMDCs decreased in SHIP1-KO culture, due to reduction of immature BMDCs, suggesting SHIP1 is critical for lineage commitment rather than for maturation from myeloid precursors to DCs. We further showed that F4/80+/MHCIIlow BM macrophage-like cells (BMMs) were the main population of SHIP1-KO BM culture. Treatment of wild-type BM culture with 3 α-aminocholestane (3AC), a specific inhibitor for functional activity of SHIP1, caused a similar developmental defect in BMDCs as seen in SHIP1-KO cells, resulting in the absence of BMDC colony, and increased number of BMMs in BM culture. In conclusion, our results suggest that differentiation of BMDCs are markedly impaired under SHIP1 deficient condition, which causes skewed development of myeloid lineage cells manifested as pathological conditions associated with an excess of macrophage population.
Collapse
Affiliation(s)
- Eui Young So
- a Division of Hematology and Oncology, Department of Medicine, Rhode Island Hospital , Warren Alpert Medical School of Brown University , Providence , USA
| | - Changqi Sun
- b Division of Rheumatology, Department of Medicine, Rhode Island Hospital , Warren Alpert Medical School of Brown University , Providence , USA
| | - Anthony M Reginato
- b Division of Rheumatology, Department of Medicine, Rhode Island Hospital , Warren Alpert Medical School of Brown University , Providence , USA
| | - Patrycia M Dubielecka
- a Division of Hematology and Oncology, Department of Medicine, Rhode Island Hospital , Warren Alpert Medical School of Brown University , Providence , USA
| | - Toru Ouchi
- c Department of Cancer Genetics , Roswell Park Cancer Institute , Buffalo , USA
| | - Olin D Liang
- a Division of Hematology and Oncology, Department of Medicine, Rhode Island Hospital , Warren Alpert Medical School of Brown University , Providence , USA
| |
Collapse
|
45
|
Sun L, Rautela J, Delconte RB, Souza-Fonseca-Guimaraes F, Carrington EM, Schenk RL, Herold MJ, Huntington ND, Lew AM, Xu Y, Zhan Y. GM-CSF Quantity Has a Selective Effect on Granulocytic vs. Monocytic Myeloid Development and Function. Front Immunol 2018; 9:1922. [PMID: 30210491 PMCID: PMC6120981 DOI: 10.3389/fimmu.2018.01922] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/06/2018] [Indexed: 01/14/2023] Open
Abstract
GM-CSF promotes myeloid differentiation of cultured bone marrow cells into cells of the granulocytic and monocytic lineage; the latter can further differentiate into monocytes/macrophages and dendritic cells. How GM-CSF selects for these different myeloid fates is unresolved. GM-CSF levels can change either iatrogenically (e.g., augmenting leukopoiesis after radiotherapy) or naturally (e.g., during infection or inflammation) resulting in different immunological outcomes. Therefore, we asked whether the dose of GM-CSF may regulate the development of three types of myeloid cells. Here, we showed that GM-CSF acted as a molecular rheostat where the quantity determined which cell type was favored; moreover, the cellular process by which this was achieved was different for each cell type. Thus, low quantities of GM-CSF promoted the granulocytic lineage, mainly through survival. High quantities promoted the monocytic lineage, mainly through proliferation, whereas moderate quantities promoted moDCs, mainly through differentiation. Finally, we demonstrated that monocytes/macrophages generated with different doses of GM-CSF differed in function. We contend that this selective effect of GM-CSF dose on myeloid differentiation and function should be taken into consideration during pathophysiological states that may alter GM-CSF levels and during GM-CSF agonistic or antagonistic therapy.
Collapse
Affiliation(s)
- Li Sun
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, China
| | - Jai Rautela
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Rebecca B Delconte
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Fernando Souza-Fonseca-Guimaraes
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Emma M Carrington
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Robyn L Schenk
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Marco J Herold
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Nicholas D Huntington
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Andrew M Lew
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,Department of Immunology and Microbiology, University of Melbourne, Parkville, VIC, Australia
| | - Yuekang Xu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, China
| | - Yifan Zhan
- The Walter & Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,Guangzhou Women and Children's Medical Centre, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
46
|
Rogers PB, Schwartz EH. Generation of Large Numbers of Myeloid Progenitors and Dendritic Cell Precursors from Murine Bone Marrow Using a Novel Cell Sorting Strategy. J Vis Exp 2018:57365. [PMID: 30148496 PMCID: PMC6126687 DOI: 10.3791/57365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cultures of monocyte-derived dendritic cells (moDC) generated from mouse bone marrow using Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF) have recently been recognized to be more heterogeneous than previously appreciated. These cultures routinely contain moDC as well monocyte-derived macrophages (moMac), and even some less developed cells such as monocytes. The goal of this protocol is to provide a consistent method for identification and separation of the many cell types present in these cultures as they develop, so that their specific functions may be further investigated. The sorting strategy presented here separates cells first into four populations based on expression of Ly6C and CD115, both of which are expressed transiently by cells as they develop in GM-CSF-driven culture. These four populations include Common myeloid progenitors or CMP (Ly6C-, CD115-), granulocyte/macrophage progenitors or GMP (Ly6C+, CD115-), monocytes (Ly6C+, CD115+), and monocyte-derived macrophages or moMac (Ly6C-, CD115+). CD11c is also added to the sorting strategy to distinguish two populations within the Ly6C-, CD115- population: CMP (CD11c-) and moDC (CD11c+). Finally, two populations may be further distinguished within the Ly6C-, CD115+ population based on the level of MHC class II expression. MoMacs express lower levels of MHC class II, while a monocyte-derived DC precursor (moDP) expresses higher MHC class II. This method allows for the reliable isolation of several developmentally distinct populations in numbers sufficient for a variety of functional and developmental analyses. We highlight one such functional readout, the differential responses of these cell types to stimulation with Pathogen-Associated Molecular Patterns (PAMPs).
Collapse
|
47
|
Li Y, Li Y, Lu W, Li H, Wang Y, Luo H, Wu Y, Dong W, Bai G, Zhang Y. Integrated Network Pharmacology and Metabolomics Analysis of the Therapeutic Effects of Zi Dian Fang on Immune Thrombocytopenic Purpura. Front Pharmacol 2018; 9:597. [PMID: 29971001 PMCID: PMC6018083 DOI: 10.3389/fphar.2018.00597] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 05/18/2018] [Indexed: 12/25/2022] Open
Abstract
Current hormone-based treatments for immune thrombocytopenic purpura (ITP) are associated with potentially serious adverse reactions. Zi Dian Fang (ZDF) is a multi-target Traditional Chinese Medicine (TCM) used to treat both the symptoms and root causes of ITP, with fewer side effects than hormone-based treatments. This study analysis of the therapeutic effects of ZDF on ITP from three aspects: platelet proliferation, immunoregulation, and inflammation. After detection of 52 chemical constituents of ZDF by UPLC-Q-TOF/MS, The main targets and pathways affected by ZDF were screened by network pharmacology and verified by Western blot and ELISA. Meanwhile, metabolomics analysis were applied to a mouse model of ITP to identify and screen endogenous terminal metabolites differentially regulated by ZDF. Integrated network pharmacology and metabolomics analysis of the therapeutic effects of ZDF on ITP may be as follows: ZDF counteracts ITP symptoms mainly by inhibiting Ras/MAPKs (Ras/Mitogen-activated protein kinases) pathway, and the expression of upstream protein (Ras) and downstream protein (p-ERK, p-JNK, p-p38) were inhibited, which affects the content of effect index associated with proliferation (Thrombopoietin, TPO; Granulocyte-macrophage colony stimulating factor, GM-CSF), inflammation (Tumor necrosis factor-α, TNF-α; Interleukin-6, IL-6), immune (Interleukin-2, IL-2; Interferon-gamma, IFN-γ; Interleukin-4, IL-4), so that the body’s arginine, Δ12-prostaglandin j2 (Δ12-PGJ2), 9-cis-Retinoic Acid, sphingosine-1-phosphate (S1P), oleic acid amide and other 12 endogenous metabolites significantly changes. Considering the established safety profile, the present study suggests ZDF may be a useful alternative to hormone-based therapies for the treatment of ITP.
Collapse
Affiliation(s)
- Yubo Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yamei Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenliang Lu
- Tasly Institute, Tasly Pharmaceutical Group, Tianjin, China
| | - Hongbin Li
- Tasly Institute, Tasly Pharmaceutical Group, Tianjin, China
| | - Yuming Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Houmin Luo
- Tianjin State Key Laboratory of Modern Chinese Medicine, School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuanyuan Wu
- Tianjin State Key Laboratory of Modern Chinese Medicine, School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenying Dong
- Tianjin State Key Laboratory of Modern Chinese Medicine, School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Gang Bai
- College of Pharmacy, Nankai University, Tianjin, China
| | - Yanjun Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
48
|
Schutt CR, Gendelman HE, Mosley RL. Tolerogenic bone marrow-derived dendritic cells induce neuroprotective regulatory T cells in a model of Parkinson's disease. Mol Neurodegener 2018; 13:26. [PMID: 29783988 PMCID: PMC5963189 DOI: 10.1186/s13024-018-0255-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 05/02/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Administration of granulocyte-macrophage colony-stimulating factor (GM-CSF) increases regulatory T cell (Treg) number and function with control of neuroinflammation and neuronal protection in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of Parkinson's disease (PD). Recently, we demonstrated in an early phase 1 clinical trial that GM-CSF also improves motor skills in PD patients. However, the mechanisms of Treg induction and its effects on neuroprotective responses remain unknown. As GM-CSF induces tolerogenic dendritic cells (DCs) that in turn convert conventional T cells to Tregs, the pathways for DC induction of Tregs were assessed. METHODS Following differentiation, bone marrow-derived dendritic cells (BMDCs) were cultured in media or GM-CSF with or without post-culture stimulation with nitrated α-synuclein (N-α-Syn). Expression of cell surface co-stimulatory molecules and proinflammatory cytokines, and induction of Tregs were evaluated. The neuroprotective capacity of tolerogenic BMDCs was assessed by adoptive transfer to MPTP-intoxicated mice. The extent of neuroinflammation and numbers of surviving dopaminergic neurons were assessed in relation to Treg numbers. RESULTS Co-culture of differentiated BMDCs with conventional T cells led to Treg induction. Stimulation of BMDCs with N-α-Syn increased expression of co-stimulatory molecules and proinflammatory cytokines, with modest increases in Treg numbers. In contrast, continued culture of BMDCs with GM-CSF modestly altered expression of co-stimulatory molecules and proinflammatory cytokines and chemokines, but decreased Treg induction. Continued culture in GM-CSF and combined stimulation with N-α-Syn reduced Treg induction to the lowest levels. Adoptive transfer of tolerogenic BMDCs to MPTP-intoxicated mice increased splenic Tregs, attenuated neuroinflammatory responses, and protected nigrostriatal dopaminergic neurons. CONCLUSIONS GM-CSF acts broadly to differentiate DCs and affect immune transformation from effector to regulatory immune responses. DCs skew such immune responses by increasing Treg numbers and activities that serve to attenuate proinflammatory responses and augment neuroprotection.
Collapse
Affiliation(s)
- Charles R Schutt
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985930 Nebraska Medical Center, Omaha, NE 68198-5930, USA.
| |
Collapse
|
49
|
Olatunde AC, Abell LP, Landuyt AE, Hiltbold Schwartz E. Development of endocytosis, degradative activity, and antigen processing capacity during GM-CSF driven differentiation of murine bone marrow. PLoS One 2018; 13:e0196591. [PMID: 29746488 PMCID: PMC5944997 DOI: 10.1371/journal.pone.0196591] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 04/16/2018] [Indexed: 12/22/2022] Open
Abstract
Dendritic cells (DC) are sentinels of the immune system, alerting and enlisting T cells to clear pathogenic threats. As such, numerous studies have demonstrated their effective uptake and proteolytic activities coupled with antigen processing and presentation functions. Yet, less is known about how these cellular mechanisms change and develop as myeloid cells progress from progenitor cells to more differentiated cell types such as DC. Thus, our study comparatively examined these functions at different stages of myeloid cell development driven by the GM-CSF. To measure these activities at different stages of development, GM-CSF driven bone marrow cells were sorted based on expression of Ly6C, CD115, and CD11c. This strategy enables isolation of cells representing five distinct myeloid cell types: Common Myeloid Progenitor (CMP), Granulocyte/Macrophage Progenitor (GMP), monocytes, monocyte-derived Macrophage/monocyte-derived Dendritic cell Precursors (moMac/moDP), and monocyte-derived DC (moDC). We observed significant differences in the uptake capacity, proteolysis, and antigen processing and presentation functions between these myeloid cell populations. CMP showed minimal uptake capacity with no detectable antigen processing and presenting function. The GMP population showed higher uptake capacity, modest proteolytic activity, and little T cell stimulatory function. In the monocyte population, the uptake capacity reached its peak, yet this cell type had minimal antigen processing and presentation function. Finally, moMac/moDP and moDC had a modestly decreased uptake capacity, high degradative capacity and strong antigen processing and presentation functions. These insights into when antigen processing and presentation function develop in myeloid cells during GM-CSF driven differentiation are crucial to the development of vaccines, allowing targeting of the most qualified cells as an ideal vaccine vehicles.
Collapse
Affiliation(s)
| | - Laura P. Abell
- Department of Biological Sciences, Auburn University, Auburn, Alabama
| | - Ashley E. Landuyt
- Department of Biological Sciences, Auburn University, Auburn, Alabama
| | | |
Collapse
|
50
|
Stable incorporation of GM-CSF into dissolvable microneedle patch improves skin vaccination against influenza. J Control Release 2018; 276:1-16. [PMID: 29496540 PMCID: PMC5967648 DOI: 10.1016/j.jconrel.2018.02.033] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 02/17/2018] [Accepted: 02/19/2018] [Indexed: 11/20/2022]
Abstract
The widely used influenza subunit vaccine would benefit from increased protection rates in vulnerable populations. Skin immunization by microneedle (MN) patch can increase vaccine immunogenicity, as well as increase vaccination coverage due to simplified administration. To further increase immunogenicity, we used granulocyte-macrophage colony stimulating factor (GM-CSF), an immunomodulatory cytokine already approved for skin cancer therapy and cancer support treatment. GM-CSF has been shown to be upregulated in skin following MN insertion. The GM-CSF-adjuvanted vaccine induced robust and long-lived antibody responses cross-reactive to homosubtypic and heterosubtypic influenza viruses. Addition of GM-CSF resulted in increased memory B cell persistence relative to groups given influenza vaccine alone and led to rapid lung viral clearance following lethal infection with homologous virus in the mouse model. Here we demonstrate that successful incorporation of the thermolabile cytokine GM-CSF into MN resulted in improved vaccine-induced protective immunity holding promise as a novel approach to improved influenza vaccination. To our knowledge, this is the first successful incorporation of a cytokine adjuvant into dissolvable MNs, thus advancing and diversifying the rapidly developing field of MN vaccination technology.
Collapse
|