1
|
Du F, Ye Z, He A, Yuan J, Su M, Jia Q, Wang H, Yang P, Yang Z, Ning P, Wang Z. An engineered α1β1 integrin-mediated FcγRI signaling component to control enhanced CAR macrophage activation and phagocytosis. J Control Release 2025; 377:689-703. [PMID: 39617174 DOI: 10.1016/j.jconrel.2024.11.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024]
Abstract
Treatment of solid tumors remains difficult, and therefore there has been increased focus on chimeric antigen receptor macrophages (CAR-M) to challenge solid tumors. However, CAR domain design of of adoptive cell therapy, which leads to differences in antitumor activity and triggered antitumor potential, remains poorly understood for macrophages. We developed an α1β1 integrin-mediated Fc-gamma receptor I (FcγRI) signaling component for CAR-M specific activation and its antitumor potential. We evaluated CAR-M effects with α1β1 integrin-mediated FcγRI signaling (ACT CAR-M) on the activation and antitumor phagocytic response of macrophages in vitro. Subcutaneous tumor model in BALB/c mice and carcinomatosis model in immunodeficient mice were used to test the antitumor effect of ACT CAR-M compared with CD3ζ CAR-M. The α1β1 integrin-mediated FcγRI signaling engagement of CAR-M was associated with enhanced macrophage activation and specific phagocytosis in primary human macrophages, and significantly improved tumor control and survival in multiple cancer models when compared to CD3ζ CAR-M. RNA-sequencing suggested that α1β1 integrin-mediated FcγRI engagement increased antitumor immunity by enhancing pro-inflammatory M1 phenotype-associated pathways, such as Toll-like receptor signaling, tumor necrosis factor signaling, and IL-17 signaling. α1β1 integrin-mediated FcγRI signaling engagement markedly enhanced antitumor effects of CAR-M immunotherapy, which is proposed as an advanced engineering CAR domain material to expand the clinical application of CAR-M.
Collapse
Affiliation(s)
- Fuyu Du
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China
| | - Zixuan Ye
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China
| | - Anna He
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China
| | - Jingtong Yuan
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China
| | - Maozhi Su
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China
| | - Qingan Jia
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710038, China
| | - Huaiyu Wang
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Peng Yang
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China
| | - Zuo Yang
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China
| | - Pengbo Ning
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China.
| | - Zhongliang Wang
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710071, PR China; Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, Xi'an, Shaanxi 710071, PR China.
| |
Collapse
|
2
|
Gudgeon J, Dannoura A, Chatterjee R, Sidgwick F, Raymond BB, Frey AM, Marin-Rubio JL, Trost M. Mass spectrometry-based proteomic exploration of diverse murine macrophage cellular models. Life Sci Alliance 2025; 8:e202402760. [PMID: 39510801 PMCID: PMC11544424 DOI: 10.26508/lsa.202402760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 11/15/2024] Open
Abstract
Immortalised cell lines that mimic their primary cell counterparts are fundamental to research, particularly when large cell numbers are required. Here, we report that immortalisation of bone marrow-derived macrophages (iBMDMs) using the J2 virus resulted in the loss of a protein of interest, MSR1, in WT cells by an unknown mechanism. This led us to perform an in-depth mass spectrometry-based proteomic characterisation of common murine macrophage cell lines (J774A.1, RAW264.7, and BMA3.1A7), in comparison with the iBMDMs, as well as primary BMDMs from both C57BL/6 and BALB/c mice. This analysis revealed striking differences in protein profiles associated with macrophage polarisation, phagocytosis, pathogen recognition, and interferon signalling. Among the cell lines, J774A.1 cells were the most similar to the gold standard primary BMDM model, whereas BMA3.1A7 cells were the least similar because of the reduction in abundance of several key proteins related closely to macrophage function. This comprehensive proteomic dataset offers valuable insights into the use and suitability of macrophage cell lines for cell signalling and inflammation research.
Collapse
Affiliation(s)
- Jack Gudgeon
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Abeer Dannoura
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Ritika Chatterjee
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Frances Sidgwick
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Andrew M Frey
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Matthias Trost
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
3
|
Mingot-Castellano ME. Sovleplenib in immune thrombocytopenia. Lancet Haematol 2024; 11:e552-e553. [PMID: 38885671 DOI: 10.1016/s2352-3026(24)00180-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024]
Affiliation(s)
- María Eva Mingot-Castellano
- Hematology Department, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina, Universidad de Sevilla, Seville 41013, Spain.
| |
Collapse
|
4
|
Chupradit K, Muneekaew S, Wattanapanitch M. Engineered CD147-CAR macrophages for enhanced phagocytosis of cancers. Cancer Immunol Immunother 2024; 73:170. [PMID: 38954079 PMCID: PMC11219683 DOI: 10.1007/s00262-024-03759-6] [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: 04/04/2024] [Accepted: 06/11/2024] [Indexed: 07/04/2024]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has shown promising results in hematologic malignancies, but its effectiveness in solid cancers remains challenging. Macrophages are immune cells residing within the tumor microenvironment. They can phagocytose tumor cells. Recently, CAR macrophages (CAR-M) have been a promising candidate for treating solid cancers. One of the common cancer antigens overexpressed in various types of cancer is CD147. CAR-T and NK cells targeting CD147 antigen have shown significant efficacy against hepatocellular carcinoma. Nevertheless, CAR-M targeting the CD147 molecule has not been investigated. In this study, we generated CAR targeting the CD147 molecule using the THP-1 monocytic cell line (CD147 CAR-M). The CD147 CAR-M exhibited typical macrophage characteristics, including phagocytosis of zymosan bioparticles and polarization ability toward M1 and M2 phenotypes. Furthermore, the CD147 CAR-M demonstrated enhanced anti-tumor activity against K562 and MDA-MB-231 cells without exhibiting off-target cytotoxicity against normal cells. Our research provides valuable insights into the potential of CD147 CAR-M as a promising platform for cancer immunotherapy, with applications in both hematologic malignancies and solid cancers.
Collapse
Affiliation(s)
- Koollawat Chupradit
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Saitong Muneekaew
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Methichit Wattanapanitch
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
5
|
Yu L, Zhang D, Yin Y, Li X, Bai C, Zhou Q, Liu X, Tian X, Xu D, Yu X, Zhao S, Hu R, Guo F, Yang Y, Ren Y, Chen G, Zeng J, Feng J. Tibial cortex transverse transport surgery improves wound healing in patients with severe type 2 DFUs by activating a systemic immune response: a cross-sectional study. Int J Surg 2024; 111:01279778-990000000-01748. [PMID: 38954658 PMCID: PMC11745691 DOI: 10.1097/js9.0000000000001897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Tibial cortex transverse transport (TTT) surgery has become an ideal treatment for patients with type 2 severe diabetic foot ulcerations (DFUs) while conventional treatments are ineffective. Based on our clinical practice experience, the protective immune response from TTT surgery may play a role against infections to promote wound healing in patients with DFUs. Therefore, this research aimed to systematically study the specific clinical efficacy and the mechanism of TTT surgery. MATERIALS AND METHODS Between June 2022 and September 2023, 68 patients with type 2 severe DFUs were enrolled and therapized by TTT surgery in this cross-sectional and experimental study. Major clinical outcomes including limb salvage rate and antibiotics usage rate were investigated. Ten clinical characteristics and laboratory features of glucose metabolism and kidney function were statistically analyzed. Blood samples from 6 key time points of TTT surgery were collected for label-free proteomics and clinical immune biomarker analysis. Besides, tissue samples from 3 key time points were for spatially resolved metabolomics and transcriptomics analysis, as well as applied to validate the key TTT-regulated molecules by RT-qPCR. RESULTS Notably, 64.7% of patients did not use antibiotics during the entire TTT surgery. TTT surgery can achieve a high limb salvage rate of 92.6% in patients with unilateral or bilateral DFUs. Pathway analysis of a total of 252 differentially expressed proteins (DEPs) from the proteomic revealed that the immune response induced by TTT surgery at different stages was first comprehensively verified through multi-omics combined with immune biomarker analysis. The function of upward transport was activating the systemic immune response, and wound healing occurs with downward transport. The spatial metabolic characteristics of skin tissue from patients with DFUs indicated downregulated levels of stearoylcarnitine and the glycerophospholipid metabolism pathway in skin tissue from patients with severe DFUs. Finally, the expressions of PRNP (prion protein) to activate the immune response, PLCB3 (PLCB3, phospholipase C beta 3) and VE-cadherin to play roles in neovascularization, and PPDPF (pancreatic progenitor cell differentiation and proliferation factor), LAMC2 (laminin subunit gamma 2) and SPRR2G (small proline rich protein 2G) to facilitate the developmental process mainly keratinocyte differentiation were statistically significant in skin tissues through transcriptomic and RT-qPCR analysis. CONCLUSION Tibial cortex transverse transport (TTT) surgery demonstrates favorable outcomes for patients with severe type 2 DFUs by activating a systemic immune response, contributing to anti-infection, ulcer recurrence, and the limb salvage rate for unilateral or bilateral DFUs. The specific clinical immune responses, candidate proteins, genes, and metabolic characteristics provide directions for in-depth mechanistic research on TTT surgery. Further research and public awareness are needed to optimize TTT surgery in patients with severe type 2 DFUs.
Collapse
Affiliation(s)
- Lin Yu
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China
- NHC Key Laboratory of Nuclear Surgery Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China
| | - Dingwei Zhang
- Department of Orthopaedics, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Yuan Yin
- NHC Key Laboratory of Nuclear Surgery Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China
| | - Xiaoya Li
- Department of Orthopaedics, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Chunxia Bai
- Department of Orthopaedics, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Qian Zhou
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China
| | - Xinyi Liu
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China
| | - Xiaojun Tian
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China
| | - Daofei Xu
- Department of Orthopaedics, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Xianjun Yu
- Department of Orthopaedics, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Sichun Zhao
- Department of Orthopaedics, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Rong Hu
- Department of Orthopaedics, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Fudie Guo
- Department of Orthopaedics, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Yuwei Yang
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China
| | - Yan Ren
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China
| | - Gang Chen
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China
| | - Jiawei Zeng
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China
| | - Jiafu Feng
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China
| |
Collapse
|
6
|
Kang J, Lee H, Joo J, Song J, Kim H, Kim YH, Park HR. Comparison of genetic and epigenetic profiles of periodontitis according to the presence of type 2 diabetes. MedComm (Beijing) 2024; 5:e620. [PMID: 38903536 PMCID: PMC11187843 DOI: 10.1002/mco2.620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/22/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) and periodontitis (PD) have intricated connections as chronic inflammatory diseases. While the immune response is a key factor that accounts for their association, the underlying mechanisms remain unclear. To gain a deeper understanding of the connection, we conducted research using a multiomics approach. We generated whole genome and methylation profiling array data from the periodontium of PD patients with DM (PDDM) and without DM to confirm genetic and epigenetic changes. Independent bulk and single-cell RNA sequencing data were employed to verify the expression levels of hypo-methylated genes. We observed a gradual rise in C>T base substitutions and hypomethylation in PD and PDDM patients compared with healthy participants. Furthermore, specific genetic and epigenetic alterations were prominently associated with the Fc-gamma receptor-mediated phagocytosis pathway. The upregulation of these genes was confirmed in both the periodontal tissues of PD patients and the pancreatic tissues of T2DM patients. Through single-cell RNA analysis of peripheral blood mononuclear cells, substantial upregulation of Fc-gamma receptors and related genes was particularly identified in monocytes. Our findings suggest that targeting the Fc-gamma signaling pathway in monocytes holds promise as a potential treatment strategy for managing systemic complications associated with diabetes.
Collapse
Affiliation(s)
- Junho Kang
- Department of ResearchKeimyung University Dongsan Medical CenterDalseo‐guDaeguRepublic of Korea
| | - Hansong Lee
- Department of BioMedical InformaticsMedical Research Institute, Pusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
| | - Ji‐Young Joo
- Department of PeriodontologySchool of DentistryPusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
| | - Jae‐Min Song
- Department of Oral and Maxillofacial SurgerySchool of DentistryPusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
| | - Hyun‐Joo Kim
- Department of PeriodontologyDental and Life Science InstituteSchool of DentistryPusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
- Department of Periodontology and Dental Research InstitutePusan National University Dental HospitalYangsan‐siGyeongsangnam‐doRepublic of Korea
- Periodontal Disease Signaling Network Research CenterSchool of DentistryPusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
| | - Yun Hak Kim
- Periodontal Disease Signaling Network Research CenterSchool of DentistryPusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
- Department of Biomedical Informatics School of MedicinePusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
- Department of AnatomySchool of MedicinePusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
| | - Hae Ryoun Park
- Department of Periodontology and Dental Research InstitutePusan National University Dental HospitalYangsan‐siGyeongsangnam‐doRepublic of Korea
- Periodontal Disease Signaling Network Research CenterSchool of DentistryPusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
- Department of Oral PathologySchool of DentistryPusan National UniversityYangsan‐siGyeongsangnam‐doRepublic of Korea
| |
Collapse
|
7
|
L’Estrange-Stranieri E, Gottschalk TA, Wright MD, Hibbs ML. The dualistic role of Lyn tyrosine kinase in immune cell signaling: implications for systemic lupus erythematosus. Front Immunol 2024; 15:1395427. [PMID: 39007135 PMCID: PMC11239442 DOI: 10.3389/fimmu.2024.1395427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/17/2024] [Indexed: 07/16/2024] Open
Abstract
Systemic lupus erythematosus (SLE, lupus) is a debilitating, multisystem autoimmune disease that can affect any organ in the body. The disease is characterized by circulating autoantibodies that accumulate in organs and tissues, which triggers an inflammatory response that can cause permanent damage leading to significant morbidity and mortality. Lyn, a member of the Src family of non-receptor protein tyrosine kinases, is highly implicated in SLE as remarkably both mice lacking Lyn or expressing a gain-of-function mutation in Lyn develop spontaneous lupus-like disease due to altered signaling in B lymphocytes and myeloid cells, suggesting its expression or activation state plays a critical role in maintaining tolerance. The past 30 years of research has begun to elucidate the role of Lyn in a duplicitous signaling network of activating and inhibitory immunoreceptors and related targets, including interactions with the interferon regulatory factor family in the toll-like receptor pathway. Gain-of-function mutations in Lyn have now been identified in human cases and like mouse models, cause severe systemic autoinflammation. Studies of Lyn in SLE patients have presented mixed findings, which may reflect the heterogeneity of disease processes in SLE, with impairment or enhancement in Lyn function affecting subsets of SLE patients that may be a means of stratification. In this review, we present an overview of the phosphorylation and protein-binding targets of Lyn in B lymphocytes and myeloid cells, highlighting the structural domains of the protein that are involved in its function, and provide an update on studies of Lyn in SLE patients.
Collapse
Affiliation(s)
- Elan L’Estrange-Stranieri
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Timothy A. Gottschalk
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Mark D. Wright
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Margaret L. Hibbs
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
8
|
Omer S, Li J, Yang CX, Harrison RE. Ninein promotes F-actin cup formation and inward phagosome movement during phagocytosis in macrophages. Mol Biol Cell 2024; 35:ar26. [PMID: 38117588 PMCID: PMC10916867 DOI: 10.1091/mbc.e23-06-0216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/30/2023] [Accepted: 12/12/2023] [Indexed: 12/22/2023] Open
Abstract
Phagocytosis by macrophages is a highly polarized process to destroy large target cells. Binding to particles induces extensive cortical actin-generated forces that drive the formation of elaborate pseudopods around the target particle. Postinternalization, the resultant phagosome is driven toward the cell interior on microtubules (MTs) by cytoplasmic dynein. However, it is unclear whether dynein and cargo-adaptors contribute to the earlier steps of particle internalization and phagosome formation. Here we reveal that ninein, a MT minus-end-associated protein that localizes to the centrosome, is also present at the phagocytic cup in macrophages. Ninein depletion impairs particle internalization by delaying the early F-actin recruitment to sites of particle engagement and cup formation, with no impact on F-actin dynamics beyond this initial step. Ninein forms membrane-bound clusters on phagocytic cups that do not nucleate acentrosomal MTs but instead mediate the assembly of dynein-dynactin complex at active phagocytic membranes. Both ninein depletion and pharmacological inhibition of dynein activity reduced inward displacement of bound particles into macrophages. We found that ninein and dynein motor activity were required for timely retrograde movement of phagosomes and for phagolysosome formation. Taken together, these data show that ninein, alone and with dynein, play significant roles during phagocytosis.
Collapse
Affiliation(s)
- Safia Omer
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4
| | - Jiahao Li
- Department of Cell & Systems Biology, University of Toronto Scarborough, Toronto, Ontario M1C 1A4
| | - Claire X. Yang
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4
| | - Rene E. Harrison
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4
- Department of Cell & Systems Biology, University of Toronto Scarborough, Toronto, Ontario M1C 1A4
| |
Collapse
|
9
|
Mititelu A, Onisâi MC, Roșca A, Vlădăreanu AM. Current Understanding of Immune Thrombocytopenia: A Review of Pathogenesis and Treatment Options. Int J Mol Sci 2024; 25:2163. [PMID: 38396839 PMCID: PMC10889445 DOI: 10.3390/ijms25042163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
The management of immune thrombocytopenia (ITP) and the prediction of patient response to therapy still represent a significant and constant challenge in hematology. ITP is a heterogeneous disease with an unpredictable evolution. Although the pathogenesis of ITP is currently better known and its etiology has been extensively studied, up to 75% of adult patients with ITP may develop chronicity, which represents a significant burden on patients' quality of life. A major risk of ITP is bleeding, but knowledge on the exact relationship between the degree of thrombocytopenia and bleeding symptoms, especially at a lower platelet count, is lacking. The actual management of ITP is based on immune suppression (corticosteroids and intravenous immunoglobulins), or the use of thrombopoietin receptor agonists (TPO-RAs), rituximab, or spleen tyrosine kinase (Syk) inhibitors. A better understanding of the underlying pathology has facilitated the development of a number of new targeted therapies (Bruton's tyrosine kinase inhibitors, neonatal Fc receptors, strategies targeting B and plasma cells, strategies targeting T cells, complement inhibitors, and newer TPO-RAs for improving megakaryopoiesis), which seem to be highly effective and well tolerated and result in a significant improvement in patients' quality of life. The disadvantage is that there is a lack of knowledge of the predictive factors of response to treatments, which would help in the development of an optimized treatment algorithm for selected patients.
Collapse
Affiliation(s)
- Alina Mititelu
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, Emergency University Hospital of Bucharest, 050098 Bucharest, Romania; (M.-C.O.); (A.M.V.)
| | - Minodora-Cezarina Onisâi
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, Emergency University Hospital of Bucharest, 050098 Bucharest, Romania; (M.-C.O.); (A.M.V.)
| | - Adrian Roșca
- Department of Physiology, Carol Davila University of Medicine and Pharmacy, 050471 Bucharest, Romania;
| | - Ana Maria Vlădăreanu
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, Emergency University Hospital of Bucharest, 050098 Bucharest, Romania; (M.-C.O.); (A.M.V.)
| |
Collapse
|
10
|
Zhang S, Hu D, Zhuo Y, Cui L, Li D, Zhang L, Yang L, Wang X. Protective effect of liriodendrin on IgG immune complex-induced acute lung injury via inhibiting SRC/STAT3/MAPK signaling pathway: a network pharmacology research. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3269-3283. [PMID: 37243760 DOI: 10.1007/s00210-023-02534-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/16/2023] [Indexed: 05/29/2023]
Abstract
The primary objectives of this research were to investigate the protective effects of liriodendrin against IgG immune complex (IgG-IC)-induced acute lung injury (ALI) and to elucidate the underlying mechanisms. This study employed a mouse and cell model of IgG-IC-induced acute lung injury. Lung tissue was stained with hematoxylin-eosin to observe pathological alterations and arterial blood gas analysis was tested. Inflammatory cytokines, including interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-alpha (TNF-α), were measured using ELISA. The mRNA expression of inflammatory cytokines was assessed via RT-qPCR. Molecular docking and enrichment analysis were combined to identify the most potential signaling pathways modulated by liriodendrin, which were then verified using western blot analysis in IgG-IC-induced ALI models. We identified 253 shared targets between liriodendrin and IgG-IC-induced acute lung injury from the database. Through network pharmacology, enrichment analysis, and molecular docking, SRC was determined to be the most closely associated target of liriodendrin in IgG-IC-induced ALI. Pretreatment with liriodendrin notably reduced the increased cytokine secretion of IL-1β, IL-6, and TNF-α. Histopathological analysis of lung tissue demonstrated a protective effect of liriodendrin on IgG-IC-induced acute lung injury in mice. Arterial blood gas analysis showed liriodendrin ameliorated acidosis and hypoxemia efficiently. Further studies revealed that liriodendrin pretreatment substantially attenuated the elevated phosphorylation levels of SRC's downstream components (JNK, P38, and STAT3), suggesting that liriodendrin may protect against IgG-IC-induced ALI via the SRC/STAT3/MAPK pathway. Our findings indicate that liriodendrin protects against IgG-IC-induced acute lung injury by inhibiting the SRC/STAT3/MAPK signaling pathway, suggesting that liriodendrin may serve as a potential treatment for acute lung injury caused by IgG-IC.
Collapse
Affiliation(s)
- Sijia Zhang
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Dongsheng Hu
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Yuzhen Zhuo
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Lingzhi Cui
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Dihua Li
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Lanqiu Zhang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Lei Yang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China.
| | - Ximo Wang
- Graduate School, Tianjin Medical University, Tianjin, China.
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China.
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin University, Tianjin, China.
| |
Collapse
|
11
|
Pei L, Hou Y, Feng Y, Li F, Su H, Zhang Y, Song Y, Liu K, Cao G. Equine β-defensin 1 regulates cytokine expression and phagocytosis in S. aureus-infected mouse monocyte macrophages via the Paxillin-FAK-PI3K pathway. Int Immunopharmacol 2023; 123:110793. [PMID: 37582311 DOI: 10.1016/j.intimp.2023.110793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 08/17/2023]
Abstract
β-defensin-1 (BD-1) is a rich source of disulfide bonds and antibacterial peptides that exhibit direct bactericidal function. The expression of BD-1 is primarily induced by external stimulation and is known to correlate with TLR-mediated inflammation, suggesting its association with innate immune responses. Equine β-defensin-1 (eBD-1) belongs to the BD-1 family. Our previous study demonstrated that eBD-1 enhances cytokine expression and promotes macrophage phagocytosis of S. aureus, although the underlying mechanism remains unknown. In this study, we utilized a PI-3K inhibitor (PKI-402) to treat eBD-1 -treated S. aureus-infected macrophages in vitro. Our results revealed that PKI-402 decreased the expression of eBD-1-promoted TNF-α, IL-6, CXCL10, CD40, RANTES, and p65 mRNA. To further investigate the relationship between eBD-1 and phagocytosis, we examined the expression of paxillin and FcγRIII (CD16 receptor) using western blot and immunofluorescence techniques. Our findings demonstrated that eBD-1 enhanced CD16 and paxillin expression in S. aureus -infected macrophages. Considering the correlation between paxillin expression and focal adhesion kinase (FAK), we transfected FAK siRNA into macrophages and evaluated paxillin expression using western blot analysis. Additionally, we quantified the number of S. aureus phagocytosed by macrophages. The results indicated a reduction in both paxillin expression and the number of S. aureus phagocytosed by macrophages upon FAK siRNA treatment. Our study showed the eBD-1 promotes cytokine mRNA expression in S. aureus-infected macrophages regulated by PI-3K-NF-κB pathway, and it increases macrophage phagocytosis of S. aureus associated with the FAK-paxillin signaling pathway.
Collapse
Affiliation(s)
- Le Pei
- Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, 010013, China; Inner Mongolia Key Laboratory of Basic Veterinary Medicine, College of Veterinary, Inner Mongolia Agricultural University, Huhhot, 010018, China
| | - Yongyue Hou
- Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, 010013, China
| | - Ying Feng
- Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, 010013, China
| | - Feng Li
- Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, 010013, China
| | - Hong Su
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, College of Veterinary, Inner Mongolia Agricultural University, Huhhot, 010018, China
| | - Yuemei Zhang
- Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, 010013, China
| | - Yue Song
- Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, 010013, China
| | - Kun Liu
- School of Public Healthy, Inner Mongolia Medical University, Huhhot 010110, China.
| | - Guifang Cao
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, College of Veterinary, Inner Mongolia Agricultural University, Huhhot, 010018, China.
| |
Collapse
|
12
|
Bossart J, Rippl A, Barton Alston AE, Flühmann B, Digigow R, Buljan M, Ayala-Nunez V, Wick P. Uncovering the dynamics of cellular responses induced by iron-carbohydrate complexes in human macrophages using quantitative proteomics and phosphoproteomics. Biomed Pharmacother 2023; 166:115404. [PMID: 37657262 DOI: 10.1016/j.biopha.2023.115404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/22/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023] Open
Abstract
Iron-carbohydrate complexes are widely used to treat iron deficiencies. Macrophages play a crucial role in the uptake and fate of these nanomedicines, however, how complexed iron carbohydrates are taken up and metabolized by macrophages is still not fully understood. Using a (phospho-)proteomics approach, we assessed differences in protein expression and phosphorylation in M2 macrophages triggered by iron sucrose (IS). Our results show that IS alters the expression of multiple receptors, indicative of a complex entry mechanism. Besides, IS induced an increase in intracellular ferritin, the loss of M2 polarization, protective mechanisms against ferroptosis, and an autophagic response. These data indicate that macrophages can use IS as a source of iron for its storage and later release, however, the excess of iron can cause oxidative stress, which can be successfully regulated by the cells. When comparing IS with ferric carboxymaltose (FCM) and iron isomaltoside-1000 (IIM), complexes with a higher carbohydrate ligand stability, we observed that FCM and IIM are metabolized at a slower rate, and trigger M2 polarization loss to a lower extent. These results indicate that the surface characteristics of the iron-carbohydrate complexes may influence the cell responses. Our data show that the application of (phospho-)proteomics can lead to a better understanding of metabolic processes, including the uptake, biodegradation and bioavailability of nanomedicines.
Collapse
Affiliation(s)
- Jonas Bossart
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Particles-Biology Interactions Laboratory, CH-9014 St. Gallen, Switzerland; SIB, Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland; ETH Zurich, Department of Health Sciences and Technology (D-HEST), CH-8093 Zurich, Switzerland
| | - Alexandra Rippl
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Particles-Biology Interactions Laboratory, CH-9014 St. Gallen, Switzerland
| | | | | | | | - Marija Buljan
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Particles-Biology Interactions Laboratory, CH-9014 St. Gallen, Switzerland; SIB, Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Vanesa Ayala-Nunez
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Particles-Biology Interactions Laboratory, CH-9014 St. Gallen, Switzerland.
| | - Peter Wick
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Particles-Biology Interactions Laboratory, CH-9014 St. Gallen, Switzerland.
| |
Collapse
|
13
|
Taya T, Teruyama F, Gojo S. Host-directed therapy for bacterial infections -Modulation of the phagolysosome pathway. Front Immunol 2023; 14:1227467. [PMID: 37841276 PMCID: PMC10570837 DOI: 10.3389/fimmu.2023.1227467] [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: 05/23/2023] [Accepted: 09/11/2023] [Indexed: 10/17/2023] Open
Abstract
Bacterial infections still impose a significant burden on humanity, even though antimicrobial agents have long since been developed. In addition to individual severe infections, the f fatality rate of sepsis remains high, and the threat of antimicrobial-resistant bacteria grows with time, putting us at inferiority. Although tremendous resources have been devoted to the development of antimicrobial agents, we have yet to recover from the lost ground we have been driven into. Looking back at the evolution of treatment for cancer, which, like infectious diseases, has the similarity that host immunity eliminates the lesion, the development of drugs to eliminate the tumor itself has shifted from a single-minded focus on drug development to the establishment of a treatment strategy in which the de-suppression of host immunity is another pillar of treatment. In infectious diseases, on the other hand, the development of therapies that strengthen and support the immune system has only just begun. Among innate immunity, the first line of defense that bacteria encounter after invading the host, the molecular mechanisms of the phagolysosome pathway, which begins with phagocytosis to fusion with lysosome, have been elucidated in detail. Bacteria have a large number of strategies to escape and survive the pathway. Although the full picture is still unfathomable, the molecular mechanisms have been elucidated for some of them, providing sufficient clues for intervention. In this article, we review the host defense mechanisms and bacterial evasion mechanisms and discuss the possibility of host-directed therapy for bacterial infection by intervening in the phagolysosome pathway.
Collapse
Affiliation(s)
- Toshihiko Taya
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Fumiya Teruyama
- Pharmacology Research Department, Tokyo New Drug Research Laboratories, Kowa Company, Ltd., Tokyo, Japan
- Department of Regenerative Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoshi Gojo
- Department of Regenerative Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
14
|
Gunalp S, Helvaci DG, Oner A, Bursalı A, Conforte A, Güner H, Karakülah G, Szegezdi E, Sag D. TRAIL promotes the polarization of human macrophages toward a proinflammatory M1 phenotype and is associated with increased survival in cancer patients with high tumor macrophage content. Front Immunol 2023; 14:1209249. [PMID: 37809073 PMCID: PMC10551148 DOI: 10.3389/fimmu.2023.1209249] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/30/2023] [Indexed: 10/10/2023] Open
Abstract
Background TNF-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that can either induce cell death or activate survival pathways after binding to death receptors (DRs) DR4 or DR5. TRAIL is investigated as a therapeutic agent in clinical trials due to its selective toxicity to transformed cells. Macrophages can be polarized into pro-inflammatory/tumor-fighting M1 macrophages or anti-inflammatory/tumor-supportive M2 macrophages and an imbalance between M1 and M2 macrophages can promote diseases. Therefore, identifying modulators that regulate macrophage polarization is important to design effective macrophage-targeted immunotherapies. The impact of TRAIL on macrophage polarization is not known. Methods Primary human monocyte-derived macrophages were pre-treated with either TRAIL or with DR4 or DR5-specific ligands and then polarized into M1, M2a, or M2c phenotypes in vitro. The expression of M1 and M2 markers in macrophage subtypes was analyzed by RNA sequencing, qPCR, ELISA, and flow cytometry. Furthermore, the cytotoxicity of the macrophages against U937 AML tumor targets was assessed by flow cytometry. TCGA datasets were also analyzed to correlate TRAIL with M1/M2 markers, and the overall survival of cancer patients. Results TRAIL increased the expression of M1 markers at both mRNA and protein levels while decreasing the expression of M2 markers at the mRNA level in human macrophages. TRAIL also shifted M2 macrophages towards an M1 phenotype. Our data showed that both DR4 and DR5 death receptors play a role in macrophage polarization. Furthermore, TRAIL enhanced the cytotoxicity of macrophages against the AML cancer cells in vitro. Finally, TRAIL expression was positively correlated with increased expression of M1 markers in the tumors from ovarian and sarcoma cancer patients and longer overall survival in cases with high, but not low, tumor macrophage content. Conclusions TRAIL promotes the polarization of human macrophages toward a proinflammatory M1 phenotype via both DR4 and DR5. Our study defines TRAIL as a new regulator of macrophage polarization and suggests that targeting DRs can enhance the anti-tumorigenic response of macrophages in the tumor microenvironment by increasing M1 polarization.
Collapse
Affiliation(s)
- Sinem Gunalp
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Genomic Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | - Derya Goksu Helvaci
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Faculty of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Aysenur Oner
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Genomic Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | | | - Alessandra Conforte
- School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Hüseyin Güner
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Molecular Biology and Genetics, Faculty of Life and Natural Science, Abdullah Gül University, Kayseri, Türkiye
| | - Gökhan Karakülah
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Genomic Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | - Eva Szegezdi
- School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Duygu Sag
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Genomic Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, Izmir, Türkiye
| |
Collapse
|
15
|
Chen M, Menon MC, Wang W, Fu J, Yi Z, Sun Z, Liu J, Li Z, Mou L, Banu K, Lee SW, Dai Y, Anandakrishnan N, Azeloglu EU, Lee K, Zhang W, Das B, He JC, Wei C. HCK induces macrophage activation to promote renal inflammation and fibrosis via suppression of autophagy. Nat Commun 2023; 14:4297. [PMID: 37463911 PMCID: PMC10354075 DOI: 10.1038/s41467-023-40086-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 07/12/2023] [Indexed: 07/20/2023] Open
Abstract
Renal inflammation and fibrosis are the common pathways leading to progressive chronic kidney disease (CKD). We previously identified hematopoietic cell kinase (HCK) as upregulated in human chronic allograft injury promoting kidney fibrosis; however, the cellular source and molecular mechanisms are unclear. Here, using immunostaining and single cell sequencing data, we show that HCK expression is highly enriched in pro-inflammatory macrophages in diseased kidneys. HCK-knockout (KO) or HCK-inhibitor decreases macrophage M1-like pro-inflammatory polarization, proliferation, and migration in RAW264.7 cells and bone marrow-derived macrophages (BMDM). We identify an interaction between HCK and ATG2A and CBL, two autophagy-related proteins, inhibiting autophagy flux in macrophages. In vivo, both global or myeloid cell specific HCK-KO attenuates renal inflammation and fibrosis with reduces macrophage numbers, pro-inflammatory polarization and migration into unilateral ureteral obstruction (UUO) kidneys and unilateral ischemia reperfusion injury (IRI) models. Finally, we developed a selective boron containing HCK inhibitor which can reduce macrophage pro-inflammatory activity, proliferation, and migration in vitro, and attenuate kidney fibrosis in the UUO mice. The current study elucidates mechanisms downstream of HCK regulating macrophage activation and polarization via autophagy in CKD and identifies that selective HCK inhibitors could be potentially developed as a new therapy for renal fibrosis.
Collapse
Affiliation(s)
- Man Chen
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Critical Care Medicine, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
- Department of Critical Care Medicine, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Madhav C Menon
- Division of Nephrology, Yale School of Medicine, New Haven, CT, USA
| | - Wenlin Wang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Fu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhengzi Yi
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zeguo Sun
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Liu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhengzhe Li
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lingyun Mou
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Khadija Banu
- Division of Nephrology, Yale School of Medicine, New Haven, CT, USA
| | - Sui-Wan Lee
- Center for Comparative Medicine and Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ying Dai
- Center for Comparative Medicine and Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nanditha Anandakrishnan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evren U Azeloglu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kyung Lee
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Weijia Zhang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bhaskar Das
- Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA.
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Renal Section, James J. Peters VAMC, Bronx, NY, USA.
| | - Chengguo Wei
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
16
|
Fu W, Xie Z, Bai M, Zhang Z, Zhao Y, Tian J. Proteomics analysis of methionine enkephalin upregulated macrophages against infection by the influenza-A virus. Proteome Sci 2023; 21:4. [PMID: 37041527 PMCID: PMC10088144 DOI: 10.1186/s12953-023-00205-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/03/2023] [Indexed: 04/13/2023] Open
Abstract
Macrophages have a vital role in phagocytosis and antiviral effect against invading influenza viruses. Previously, we found that methionine enkephalin (MENK) inhibited influenza virus infection by upregulating the "antiviral state" of macrophages. To investigate the immunoregulatory mechanism of action of MENK on macrophages, we employed proteomic analysis to identify differentially expressed proteins (DEPs) between macrophages infected with the influenza-A virus and cells infected with the influenza-A virus after pretreatment with MENK. A total of 215 DEPs were identified: 164 proteins had upregulated expression and 51 proteins had downregulated expression. Proteomics analysis showed that DEPs were highly enriched in "cytokine-cytokine receptor interaction", "phagosome", and "complement and coagulation cascades pathway". Proteomics analysis revealed that MENK could be an immune modulator or prophylactic for the prevention and treatment of influenza. MENK promoted the polarization of M1 macrophages, activated inflammatory responses, and enhanced phagocytosis and killing function by upregulating opsonizing receptors.
Collapse
Affiliation(s)
- Wenrui Fu
- Graduate College, Jinzhou Medical University, Jinzhou, 121000, China
| | - Zifeng Xie
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, 121000, China
| | - Mei Bai
- Department of Microbiology, Jinzhou Center for Disease Control and Prevention, Jinzhou, 121000, China
| | - Zhen Zhang
- Department of Microbiology, Jinzhou Center for Disease Control and Prevention, Jinzhou, 121000, China
| | - Yuanlong Zhao
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, 121000, China
| | - Jing Tian
- Department of Immunology, School of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, 121000, China.
| |
Collapse
|
17
|
Cai Y, Zuo X, Zuo Y, Wu S, Pang W, Ma K, Yi Q, Tan L, Deng H, Qu X, Chen X. Transcriptomic analysis reveals shared gene signatures and molecular mechanisms between obesity and periodontitis. Front Immunol 2023; 14:1101854. [PMID: 37063877 PMCID: PMC10090675 DOI: 10.3389/fimmu.2023.1101854] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/24/2023] [Indexed: 03/31/2023] Open
Abstract
BackgroundBoth obesity (OB) and periodontitis (PD) are chronic non-communicable diseases, and numerous epidemiological studies have demonstrated the association between these two diseases. However, the molecular mechanisms that could explain the association between OB and PD are largely unclear. This study aims to investigate the common gene signatures and biological pathways in OB and PD through bioinformatics analysis of publicly available transcriptome datasets.MethodsThe RNA expression profile datasets of OB (GSE104815) and PD (GSE106090) were used as training data, and GSE152991 and GSE16134 as validation data. After screening for differentially expressed genes (DEGs) shared by OB and PD, gene enrichment analysis, protein-protein interaction (PPI) network construction, GeneMANIA analysis, immune infiltration analysis and gene set enrichment analysis (GSEA) were performed. In addition, receiver operating characteristic (ROC) curves were used to assess the predictive accuracy of the hub gene. Finally, we constructed the hub gene-associated TF-miRNA-mRNA regulatory network.ResultsWe identified a total of 147 DEGs shared by OB and PD (38 down-regulated and 109 up-regulated). Functional analysis showed that these genes were mainly enriched in immune-related pathways such as B cell receptor signalling, leukocyte migration and cellular defence responses. 14 hub genes (FGR, MNDA, NCF2, FYB1, EVI2B, LY86, IGSF6, CTSS, CXCR4, LCK, FCN1, CXCL2, P2RY13, MMP7) showed high sensitivity and specificity in the ROC curve analysis. The results of immune infiltration analysis showed that immune cells such as macrophages, activated CD4 T cells and immune B cells were present at high infiltration levels in both OB and PD samples.The results of GeneMANIA analysis and GSEA analysis suggested that five key genes (FGR, LCK, FYB1, LY86 and P2RY13) may be strongly associated with macrophages. Finally, we constructed a TF-miRNA-mRNA regulatory network consisting of 233 transcription factors (TFs), 8 miRNAs and 14 mRNAs based on the validated information obtained from the database.ConclusionsFive key genes (FGR, LCK, FYB1, LY86, P2RY13) may be important biomarkers of OB and PD. These genes may play an important role in the pathogenesis of OB and PD by affecting macrophage activity and participating in immune regulation and inflammatory responses.
Collapse
Affiliation(s)
- Yisheng Cai
- Laboratory of Molecular and Statistical Genetics and Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xuemei Zuo
- Laboratory of Molecular and Statistical Genetics and Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yuyang Zuo
- Laboratory of Molecular and Statistical Genetics and Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Shuang Wu
- Laboratory of Molecular and Statistical Genetics and Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Weiwei Pang
- Laboratory of Molecular and Statistical Genetics and Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Keqiang Ma
- Laboratory of Molecular and Statistical Genetics and Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Qiaorong Yi
- Laboratory of Molecular and Statistical Genetics and Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Lijun Tan
- Laboratory of Molecular and Statistical Genetics and Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Hongwen Deng
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, United States
| | - Xiaochao Qu
- Laboratory of Molecular and Statistical Genetics and Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, China
- *Correspondence: Xiaochao Qu, ; Xiangding Chen,
| | - Xiangding Chen
- Laboratory of Molecular and Statistical Genetics and Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, China
- *Correspondence: Xiaochao Qu, ; Xiangding Chen,
| |
Collapse
|
18
|
Francis D, Bhairaddy A, Joy A, Hari GV, Francis A. Secretory proteins in the orchestration of microbial virulence: The curious case of Staphylococcus aureus. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 133:271-350. [PMID: 36707204 DOI: 10.1016/bs.apcsb.2022.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Microbial virulence showcases an excellent model for adaptive changes that enable an organism to survive and proliferate in a hostile environment and exploit host resources to its own benefit. In Staphylococcus aureus, an opportunistic pathogen of the human host, known for the diversity of the disease conditions it inflicts and the rapid evolution of antibiotic resistance, virulence is a consequence of having a highly plastic genome that is amenable to quick reprogramming and the ability to express a diverse arsenal of virulence factors. Virulence factors that are secreted to the host milieu effectively manipulate the host conditions to favor bacterial survival and growth. They assist in colonization, nutrient acquisition, immune evasion, and systemic spread. The structural and functional characteristics of the secreted virulence proteins have been shaped to assist S. aureus in thriving and disseminating effectively within the host environment and exploiting the host resources to its best benefit. With the aim of highlighting the importance of secreted virulence proteins in bacterial virulence, the present chapter provides a comprehensive account of the role of the major secreted proteins of S. aureus in orchestrating its virulence in the human host.
Collapse
Affiliation(s)
- Dileep Francis
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India.
| | - Anusha Bhairaddy
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India
| | - Atheene Joy
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India
| | | | - Ashik Francis
- Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| |
Collapse
|
19
|
Lalnunthangi A, Dakpa G, Tiwari S. Multifunctional role of the ubiquitin proteasome pathway in phagocytosis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 194:179-217. [PMID: 36631192 DOI: 10.1016/bs.pmbts.2022.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Phagocytosis is a specialized form of endocytosis where large cells and particles (>0.5μm) are engulfed by the phagocytic cells, and ultimately digested in the phagolysosomes. This process not only eliminates unwanted particles and pathogens from the extracellular sources, but also eliminates apoptotic cells within the body, and is critical for maintenance of tissue homeostasis. It is believed that both endocytosis and phagocytosis share common pathways after particle internalization, but specialized features and differences between these two routes of internalization are also likely. The recruitment and removal of each protein/particle during the maturation of endocytic/phagocytic vesicles has to be tightly regulated to ensure their timely action. Ubiquitin proteasome pathway (UPP), degrades unwanted proteins by post-translational modification of proteins with chains of conserved protein Ubiquitin (Ub), with subsequent recognition of Ub chains by the 26S proteasomes and substrate degradation by this protease. This pathway utilizes different Ub linkages to modify proteins to regulate protein-protein interaction, localization, and activity. Due to its vast number of targets, it is involved in many cellular pathways, including phagocytosis. This chapters describes the basic steps and signaling in phagocytosis and different roles that UPP plays at multiple steps in regulating phagocytosis directly, or through its interaction with other phagosomal proteins. How aberrations in UPP function affect phagocytosis and their association with human diseases, and how pathogens exploit this pathway for their own benefit is also discussed.
Collapse
Affiliation(s)
| | | | - Swati Tiwari
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
20
|
Abstract
Neurons in the developing brain undergo extensive structural refinement as nascent circuits adopt their mature form. This physical transformation of neurons is facilitated by the engulfment and degradation of axonal branches and synapses by surrounding glial cells, including microglia and astrocytes. However, the small size of phagocytic organelles and the complex, highly ramified morphology of glia have made it difficult to define the contribution of these and other glial cell types to this crucial process. Here, we used large-scale, serial section transmission electron microscopy (TEM) with computational volume segmentation to reconstruct the complete 3D morphologies of distinct glial types in the mouse visual cortex, providing unprecedented resolution of their morphology and composition. Unexpectedly, we discovered that the fine processes of oligodendrocyte precursor cells (OPCs), a population of abundant, highly dynamic glial progenitors, frequently surrounded small branches of axons. Numerous phagosomes and phagolysosomes (PLs) containing fragments of axons and vesicular structures were present inside their processes, suggesting that OPCs engage in axon pruning. Single-nucleus RNA sequencing from the developing mouse cortex revealed that OPCs express key phagocytic genes at this stage, as well as neuronal transcripts, consistent with active axon engulfment. Although microglia are thought to be responsible for the majority of synaptic pruning and structural refinement, PLs were ten times more abundant in OPCs than in microglia at this stage, and these structures were markedly less abundant in newly generated oligodendrocytes, suggesting that OPCs contribute substantially to the refinement of neuronal circuits during cortical development.
Collapse
|
21
|
Xiong Y, Li Y, Cui X, Zhang L, Yang X, Liu H. ADAP restraint of STAT1 signaling regulates macrophage phagocytosis in immune thrombocytopenia. Cell Mol Immunol 2022; 19:898-912. [PMID: 35637282 PMCID: PMC9149338 DOI: 10.1038/s41423-022-00881-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 05/09/2022] [Indexed: 01/08/2023] Open
Abstract
Heightened platelet phagocytosis by macrophages accompanied by an increase in IFN-γ play key roles in the etiology of immune thrombocytopenia (ITP); however, it remains elusive how macrophage-mediated platelet clearance is regulated in ITP. Here, we report that adhesion and degranulation-protein adaptor protein (ADAP) restrains platelet phagocytosis by macrophages in ITP via modulation of signal transducer and activator of transcription 1 (STAT1)-FcγR signaling. We show that ITP was associated with the underexpression of ADAP in splenic macrophages. Furthermore, macrophages from Adap-/- mice exhibited elevated platelet phagocytosis and upregulated proinflammatory signaling, and thrombocytopenia in Adap-/- mice was mitigated by the depletion of macrophages. Mechanistically, ADAP interacted and competed with STAT1 binding to importin α5. ADAP deficiency potentiated STAT1 nuclear entry, leading to a selective enhancement of FcγRI/IV transcription in macrophages. Moreover, pharmacological inhibition of STAT1 or disruption of the STAT1-importin α5 interaction relieved thrombocytopenia in Adap-/- mice. Thus, our findings not only reveal a critical role for ADAP as an intracellular immune checkpoint for shaping macrophage phagocytosis in ITP but also identify the ADAP-STAT1-importin α5 module as a promising therapeutic target in the treatment of ITP.
Collapse
Affiliation(s)
- Yiwei Xiong
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Yanli Li
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Xinxing Cui
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Lifeng Zhang
- Department of General Surgery, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou, 215123, China
| | - Xiaodong Yang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Hebin Liu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China.
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China.
| |
Collapse
|
22
|
Chai K, Zhang X, Chen S, Gu H, Tang H, Cao P, Wang G, Ye W, Wan F, Liang J, Shen D. Application of weighted co-expression network analysis and machine learning to identify the pathological mechanism of Alzheimer's disease. Front Aging Neurosci 2022; 14:837770. [PMID: 35912089 PMCID: PMC9326231 DOI: 10.3389/fnagi.2022.837770] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Aberrant deposits of neurofibrillary tangles (NFT), the main characteristic of Alzheimer's disease (AD), are highly related to cognitive impairment. However, the pathological mechanism of NFT formation is still unclear. This study explored differences in gene expression patterns in multiple brain regions [entorhinal, temporal, and frontal cortex (EC, TC, FC)] with distinct Braak stages (0- VI), and identified the hub genes via weighted gene co-expression network analysis (WGCNA) and machine learning. For WGCNA, consensus modules were detected and correlated with the single sample gene set enrichment analysis (ssGSEA) scores. Overlapping the differentially expressed genes (DEGs, Braak stages 0 vs. I-VI) with that in the interest module, metascape analysis, and Random Forest were conducted to explore the function of overlapping genes and obtain the most significant genes. We found that the three brain regions have high similarities in the gene expression pattern and that oxidative damage plays a vital role in NFT formation via machine learning. Through further filtering of genes from interested modules by Random Forest, we screened out key genes, such as LYN, LAPTM5, and IFI30. These key genes, including LYN, LAPTM5, and ARHGDIB, may play an important role in the development of AD through the inflammatory response pathway mediated by microglia.
Collapse
Affiliation(s)
- Keping Chai
- Department of Pediatrics, Zhejiang Hospital, Hangzhou, China
- *Correspondence: Keping Chai
| | - Xiaolin Zhang
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan, China
| | - Shufang Chen
- Department of Pediatrics, Zhejiang Hospital, Hangzhou, China
| | - Huaqian Gu
- Department of Pediatrics, Zhejiang Hospital, Hangzhou, China
| | - Huitao Tang
- Department of Pediatrics, Zhejiang Hospital, Hangzhou, China
| | - Panlong Cao
- Department of Pediatrics, Zhejiang Hospital, Hangzhou, China
| | - Gangqiang Wang
- Department of Pediatrics, Zhejiang Hospital, Hangzhou, China
| | - Weiping Ye
- Department of Pediatrics, Zhejiang Hospital, Hangzhou, China
| | - Feng Wan
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan, China
- Feng Wan
| | - Jiawei Liang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
- Jiawei Liang
| | - Daojiang Shen
- Department of Pediatrics, Zhejiang Hospital, Hangzhou, China
- Daojiang Shen
| |
Collapse
|
23
|
Wan J, Zhao X, Liu J, Chen K, Li C. Src kinase mediates coelomocytes phagocytosis via interacting with focal adhesion kinase in Vibrio splendidus challenged Apostichopus japonicus. FISH & SHELLFISH IMMUNOLOGY 2022; 124:411-420. [PMID: 35462003 DOI: 10.1016/j.fsi.2022.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 06/14/2023]
Abstract
Immune cells have many efficient ways to participate in the host immunity, including phagocytosis, which is an important pathway to eliminate pathogens. Only β-integrin-mediated phagocytosis pathways have been confirmed in Apostichopus japonicus. The Src family kinases (SFKs), a class of non-receptor tyrosine kinases plays an important role in the regulation of phagocytic signals in invertebrates. However, the SFK-mediated phagocytic mechanism is largely unknown in A. japonicus. In this study, a novel SFK homologue (AjSrc) with a conservative SH3 domain, an SH2 domain, and a tyrosine kinase domain was identified from A. japonicus. Both gene and protein expression of AjSrc and phosphorylation levels increased under Vibrio splendidus challenged, reaching the highest level at 24 h. Knock-down of AjSrc could depress coelomocytes' phagocytosis by 25% compared to the control group. To better understand the mechanism of AjSrc-mediated phagocytosis, focal adhesion kinase (FAK) was identified by a Co-immunoprecipitation experiment to be verified as an interactive protein of AjSrc. The phagocytosis rates of coelomocytes were decreased by 33% and 37% in AjFAK and AjSrc + AjFAK interference groups compared with the control group, respectively. Furthermore, the phosphorylation level of AjFAK was increased and reached the maximum level at 24 h post V. splendidus infection, as the same as that of AjSrc. Our results suggested that AjSrc could mediate V. splendidus-induced coelomocytes' phagocytosis via interacting with AjFAK and co-phosphorylation. This study enriched the mechanism of phagocytosis in echinoderm and provided the new theoretical foundation for disease control of sea cucumber.
Collapse
Affiliation(s)
- Junjie Wan
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China; State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, 315211, PR China
| | - Xuelin Zhao
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China; State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, 315211, PR China.
| | - Jiqing Liu
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China; State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, 315211, PR China
| | - Kaiyu Chen
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China; State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, 315211, PR China
| | - Chenghua Li
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China; State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, 315211, PR China.
| |
Collapse
|
24
|
Cao X, Chen J, Li B, Dang J, Zhang W, Zhong X, Wang C, Raoof M, Sun Z, Yu J, Fakih MG, Feng M. Promoting antibody-dependent cellular phagocytosis for effective macrophage-based cancer immunotherapy. SCIENCE ADVANCES 2022; 8:eabl9171. [PMID: 35302839 PMCID: PMC8932662 DOI: 10.1126/sciadv.abl9171] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/26/2022] [Indexed: 05/16/2023]
Abstract
Macrophages are essential in eliciting antibody-dependent cellular phagocytosis (ADCP) of cancer cells. However, a satisfactory anticancer efficacy of ADCP is contingent on early antibody administration, and resistance develops along with cancer progression. Here, we investigate the mechanisms underlying ADCP and demonstrate an effective combinatorial strategy to potentiate its efficacy. We identified paclitaxel as a universal adjuvant that efficiently potentiated ADCP by a variety of anticancer antibodies in multiple cancers. Rather than eliciting cytotoxicity on cancer cells, paclitaxel polarized macrophages toward a state with enhanced phagocytic ability. Paclitaxel-treated macrophages down-regulated cell surface CSF1R whose expression was negatively correlated with patient survival in multiple malignancies. The suppression of CSF1R in macrophages enhanced ADCP of cancer cells, suggesting a role of CSF1R in regulating macrophage phagocytic ability. Together, these findings define a potent strategy for using conventional anticancer drugs to stimulate macrophage phagocytosis and promote the therapeutic efficacy of clinical anticancer antibodies.
Collapse
Affiliation(s)
- Xu Cao
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Jing Chen
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Bolei Li
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Jessica Dang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Wencan Zhang
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Xiancai Zhong
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Chongkai Wang
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA 91010, USA
| | - Mustafa Raoof
- Department of Surgery, City of Hope, Duarte, CA 91010, USA
| | - Zuoming Sun
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Jianhua Yu
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope, Duarte, CA 91010, USA
| | - Marwan G. Fakih
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA 91010, USA
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
25
|
Kralova J, Pavliuchenko N, Fabisik M, Ilievova K, Spoutil F, Prochazka J, Pokorna J, Sedlacek R, Brdicka T. The receptor-type protein tyrosine phosphatase CD45 promotes onset and severity of IL-1β-mediated autoinflammatory osteomyelitis. J Biol Chem 2021; 297:101131. [PMID: 34461100 PMCID: PMC8455366 DOI: 10.1016/j.jbc.2021.101131] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 08/20/2021] [Accepted: 08/26/2021] [Indexed: 11/17/2022] Open
Abstract
A number of human autoinflammatory diseases manifest with severe inflammatory bone destruction. Mouse models of these diseases represent valuable tools that help us to understand molecular mechanisms triggering this bone autoinflammation. The Pstpip2cmo mouse strain is among the best characterized of these; it harbors a mutation resulting in the loss of adaptor protein PSTPIP2 and development of autoinflammatory osteomyelitis. In Pstpip2cmo mice, overproduction of interleukin-1β (IL-1β) and reactive oxygen species by neutrophil granulocytes leads to spontaneous inflammation of the bones and surrounding soft tissues. However, the upstream signaling events leading to this overproduction are poorly characterized. Here, we show that Pstpip2cmo mice deficient in major regulator of Src-family kinases (SFKs) receptor-type protein tyrosine phosphatase CD45 display delayed onset and lower severity of the disease, while the development of autoinflammation is not affected by deficiencies in Toll-like receptor signaling. Our data also show deregulation of pro-IL-1β production by Pstpip2cmo neutrophils that are attenuated by CD45 deficiency. These data suggest a role for SFKs in autoinflammation. Together with previously published work on the involvement of protein tyrosine kinase spleen tyrosine kinase, they point to the role of receptors containing immunoreceptor tyrosine-based activation motifs, which after phosphorylation by SFKs recruit spleen tyrosine kinase for further signal propagation. We propose that this class of receptors triggers the events resulting in increased pro-IL-1β synthesis and disease initiation and/or progression.
Collapse
Affiliation(s)
- Jarmila Kralova
- Laboratory of Leukocyte Signaling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Nataliia Pavliuchenko
- Laboratory of Leukocyte Signaling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Charles University, Faculty of Science, Prague, Czech Republic
| | - Matej Fabisik
- Laboratory of Leukocyte Signaling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Charles University, Faculty of Science, Prague, Czech Republic
| | - Kristyna Ilievova
- Laboratory of Leukocyte Signaling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Frantisek Spoutil
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Jan Prochazka
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic; Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Jana Pokorna
- Laboratory of Leukocyte Signaling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic; Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Tomas Brdicka
- Laboratory of Leukocyte Signaling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
26
|
Nazareth L, St John J, Murtaza M, Ekberg J. Phagocytosis by Peripheral Glia: Importance for Nervous System Functions and Implications in Injury and Disease. Front Cell Dev Biol 2021; 9:660259. [PMID: 33898462 PMCID: PMC8060502 DOI: 10.3389/fcell.2021.660259] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/17/2021] [Indexed: 12/30/2022] Open
Abstract
The central nervous system (CNS) has very limited capacity to regenerate after traumatic injury or disease. In contrast, the peripheral nervous system (PNS) has far greater capacity for regeneration. This difference can be partly attributed to variances in glial-mediated functions, such as axon guidance, structural support, secretion of growth factors and phagocytic activity. Due to their growth-promoting characteristic, transplantation of PNS glia has been trialed for neural repair. After peripheral nerve injuries, Schwann cells (SCs, the main PNS glia) phagocytose myelin debris and attract macrophages to the injury site to aid in debris clearance. One peripheral nerve, the olfactory nerve, is unique in that it continuously regenerates throughout life. The olfactory nerve glia, olfactory ensheathing cells (OECs), are the primary phagocytes within this nerve, continuously clearing axonal debris arising from the normal regeneration of the nerve and after injury. In contrast to SCs, OECs do not appear to attract macrophages. SCs and OECs also respond to and phagocytose bacteria, a function likely critical for tackling microbial invasion of the CNS via peripheral nerves. However, phagocytosis is not always effective; inflammation, aging and/or genetic factors may contribute to compromised phagocytic activity. Here, we highlight the diverse roles of SCs and OECs with the focus on their phagocytic activity under physiological and pathological conditions. We also explore why understanding the contribution of peripheral glia phagocytosis may provide us with translational strategies for achieving axonal regeneration of the injured nervous system and potentially for the treatment of certain neurological diseases.
Collapse
Affiliation(s)
- Lynn Nazareth
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - James St John
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Mariyam Murtaza
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Jenny Ekberg
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| |
Collapse
|
27
|
Dixon CL, Mekhail K, Fairn GD. Examining the Underappreciated Role of S-Acylated Proteins as Critical Regulators of Phagocytosis and Phagosome Maturation in Macrophages. Front Immunol 2021; 12:659533. [PMID: 33868308 PMCID: PMC8047069 DOI: 10.3389/fimmu.2021.659533] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/15/2021] [Indexed: 12/04/2022] Open
Abstract
Phagocytosis is a receptor-mediated process used by cells to engulf a wide variety of particulates, including microorganisms and apoptotic cells. Many of the proteins involved in this highly orchestrated process are post-translationally modified with lipids as a means of regulating signal transduction, membrane remodeling, phagosome maturation and other immunomodulatory functions of phagocytes. S-acylation, generally referred to as S-palmitoylation, is the post-translational attachment of fatty acids to a cysteine residue exposed topologically to the cytosol. This modification is reversible due to the intrinsically labile thioester bond between the lipid and sulfur atom of cysteine, and thus lends itself to a variety of regulatory scenarios. Here we present an overview of a growing number of S-acylated proteins known to regulate phagocytosis and phagosome biology in macrophages.
Collapse
Affiliation(s)
- Charneal L Dixon
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Katrina Mekhail
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Gregory D Fairn
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, Toronto, ON, Canada
| |
Collapse
|
28
|
Rudd CE. How the Discovery of the CD4/CD8-p56 lck Complexes Changed Immunology and Immunotherapy. Front Cell Dev Biol 2021; 9:626095. [PMID: 33791292 PMCID: PMC8005572 DOI: 10.3389/fcell.2021.626095] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/11/2021] [Indexed: 12/22/2022] Open
Abstract
The past 25 years have seen enormous progress in uncovering the receptors and signaling mechanisms on T-cells that activate their various effecter functions. Until the late 1980s, most studies on T-cells had focused on the influx of calcium and the levels of cAMP/GMP in T-cells. My laboratory then uncovered the interaction of CD4 and CD8 co-receptors with the protein-tyrosine kinase p56lck which are now widely accepted as the initiators of the tyrosine phosphorylation cascade leading to T-cell activation. The finding explained how immune recognition receptors expressed by many immune cells, which lack intrinsic catalytic activity, can transduce activation signals via non-covalent association with non-receptor tyrosine kinases. The discovery also established the concept that a protein tyrosine phosphorylation cascade operated in T-cells. In this vein, we and others then showed that the CD4- and CD8-p56lck complexes phosphorylate the TCR complexes which led to the identification of other protein-tyrosine kinases such as ZAP-70 and an array of substrates that are now central to studies in T-cell immunity. Other receptors such as B-cell receptor, Fc receptors and others were also subsequently found to use src kinases to control cell growth. In T-cells, p56lck driven phosphorylation targets include co-receptors such as CD28 and CTLA-4 and immune cell-specific adaptor proteins such as LAT and SLP-76 which act to integrate signals proximal to surface receptors. CD4/CD8-p56lck regulated events in T-cells include intracellular calcium mobilization, integrin activation and the induction of transcription factors for gene expression. Lastly, the identification of the targets of p56lck in the TCR and CD28 provided the framework for the development of chimeric antigen receptor (CAR) therapy in the treatment of cancer. In this review, I outline a history of the development of events that led to the development of the "TCR signaling paradigm" and its implications to immunology and immunotherapy.
Collapse
Affiliation(s)
- Christopher E. Rudd
- Division of Immunology-Oncology, Centre de Recherche Hôpital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Faculty of Medicine, Universite de Montreal, Montreal, QC, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University Health Center, McGill University, Montreal, QC, Canada
| |
Collapse
|
29
|
Pidwill GR, Gibson JF, Cole J, Renshaw SA, Foster SJ. The Role of Macrophages in Staphylococcus aureus Infection. Front Immunol 2021; 11:620339. [PMID: 33542723 PMCID: PMC7850989 DOI: 10.3389/fimmu.2020.620339] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/02/2020] [Indexed: 12/23/2022] Open
Abstract
Staphylococcus aureus is a member of the human commensal microflora that exists, apparently benignly, at multiple sites on the host. However, as an opportunist pathogen it can also cause a range of serious diseases. This requires an ability to circumvent the innate immune system to establish an infection. Professional phagocytes, primarily macrophages and neutrophils, are key innate immune cells which interact with S. aureus, acting as gatekeepers to contain and resolve infection. Recent studies have highlighted the important roles of macrophages during S. aureus infections, using a wide array of killing mechanisms. In defense, S. aureus has evolved multiple strategies to survive within, manipulate and escape from macrophages, allowing them to not only subvert but also exploit this key element of our immune system. Macrophage-S. aureus interactions are multifaceted and have direct roles in infection outcome. In depth understanding of these host-pathogen interactions may be useful for future therapeutic developments. This review examines macrophage interactions with S. aureus throughout all stages of infection, with special emphasis on mechanisms that determine infection outcome.
Collapse
Affiliation(s)
- Grace R. Pidwill
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| | - Josie F. Gibson
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Joby Cole
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Stephen A. Renshaw
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Simon J. Foster
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
30
|
Yang X, Chatterjee V, Ma Y, Zheng E, Yuan SY. Protein Palmitoylation in Leukocyte Signaling and Function. Front Cell Dev Biol 2020; 8:600368. [PMID: 33195285 PMCID: PMC7655920 DOI: 10.3389/fcell.2020.600368] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022] Open
Abstract
Palmitoylation is a post-translational modification (PTM) based on thioester-linkage between palmitic acid and the cysteine residue of a protein. This covalent attachment of palmitate is reversibly and dynamically regulated by two opposing sets of enzymes: palmitoyl acyltransferases containing a zinc finger aspartate-histidine-histidine-cysteine motif (PAT-DHHCs) and thioesterases. The reversible nature of palmitoylation enables fine-tuned regulation of protein conformation, stability, and ability to interact with other proteins. More importantly, the proper function of many surface receptors and signaling proteins requires palmitoylation-meditated partitioning into lipid rafts. A growing number of leukocyte proteins have been reported to undergo palmitoylation, including cytokine/chemokine receptors, adhesion molecules, pattern recognition receptors, scavenger receptors, T cell co-receptors, transmembrane adaptor proteins, and signaling effectors including the Src family of protein kinases. This review provides the latest findings of palmitoylated proteins in leukocytes and focuses on the functional impact of palmitoylation in leukocyte function related to adhesion, transmigration, chemotaxis, phagocytosis, pathogen recognition, signaling activation, cytotoxicity, and cytokine production.
Collapse
Affiliation(s)
- Xiaoyuan Yang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Victor Chatterjee
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Yonggang Ma
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Ethan Zheng
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Sarah Y Yuan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
31
|
Yucesan E, Hatirnaz Ng O, Yalniz FF, Yilmaz H, Salihoglu A, Sudutan T, Eskazan AE, Ongoren S, Baslar Z, Soysal T, Ozbek U, Sayitoglu M, Ar MC. Copy-number variations in adult patients with chronic immune thrombocytopenia. Expert Rev Hematol 2020; 13:1277-1287. [PMID: 32885695 DOI: 10.1080/17474086.2020.1819786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVES Immune thrombocytopenia (ITP) is an autoimmune disease with heterogeneous background. FCGR2C mutations were defined in one third of the patients but genetic players have not been fully elucidated yet. Although childhood ITP present as benign, ITP in adulthood is chronic disease with treatment challenges. This study aimed to focus on adult ITP patients using a whole genome genotyping that is valuable approach to identify the responsible genomic regions for the disease. METHODS Herein 24 adult primary-refractory for ITP patients were evaluated using HumanCytoSNP12BeadChip,Illumina. Forty-six age and sex matched healthy individuals, and ptients awith nonhematological conditions were analyzed as controls. Identified CNV regions were verified by qRTPCR. T-cell receptor beta and delta (TCRB/TCRG) clonality were assessed by heteroduplex analysis in mosaic cases. RESULTS Several CNV losses and gains were defined (losses:2q,7q,17q,19p, and gains: 1q,2p,3q,4q,7q,10q,12p,13q,14q,15q,17p,20q,21p,22q,Xp). Mosaic changes of different sizes (0.2-17.77Mb) were identified in five patients and three of them showed clonality. CNV regions that were unique to ITP patients were identified for the first time and among these genes, those related to immune regulation, and cellular trafficking were noteworthy. Conclusion: Identified CNV regions harbor several candidate genes, the functions of which might shed light on the pathogenesis of chronic ITP.
Collapse
Affiliation(s)
- Emrah Yucesan
- Faculty of Medicine, Department of Medical Biology, Bezmialem Vakif University , Istanbul, Turkey
| | - Ozden Hatirnaz Ng
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University , Istanbul, Turkey
| | - Fevzi Firat Yalniz
- Cerrahpasa Faculty of Medicine, Division of Haematology, Department of Internal Medicine, Istanbul University-Cerrahpasa , Istanbul, Turkey
| | - Hulya Yilmaz
- Cerrahpasa Faculty of Medicine, Division of Haematology, Department of Internal Medicine, Istanbul University-Cerrahpasa , Istanbul, Turkey
| | - Ayse Salihoglu
- Cerrahpasa Faculty of Medicine, Division of Haematology, Department of Internal Medicine, Istanbul University-Cerrahpasa , Istanbul, Turkey
| | - Tugce Sudutan
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University , Istanbul, Turkey
| | - Ahmet Emre Eskazan
- Cerrahpasa Faculty of Medicine, Division of Haematology, Department of Internal Medicine, Istanbul University-Cerrahpasa , Istanbul, Turkey
| | - Seniz Ongoren
- Cerrahpasa Faculty of Medicine, Division of Haematology, Department of Internal Medicine, Istanbul University-Cerrahpasa , Istanbul, Turkey
| | - Zafer Baslar
- Cerrahpasa Faculty of Medicine, Division of Haematology, Department of Internal Medicine, Istanbul University-Cerrahpasa , Istanbul, Turkey
| | - Teoman Soysal
- Cerrahpasa Faculty of Medicine, Division of Haematology, Department of Internal Medicine, Istanbul University-Cerrahpasa , Istanbul, Turkey
| | - Ugur Ozbek
- Department of Medical Genetics, Acibadem Mehmet Ali Aydinlar University, School of Medicine , Istanbul, Turkey
| | - Muge Sayitoglu
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University , Istanbul, Turkey
| | - M Cem Ar
- Cerrahpasa Faculty of Medicine, Division of Haematology, Department of Internal Medicine, Istanbul University-Cerrahpasa , Istanbul, Turkey
| |
Collapse
|
32
|
Formation and Maturation of the Phagosome: A Key Mechanism in Innate Immunity against Intracellular Bacterial Infection. Microorganisms 2020; 8:microorganisms8091298. [PMID: 32854338 PMCID: PMC7564318 DOI: 10.3390/microorganisms8091298] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023] Open
Abstract
Phagocytosis is an essential mechanism in innate immune defense, and in maintaining homeostasis to eliminate apoptotic cells or microbes, such as Mycobacterium tuberculosis, Salmonella enterica, Streptococcus pyogenes and Legionella pneumophila. After internalizing microbial pathogens via phagocytosis, phagosomes undergo a series of ‘maturation’ steps, to form an increasingly acidified compartment and subsequently fuse with the lysosome to develop into phagolysosomes and effectively eliminate the invading pathogens. Through this mechanism, phagocytes, including macrophages, neutrophils and dendritic cells, are involved in the processing of microbial pathogens and antigen presentation to T cells to initiate adaptive immune responses. Therefore, phagocytosis plays a role in the bridge between innate and adaptive immunity. However, intracellular bacteria have evolved diverse strategies to survive and replicate within hosts. In this review, we describe the sequential stages in the phagocytosis process. We also discuss the immune evasion strategies used by pathogens to regulate phagosome maturation during intracellular bacterial infection, and indicate that these might be used for the development of potential therapeutic strategies for infectious diseases.
Collapse
|
33
|
Goldstein NB, Steel A, Barbulescu CC, Koster MI, Wright MJ, Jones KL, Gao B, Ward B, Woessner B, Trottier Z, Pakieser J, Hu J, Lambert KA, Shellman YG, Fujita M, Robinson WA, Roop DR, Norris DA, Birlea SA. Melanocyte Precursors in the Hair Follicle Bulge of Repigmented Vitiligo Skin Are Controlled by RHO-GTPase, KCTD10, and CTNNB1 Signaling. J Invest Dermatol 2020; 141:638-647.e13. [PMID: 32800877 DOI: 10.1016/j.jid.2020.07.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 06/18/2020] [Accepted: 07/06/2020] [Indexed: 01/09/2023]
Abstract
In repigmentation of human vitiligo, the melanocyte (MC) precursors in the hair follicle bulge proliferate, migrate, and differentiate to repopulate the depigmented epidermis. Here, we present a comprehensive characterization of pathways and signals in the bulge that control the repigmentation process. Using biopsies from patients with vitiligo, we have selectively harvested, by laser capture microdissection, MC and keratinocyte precursors from the hair follicle bulge of untreated vitiligo skin and vitiligo skin treated with narrow-band UVB. The captured material was subjected to whole transcriptome RNA-sequencing. With this strategy, we found that repigmentation in the bulge MC precursors is driven by KCTD10, a signal with unknown roles in the skin, and CTNNB1 (encoding β-catenin) and RHO guanosine triphosphatase [RHO GTPase, RHO], two signaling pathways previously shown to be involved in pigmentation biology. Knockdown studies in cultured human MCs of RHOJ, the upmost differentially expressed RHO family component, corroborated with our findings in patients with vitiligo, identified RHOJ involvement in UV response and melanization, and confirmed previously identified roles in melanocytic cell migration and apoptosis. A better understanding of mechanisms that govern repigmentation in MC precursors will enable the discovery of molecules that induce robust repigmentation phenotypes in vitiligo.
Collapse
Affiliation(s)
| | - Andrea Steel
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA
| | | | - Maranke I Koster
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA
| | - Michael J Wright
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA
| | - Kenneth L Jones
- Department of Hematology, University of Colorado, Aurora, Colorado, USA; Department of Pediatrics, University of Colorado, Aurora, Colorado, USA
| | - Bifeng Gao
- Sequencing and Microarray Core, University of Colorado, Aurora, Colorado, USA
| | - Brian Ward
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA
| | - Brian Woessner
- Sequencing and Microarray Core, University of Colorado, Aurora, Colorado, USA
| | - Zachary Trottier
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA
| | - Jen Pakieser
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA
| | - Junxiao Hu
- Department of Pediatrics, University of Colorado, Aurora, Colorado, USA; Cancer Center Biostatistics Core, University of Colorado, Aurora, Colorado, USA
| | - Karoline A Lambert
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA
| | - Yiqun G Shellman
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA
| | - Mayumi Fujita
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA; Denver Department of Veterans Affairs Medical Center, Denver, Colorado, USA
| | | | - Dennis R Roop
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA
| | - David A Norris
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA; Denver Department of Veterans Affairs Medical Center, Denver, Colorado, USA
| | - Stanca A Birlea
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA; Human Medical Genetics and Genomics Program, Aurora, Colorado, USA.
| |
Collapse
|
34
|
Saleh R, Taha RZ, Sasidharan Nair V, Toor SM, Alajez NM, Elkord E. Transcriptomic Profiling of Circulating HLA-DR - Myeloid Cells, Compared with HLA-DR + Myeloid Antigen-presenting Cells. Immunol Invest 2020; 50:952-963. [PMID: 32727251 DOI: 10.1080/08820139.2020.1795875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells with potent immunosuppressive functions, which can inhibit the activation of immune responses under a steady-state condition and pathological conditions. We performed transcriptomic profiling of circulating CD33+HLA-DR+ myeloid antigen-presenting cells (APCs) and CD33+HLA-DR- myeloid cells (potentially MDSCs) in healthy individuals. We sorted both subpopulations from peripheral blood mononuclear cells (PBMCs) of 10 healthy donors and performed RNA sequencing (RNA-Seq). We found that several signaling pathways associated with the positive regulation of immune responses, such as antigen presentation/processing, FcγR-mediated phagocytosis and immune cell trafficking, phosphoinositide 3-kinase (PI3K)/Akt signaling, DC maturation, triggering receptor expressed on myeloid cells 1 (TREM1) signaling, nuclear factor of activated T cells (NFAT) and IL-8 signaling were downregulated in CD33+HLA-DR- myeloid cells. In contrast, pathways implicated in tumor suppression and anti-inflammation, including peroxisome proliferator-activated receptor (PPAR) and phosphatase and tensin homolog (PTEN), were upregulated in CD33+HLA-DR- myeloid cells. These data indicate that PPAR/PTEN axis could be upregulated in myeloid cells to keep the immune system in check in normal physiological conditions. Our data reveal some of the molecular and functional differences between CD33+HLA-DR+ APCs and CD33+HLA-DR- myeloid cells in a steady-state condition, reflecting the potential suppressive function of CD33+HLA-DR- myeloid cells to maintain immune tolerance. For future studies, the same methodological approach could be applied to perform transcriptomic profiling of myeloid subsets in pathological conditions.
Collapse
Affiliation(s)
- Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Rowaida Z Taha
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Varun Sasidharan Nair
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Salman M Toor
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Nehad M Alajez
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| |
Collapse
|
35
|
Junker F, Gordon J, Qureshi O. Fc Gamma Receptors and Their Role in Antigen Uptake, Presentation, and T Cell Activation. Front Immunol 2020; 11:1393. [PMID: 32719679 PMCID: PMC7350606 DOI: 10.3389/fimmu.2020.01393] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/01/2020] [Indexed: 12/24/2022] Open
Abstract
The cellular uptake, intracellular processing, and presentation of foreign antigen are crucial processes for eliciting an effective adaptive host response to the majority of pathogens. The effective recognition of antigen by T cells requires that it is first processed and then presented on MHC molecules that are expressed on other cells. A critical step leading to the presentation of antigen is delivering the foreign cargo to an intracellular compartment where the antigen can be processed and loaded onto MHC molecules. Fc-gamma receptors (FcγRs) recognize IgG-coated targets, such as opsonized pathogens or immune complexes (ICs). Cross-linking leads to internalization of the cargo with associated activation of down-stream signaling cascades. FcγRs vary in their affinity for IgG and intracellular trafficking, and therefore have an opportunity to regulate antigen presentation by controlling the shuttling and processing of their cargos. In this way, they critically influence physiological and pathophysiological adaptive immune cell functions. In this review, we will cover the contribution of FcγRs to antigen-presentation with a focus on the intracellular trafficking of IgG-ICs and the pathways that support this function. We will also discuss genetic evidence linking FcγR biology to immune cell activation and autoimmune processes as exemplified by systemic lupus erythematosus (SLE).
Collapse
Affiliation(s)
- Fabian Junker
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - John Gordon
- Celentyx Ltd, Birmingham Research Park, Birmingham, United Kingdom
| | - Omar Qureshi
- Celentyx Ltd, Birmingham Research Park, Birmingham, United Kingdom
| |
Collapse
|
36
|
Peshdary V, Styles G, Gagné R, Yauk CL, Sorisky A, Atlas E. Depot-Specific Analysis of Human Adipose Cells and Their Responses to Bisphenol S. Endocrinology 2020; 161:5805237. [PMID: 32170302 DOI: 10.1210/endocr/bqaa044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/16/2020] [Indexed: 12/15/2022]
Abstract
Exposure to endocrine-disrupting chemicals (EDCs) is associated with adverse health outcomes including obesity and diabetes. Obesity, and more specifically visceral obesity, is correlated with metabolic disease. The adipose tissue is an endocrine organ and a potential target for many environmental pollutants including bisphenols. The subcutaneous (Sc) and the omental (Om, visceral) depots are composed of mature adipocytes and residing progenitors, which may be different between the depots and may be EDCs targets. Bisphenol A (BPA) is a suspected metabolic disruptor, and is being replaced with structurally similar compounds such as bisphenol S (BPS). Like BPA, BPS induces adipogenesis in murine and primary human Sc preadipocytes. However, the effect of BPS on Om preadipocytes is not known. In this study, we show that human primary progenitors from Om depots have a distinct transcriptomic signature as compared to progenitors derived from donor-matched Sc depots. Furthermore, we show that BPS increases adipogenesis both of Om and Sc preadipocytes and can mimic the action of glucocorticoids or peroxisome proliferator-activated receptor γ (PPARγ) agonists. We also show that BPS treatment, at 0.1 µM and 25 µM, modifies the adipokine profiles both of Om- and Sc-derived adipocytes in a depot-specific manner. Taken together our data show distinct gene expression profiles in the Om vs Sc progenitors and similar responses to the BPA analogue, BPS.
Collapse
Affiliation(s)
- Vian Peshdary
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
- Departments of Medicine and of Biochemistry, University of Ottawa, Ottawa, Canada
| | - George Styles
- Departments of Medicine and of Biochemistry, University of Ottawa, Ottawa, Canada
| | - Rémi Gagné
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Carole L Yauk
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Alexander Sorisky
- Departments of Medicine and of Biochemistry, University of Ottawa, Ottawa, Canada
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Ella Atlas
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
- Departments of Medicine and of Biochemistry, University of Ottawa, Ottawa, Canada
| |
Collapse
|
37
|
Walpole GFW, Grinstein S. Endocytosis and the internalization of pathogenic organisms: focus on phosphoinositides. F1000Res 2020; 9. [PMID: 32494357 PMCID: PMC7233180 DOI: 10.12688/f1000research.22393.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/07/2020] [Indexed: 12/18/2022] Open
Abstract
Despite their comparatively low abundance in biological membranes, phosphoinositides are key to the regulation of a diverse array of signaling pathways and direct membrane traffic. The role of phosphoinositides in the initiation and progression of endocytic pathways has been studied in considerable depth. Recent advances have revealed that distinct phosphoinositide species feature prominently in clathrin-dependent and -independent endocytosis as well as in phagocytosis and macropinocytosis. Moreover, a variety of intracellular and cell-associated pathogens have developed strategies to commandeer host cell phosphoinositide metabolism to gain entry and/or metabolic advantage, thereby promoting their survival and proliferation. Here, we briefly survey the current knowledge on the involvement of phosphoinositides in endocytosis, phagocytosis, and macropinocytosis and highlight several examples of molecular mimicry employed by pathogens to either “hitch a ride” on endocytic pathways endogenous to the host or create an entry path of their own.
Collapse
Affiliation(s)
- Glenn F W Walpole
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Sergio Grinstein
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
| |
Collapse
|
38
|
Torres-Gomez A, Cabañas C, Lafuente EM. Phagocytic Integrins: Activation and Signaling. Front Immunol 2020; 11:738. [PMID: 32425937 PMCID: PMC7203660 DOI: 10.3389/fimmu.2020.00738] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/31/2020] [Indexed: 01/06/2023] Open
Abstract
Phagocytic integrins are endowed with the ability to engulf and dispose of particles of different natures. Evolutionarily conserved from worms to humans, they are involved in pathogen elimination and apoptotic and tumoral cell clearance. Research in the field of integrin-mediated phagocytosis has shed light on the molecular events controlling integrin activation and their effector functions. However, there are still some aspects of the regulation of the phagocytic process that need to be clarified. Here, we have revised the molecular events controlling phagocytic integrin activation and the downstream signaling driving particle engulfment, and we have focused particularly on αMβ2/CR3, αXβ2/CR4, and a brief mention of αVβ5/αVβ3integrins.
Collapse
Affiliation(s)
- Alvaro Torres-Gomez
- Department of Immunology, Ophthalmology and Otorhinolaryngology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Carlos Cabañas
- Department of Immunology, Ophthalmology and Otorhinolaryngology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), Madrid, Spain.,Severo Ochoa Center for Molecular Biology (CSIC-UAM), Madrid, Spain
| | - Esther M Lafuente
- Department of Immunology, Ophthalmology and Otorhinolaryngology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), Madrid, Spain
| |
Collapse
|
39
|
Heimes A, Brodhagen J, Weikard R, Seyfert HM, Becker D, Meyerholz MM, Petzl W, Zerbe H, Hoedemaker M, Rohmeier L, Schuberth HJ, Schmicke M, Engelmann S, Kühn C. Hepatic Transcriptome Analysis Identifies Divergent Pathogen-Specific Targeting-Strategies to Modulate the Innate Immune System in Response to Intramammary Infection. Front Immunol 2020; 11:715. [PMID: 32411137 PMCID: PMC7202451 DOI: 10.3389/fimmu.2020.00715] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 03/30/2020] [Indexed: 12/22/2022] Open
Abstract
Mastitis is one of the major risks for public health and animal welfare in the dairy industry. Two of the most important pathogens to cause mastitis in dairy cattle are Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli). While S. aureus generally induces a chronic and subclinical mastitis, E. coli is an important etiological pathogen resulting in an acute and clinical mastitis. The liver plays a central role in both, the metabolic and inflammatory physiology of the dairy cow, which is particularly challenged in the early lactation due to high metabolic and immunological demands. In the current study, we challenged the mammary glands of Holstein cows with S. aureus or E. coli, respectively, mimicking an early lactation infection. We compared the animals' liver transcriptomes with those of untreated controls to investigate the hepatic response of the individuals. Both, S. aureus and E. coli elicited systemic effects on the host after intramammary challenge and seemed to use pathogen-specific targeting strategies to bypass the innate immune system. The most striking result of our study is that we demonstrate for the first time that S. aureus intramammary challenge causes an immune response beyond the original local site of the mastitis. We found that in the peripheral liver tissue defined biological pathways are switched on in a coordinated manner to balance the immune response in the entire organism. TGFB1 signaling plays a crucial role in this context. Important pathways involving actin and integrin, key components of the cytoskeleton, were downregulated in the liver of S. aureus infected cows. In the hepatic transcriptome of E. coli infected cows, important components of the complement system were significantly lower expressed compared to the control cows. Notably, while S. aureus inhibits the cell signaling by Rho GTPases in the liver, E. coli switches the complement system off. Also, metabolic hepatic pathways (e.g., lipid metabolism) are affected after mammary gland challenge, demonstrating that the liver restricts metabolic tasks in favor of the predominant immune response after infection. Our results provide new insights for the infection-induced modifications of the dairy cow's hepatic transcriptome following mastitis.
Collapse
Affiliation(s)
- Annika Heimes
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Dummerstorf, Germany
| | - Johanna Brodhagen
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Dummerstorf, Germany
| | - Rosemarie Weikard
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Dummerstorf, Germany
| | - Hans-Martin Seyfert
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Dummerstorf, Germany
| | - Doreen Becker
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Dummerstorf, Germany
| | - Marie M Meyerholz
- Clinic for Ruminants with Ambulatory and Herd Health Services, Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-University Munich, Oberschleißheim, Germany
| | - Wolfram Petzl
- Clinic for Ruminants with Ambulatory and Herd Health Services, Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-University Munich, Oberschleißheim, Germany
| | - Holm Zerbe
- Clinic for Ruminants with Ambulatory and Herd Health Services, Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-University Munich, Oberschleißheim, Germany
| | - Martina Hoedemaker
- Clinic for Cattle, University of Veterinary Medicine Hanover, Hanover, Germany
| | - Laura Rohmeier
- Clinic for Ruminants with Ambulatory and Herd Health Services, Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-University Munich, Oberschleißheim, Germany.,Clinic for Swine, Small Ruminants, Forensic Medicine and Ambulatory Service, University of Veterinary Medicine Hanover, Hanover, Germany
| | | | - Marion Schmicke
- Faculty of Natural Sciences III, Martin-Luther Universität Halle-Wittenberg, Halle, Germany
| | - Susanne Engelmann
- Technical University Braunschweig, Institute for Microbiology, Brunswick, Germany.,Helmholtz Centre for Infection Research, Microbial Proteomics, Brunswick, Germany
| | - Christa Kühn
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Dummerstorf, Germany.,Agricultural and Environmental Faculty, University Rostock, Rostock, Germany
| |
Collapse
|
40
|
Abstract
Phagocytosis is a pivotal immunological process, and its discovery by Elia Metchnikoff in 1882 was a step toward the establishment of the innate immune system as a separate branch of immunology. Elia Metchnikoff received the Nobel Prize in physiology and medicine for this discovery in 1908. Since its discovery almost 140 years before, phagocytosis remains the hot topic of research in immunology. The phagocytosis research has seen a great advancement since its first discovery. Functionally, phagocytosis is a simple immunological process required to engulf and remove pathogens, dead cells and tumor cells to maintain the immune homeostasis. However, mechanistically, it is a very complex process involving different mechanisms, induced and regulated by several pattern recognition receptors, soluble pattern recognition molecules, scavenger receptors (SRs) and opsonins. These mechanisms involve the formation of phagosomes, their maturation into phagolysosomes causing pathogen destruction or antigen synthesis to present them to major histocompatibility complex molecules for activating an adaptive immune response. Any defect in this mechanism may predispose the host to certain infections and inflammatory diseases (autoinflammatory and autoimmune diseases) along with immunodeficiency. The article is designed to discuss its mechanistic complexity at each level, varying from phagocytosis induction to phagolysosome resolution.
Collapse
Affiliation(s)
- Vijay Kumar
- Faculty of Medicine, Children's Health Queensland Clinical Unit, School of Clinical Medicine, Mater Research, University of Queensland, ST Lucia, Brisbane, Queensland, Australia.,Faculty of Medicine, School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
41
|
Mohammed A, Okwor I, Shan L, Onyilagha C, Uzonna JE, Gounni AS. Semaphorin 3E Regulates the Response of Macrophages to Lipopolysaccharide-Induced Systemic Inflammation. THE JOURNAL OF IMMUNOLOGY 2019; 204:128-136. [PMID: 31776203 DOI: 10.4049/jimmunol.1801514] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 10/28/2019] [Indexed: 12/19/2022]
Abstract
Semaphorin 3E (Sema3E) is a secreted protein that was initially discovered as a neuronal guidance cue. Recent evidence showed that Sema3E plays an essential role in regulating the activities of various immune cells. However, the exact role of Sema3E in macrophage function, particularly during inflammation, is not fully understood. We studied the impact of Sema3E gene deletion on macrophage function during the LPS-induced acute inflammatory response. We found that Sema3E-deficient (Sema3e-/- ) mice were better protected from LPS-induced acute inflammation as exemplified by their superior clinical score and effective temperature control compared with their wild-type littermates. This superior control of inflammatory response in Sema3e-/- mice was associated with significantly lower phosphorylation of ERK1/2, AKT, STAT3, and NF-κB, and a concomitant reduction in inducible NO synthase expression and production of TNF and IL-6 compared with their Sema3e+/+ littermates. Sema3e-/- mice also contained significantly higher numbers of activated macrophages compared with their Sema3e+/+ littermates at both baselines and after LPS challenge. In vivo-specific deletion of the Sema3E high-affinity receptor, plexinD1, on macrophages led to the improvement in clinical disease following exposure to a lethal dose of LPS. Collectively, our data show that Sema3E plays an essential role in dampening the early inflammatory response to LPS by regulating macrophage function, suggesting an essential role of this pathway in macrophage inflammatory response.
Collapse
Affiliation(s)
- Ashfaque Mohammed
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada
| | - Ifeoma Okwor
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada
| | - Lianyu Shan
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada
| | - Chukwunonso Onyilagha
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada
| | - Jude E Uzonna
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada
| | - Abdelilah S Gounni
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada
| |
Collapse
|
42
|
Szilveszter KP, Németh T, Mócsai A. Tyrosine Kinases in Autoimmune and Inflammatory Skin Diseases. Front Immunol 2019; 10:1862. [PMID: 31447854 PMCID: PMC6697022 DOI: 10.3389/fimmu.2019.01862] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/23/2019] [Indexed: 12/30/2022] Open
Abstract
Tyrosine kinases relay signals from diverse leukocyte antigen receptors, innate immune receptors, and cytokine receptors, and therefore mediate the recruitment and activation of various leukocyte populations. Non-receptor tyrosine kinases of the Jak, Src, Syk, and Btk families play major roles in various immune-mediated disorders, and small-molecule tyrosine kinase inhibitors are emerging novel therapeutics in a number of those diseases. Autoimmune and inflammatory skin diseases represent a broad spectrum of immune-mediated diseases. Genetic and pharmacological studies in humans and mice support the role of tyrosine kinases in several inflammatory skin diseases. Atopic dermatitis and psoriasis are characterized by an inflammatory microenvironment which activates cytokine receptors coupled to the Jak-Stat signaling pathway. Jak kinases are also implicated in alopecia areata and vitiligo, skin disorders mediated by cytotoxic T lymphocytes. Genetic studies indicate a critical role for Src-family kinases and Syk in animal models of autoantibody-mediated blistering skin diseases. Here, we review the various tyrosine kinase signaling pathways and their role in various autoimmune and inflammatory skin diseases. Special emphasis will be placed on identification of potential therapeutic targets, as well as on ongoing preclinical and clinical studies for the treatment of inflammatory skin diseases by small-molecule tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Kata P Szilveszter
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
| | - Tamás Németh
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
| | - Attila Mócsai
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
| |
Collapse
|
43
|
Naert R, López-Requena A, Voets T, Talavera K, Alpizar YA. Expression and Functional Role of TRPV4 in Bone Marrow-Derived CD11c + Cells. Int J Mol Sci 2019; 20:ijms20143378. [PMID: 31295806 PMCID: PMC6678969 DOI: 10.3390/ijms20143378] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/07/2019] [Accepted: 07/08/2019] [Indexed: 01/04/2023] Open
Abstract
The increase in cytosolic Ca2+ is essential in key effector functions of dendritic cells (DCs), including differentiation, maturation, cytokine expression, and phagocytosis. Although several Ca2+-permeable ion channels have been described in DCs, the contribution of transient receptor potential (TRP) channels remains poorly understood. Here, we investigated whether TRPV4 plays a role in the differentiation, maturation, and phagocytosis of granulocyte-macrophage colony-stimulating factor (GM-CSF)-induced mouse bone marrow-derived cells (BMDCs). Using intracellular Ca2+ imaging experiments, we found that TRPV4 was functionally expressed in the plasma membrane of immature CD11c+ BMDCs and that its activity and expression were downregulated in CD11c+ BMDCs matured with lipopolysaccharide (LPS). Comparative analysis of the GM-CSF-stimulated cells showed that Trpv4 knockout and wild-type bone marrow cultures had a similar distribution of differentiated cells, generating a heterogenous culture population rich in CD11c+, CD11b+ cells, and low levels of F4/80+ cells. The lack of TRPV4 did not prevent the LPS-induced nuclear translocation of NF-κB, the upregulation of the proinflammatory cytokines IL-6 and IL-12, or the upregulation of the maturation markers CD40, CD80, and CD86. In contrast, TRPV4-deficient CD11c+ BMDCs exhibited a significantly reduced endocytic capacity of IgG-coated beads, but the internalization of uncoated beads in the absence of TRPV4 was not affected. Taken together, our results demonstrate that TRPV4 was dispensable in the differentiation and maturation of mouse CD11c+ BMDCs but contributed to the mechanism underlying Fc receptor-mediated phagocytosis. Overall, our results further strengthen the role of TRPV4 in immune-related processes.
Collapse
Affiliation(s)
- Robbe Naert
- Laboratory of Ion Channel Research, Dept. of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Alejandro López-Requena
- Laboratory of Ion Channel Research, Dept. of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research, Dept. of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Karel Talavera
- Laboratory of Ion Channel Research, Dept. of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Yeranddy A Alpizar
- Laboratory of Ion Channel Research, Dept. of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium.
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium.
| |
Collapse
|
44
|
Ivanova II, Mihaylova NM, Manoylov IK, Makatsori D, Lolov S, Nikolova MH, Mamalaki A, Prechl J, Tchorbanov AI. Targeting of Influenza Viral Epitopes to Antigen-Presenting Cells by Genetically Engineered Chimeric Molecules in a Humanized NOD SCID Gamma Transfer Model. Hum Gene Ther 2019; 29:1056-1070. [PMID: 30191743 DOI: 10.1089/hum.2018.100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Antiviral DNA vaccines are a novel strategy in the vaccine development field, which basically consists of the administration of expression vectors coding viral antigen sequences into the host's cells. Targeting of conserved viral epitopes by antibody fragments specific to activating cell surface co-receptor molecules on antigen-presenting cells could be an alternative approach for inducing protective immunity. It has been shown that FcγRI on human monocytes enhances antigen presentation in vivo. Various DNA constructs, encoding a Single-chain variable antibodies (scFv) from mouse anti-human FcγRI monoclonal antibody, coupled to a sequence encoding a T- and B-cell epitope-containing influenza A virus hemagglutinin inter-subunit peptide were inserted into the eukaryotic expression vector system pTriEx-3 Neo. The constructed chimeric DNA molecules were expressed by transfected Chinese hamster ovary cells and the ability of the engineered proteins to interact with FcγRI-expressing cells was confirmed by flow cytometry. The fusion protein induced a strong signal transduction on human monocytes via FcγRI. The expression vector pTriEx-3 Neo containing the described construct was used as a naked DNA vaccine and introduced directly to experimental humanized NOD SCID gamma mice with or without boosting with the expressed fusion protein. Immunization with the generated DNA chimeric molecules and prime-boost with the expressed recombinant proteins induced significant serum levels of anti-influenza immunoglobulin G antibodies and strong cytotoxic T lymphocyte activity against influenza virus-infected cells in humanized animals.
Collapse
Affiliation(s)
- Iva I Ivanova
- 1 Laboratory of Experimental Immunology, Institute of Microbiology , Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Nikolina M Mihaylova
- 1 Laboratory of Experimental Immunology, Institute of Microbiology , Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Iliyan K Manoylov
- 1 Laboratory of Experimental Immunology, Institute of Microbiology , Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | - Stefan Lolov
- 3 Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Maria H Nikolova
- 4 National Reference Laboratory of Immunology, National Center of Infectious and Parasitic Diseases , Sofia, Bulgaria
| | - Avgi Mamalaki
- 2 Hellenic Pasteur Institute , Ampelokipi, Athens, Greece
| | - Jozsef Prechl
- 5 Immunology Research Group, Hungarian Academy of Sciences , Budapest, Hungary
| | - Andrey I Tchorbanov
- 1 Laboratory of Experimental Immunology, Institute of Microbiology , Bulgarian Academy of Sciences, Sofia, Bulgaria .,6 National Institute of Immunology , Sofia, Bulgaria
| |
Collapse
|
45
|
Naik U, Nguyen QPH, Harrison RE. Binding and uptake of single and dual-opsonized targets by macrophages. J Cell Biochem 2019; 121:183-199. [PMID: 31172552 DOI: 10.1002/jcb.29043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 05/03/2019] [Indexed: 12/15/2022]
Abstract
Our current understanding of phagocytosis is largely derived from studies of individual receptor-ligand interactions and their downstream signaling pathways. Because phagocytes are exposed to a variety of ligands on heterogeneous target particles in vivo, it is important to observe the engagement of multiple receptors simultaneously and the triggered involvement of downstream signaling pathways. Potential crosstalk between the two well-characterized opsonic receptors, FcγR and CR3, was briefly explored in the early 1970s, where macrophages were challenged with dual-opsonized targets. However, subsequent studies on receptor crosstalk were primarily restricted to using single opsonins on different targets, typically at saturating opsonin conditions. Beyond validating these initial explorations on receptor crosstalk, we identify the early signaling mechanisms that underlie the binding and phagocytosis during the simultaneous activation of both opsonic receptors, through the presence of a dual-opsonized target (immunoglobulin G [IgG] and C3bi), compared with single receptor activation. For this purpose, we used signaling protein inhibitor studies as well as live cell brightfield and fluorescent imaging to fully understand the role of tyrosine kinases, F-actin dynamics and internalization kinetics for FcγR and CR3. Importantly, opsonic receptors were studied together and in isolation, in the context of sparsely opsonized targets. We observed enhanced particle binding and a synergistic effect on particle internalization during the simultaneous activation of FcγR and CR3 engaged with sparsely opsonized targets. Inhibition of early signaling and cytoskeletal molecules revealed a differential involvement of Src kinase for FcγR- vs CR3- and dual receptor-mediated phagocytosis. Src activity recruits Syk kinase and we observed intermediate levels of Syk phosphorylation in dual-opsonized particles compared with those opsonized with IgG or C3bi alone. These results likely explain the intermediate levels of F-actin that is recruited to sites of dual-opsonized particle uptake and the notoriously delayed internalization of C3bi-opsonized targets by macrophages.
Collapse
Affiliation(s)
- Urja Naik
- Department of Cell & Systems Biology, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Quynh Phuong Hai Nguyen
- Department of Cell & Systems Biology, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Rene E Harrison
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| |
Collapse
|
46
|
Pellizzari G, Hoskin C, Crescioli S, Mele S, Gotovina J, Chiaruttini G, Bianchini R, Ilieva K, Bax HJ, Papa S, Lacy KE, Jensen-Jarolim E, Tsoka S, Josephs DH, Spicer JF, Karagiannis SN. IgE re-programs alternatively-activated human macrophages towards pro-inflammatory anti-tumoural states. EBioMedicine 2019; 43:67-81. [PMID: 30956175 PMCID: PMC6562024 DOI: 10.1016/j.ebiom.2019.03.080] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Antibody Fc-driven engagement of macrophages is critical for evoking cellular activation and effector functions and influencing tumour-associated macrophage (TAM) recruitment. We previously reported that IgE class antibodies promote restriction of cancer growth in rodent models associated with significant TAM infiltration. However, the human macrophage-associated IgE-Fc Receptor (FcεR) axis remains unexplored. We investigated the effects of anti-tumour IgE stimulation on human macrophage activation. METHODS Human blood monocyte-differentiated quiescent (M0), classically-(M1) and alternatively-(M2) activated macrophages were crosslinked with IgE and polyclonal antibodies to mimic immune complex formation. We examined surface marker expression, cytokine secretion, protein kinase phosphorylation and gene expression in IgE-stimulated macrophages and IgE antibody-dependent macrophage-mediated cytotoxicity (ADCC) against tumour cells. FINDINGS A proportion (40%) of M2 and (<20%) M0 and M1 macrophages expressed the high-affinity IgE receptor FcεRI. IgE crosslinking triggered upregulation of co-stimulatory CD80, increased TNFα, IFNγ, IL-1β, IL-12, IL-10, IL-13, CXCL9, CXCL11 and RANTES secretion by M0 and M2 and additionally enhanced MCP-1 by M2 macrophages. IgE-stimulated M1 macrophages retained secretion of pro-inflammatory cytokines. IgE crosslinking enhanced the FcεRI-dependent signalling pathway, including phosphorylation of the Lyn kinase, ERK1/2 and p38 in M2 macrophages and upregulated Lyn gene expression by M1 and M2 macrophages. Anti-tumour IgE engendered ADCC of cancer cells by all macrophage subsets. INTERPRETATION IgE can engage and re-educate alternatively-activated macrophages towards pro-inflammatory phenotypes and prime all subsets to mediate anti-tumour functions. This points to IgE-mediated cascades with potential to activate immune stroma and may be significant in the clinical development of strategies targeting tumour-resident macrophages.
Collapse
Affiliation(s)
- Giulia Pellizzari
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Coran Hoskin
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, Bush House, London WC2B 4BG, United Kingdom
| | - Silvia Crescioli
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Silvia Mele
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Jelena Gotovina
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University Vienna, Austria; Department of Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Giulia Chiaruttini
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Rodolfo Bianchini
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University Vienna, Austria; Department of Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Kristina Ilieva
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom; Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| | - Heather J Bax
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom; School of Cancer & Pharmaceutical Sciences, King's College London, Bermondsey Wing, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Sophie Papa
- School of Cancer & Pharmaceutical Sciences, King's College London, Bermondsey Wing, Guy's Hospital, London SE1 9RT, United Kingdom; Guy's and St Thomas' NHS Trust, Department of Medical Oncology, London, United Kingdom
| | - Katie E Lacy
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Erika Jensen-Jarolim
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University Vienna, Austria; Department of Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Sophia Tsoka
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, Bush House, London WC2B 4BG, United Kingdom
| | - Debra H Josephs
- School of Cancer & Pharmaceutical Sciences, King's College London, Bermondsey Wing, Guy's Hospital, London SE1 9RT, United Kingdom; Guy's and St Thomas' NHS Trust, Department of Medical Oncology, London, United Kingdom
| | - James F Spicer
- School of Cancer & Pharmaceutical Sciences, King's College London, Bermondsey Wing, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom.
| |
Collapse
|
47
|
Size-Dependent Segregation Controls Macrophage Phagocytosis of Antibody-Opsonized Targets. Cell 2019; 174:131-142.e13. [PMID: 29958103 DOI: 10.1016/j.cell.2018.05.059] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/20/2018] [Accepted: 05/29/2018] [Indexed: 12/18/2022]
Abstract
Macrophages protect the body from damage and disease by targeting antibody-opsonized cells for phagocytosis. Though antibodies can be raised against antigens with diverse structures, shapes, and sizes, it is unclear why some are more effective at triggering immune responses than others. Here, we define an antigen height threshold that regulates phagocytosis of both engineered and cancer-specific antigens by macrophages. Using a reconstituted model of antibody-opsonized target cells, we find that phagocytosis is dramatically impaired for antigens that position antibodies >10 nm from the target surface. Decreasing antigen height drives segregation of antibody-bound Fc receptors from the inhibitory phosphatase CD45 in an integrin-independent manner, triggering Fc receptor phosphorylation and promoting phagocytosis. Our work shows that close contact between macrophage and target is a requirement for efficient phagocytosis, suggesting that therapeutic antibodies should target short antigens in order to trigger Fc receptor activation through size-dependent physical segregation.
Collapse
|
48
|
Barger SR, Reilly NS, Shutova MS, Li Q, Maiuri P, Heddleston JM, Mooseker MS, Flavell RA, Svitkina T, Oakes PW, Krendel M, Gauthier NC. Membrane-cytoskeletal crosstalk mediated by myosin-I regulates adhesion turnover during phagocytosis. Nat Commun 2019; 10:1249. [PMID: 30890704 PMCID: PMC6425032 DOI: 10.1038/s41467-019-09104-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 02/21/2019] [Indexed: 11/09/2022] Open
Abstract
Phagocytosis of invading pathogens or cellular debris requires a dramatic change in cell shape driven by actin polymerization. For antibody-covered targets, phagocytosis is thought to proceed through the sequential engagement of Fc-receptors on the phagocyte with antibodies on the target surface, leading to the extension and closure of the phagocytic cup around the target. We find that two actin-dependent molecular motors, class 1 myosins myosin 1e and myosin 1f, are specifically localized to Fc-receptor adhesions and required for efficient phagocytosis of antibody-opsonized targets. Using primary macrophages lacking both myosin 1e and myosin 1f, we find that without the actin-membrane linkage mediated by these myosins, the organization of individual adhesions is compromised, leading to excessive actin polymerization, slower adhesion turnover, and deficient phagocytic internalization. This work identifies a role for class 1 myosins in coordinated adhesion turnover during phagocytosis and supports a mechanism involving membrane-cytoskeletal crosstalk for phagocytic cup closure.
Collapse
Affiliation(s)
- Sarah R Barger
- Cell and Developmental Biology Department, State University of New York Upstate Medical University, Syracuse, 13210, NY, USA
| | - Nicholas S Reilly
- Department of Physics, University of Rochester, Rochester, 14627, NY, USA
| | - Maria S Shutova
- Department of Biology, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Qingsen Li
- IFOM, FIRC Institute of Molecular Oncology, Milan, 20139, Italy
| | - Paolo Maiuri
- IFOM, FIRC Institute of Molecular Oncology, Milan, 20139, Italy
| | - John M Heddleston
- Advanced Imaging Center, Howard Hughes Medical Institute Janelia Research Campus, Ashburn, 20147, VA, USA
| | - Mark S Mooseker
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, 06520, CT, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, 06519, CT, USA
- Howard Hughes Medical Institute, Yale University, New Haven, 06519, CT, USA
| | - Tatyana Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Patrick W Oakes
- Department of Physics, University of Rochester, Rochester, 14627, NY, USA
- Department of Biology, University of Rochester, Rochester, 14627, NY, USA
| | - Mira Krendel
- Cell and Developmental Biology Department, State University of New York Upstate Medical University, Syracuse, 13210, NY, USA.
| | - Nils C Gauthier
- IFOM, FIRC Institute of Molecular Oncology, Milan, 20139, Italy.
| |
Collapse
|
49
|
Li S, Yang J, Wang L, Du F, Zhao J, Fang R. Expression profile of microRNAs in porcine alveolar macrophages after Toxoplasma gondii infection. Parasit Vectors 2019; 12:65. [PMID: 30696482 PMCID: PMC6350279 DOI: 10.1186/s13071-019-3297-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/07/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Toxoplasma gondii is an apicomplexan protozoan parasite that can cause serious clinical illnesses in both humans and animals. microRNAs (miRNAs) are non-protein-coding RNAs that can regulate the expression of target genes. A previous study found that many miRNAs were differentially expressed after T. gondii infection and exert significant effects and revealed that both host survival and the virulence of different strains can be regulated by different miRNAs. Macrophages play an important role in T. gondii infection, but few studies have investigated the relationship between miRNAs and porcine alveolar macrophages infected with T. gondii. METHODS Porcine alveolar macrophages (3D4-21) were infected with the RH (Type I) and Me49 (Type II) strains of T. gondii for 12 h and 24 h and then harvested. miRNA libraries were generated using the NEBNext® Multiplex Small RNA Library Prep Set for Illumina® (NEB, USA), and the miRNA expression levels were estimated based on transcripts per million reads (TPM). RESULTS Our study generated six miRNA expression profiles from macrophages infected with RH and Me49 compared with the control groups. The comparison of the T. gondii-infected and uninfected samples identified 81 differentially expressed miRNAs, including 36 novel miRNAs and 45 mature miRNAs. The target genes of these differentially expressed miRNAs were predicted using miRanda software, and ssc-miR-127 and ssc-miR-143-3p were predicted to regulate nitric oxide synthase 1 (NOS1) and nitric oxide synthase 3 (NOS3), respectively, which play essential roles in synthesizing nitric oxide (NO) by oxidizing L-arginine. These genes were differentially expressed in both the RH- and Me49-infected groups. A KEGG enrichment analysis indicated that the predicted target genes were involved in multiple signaling pathways, including FcγR-mediated phagocytosis, the AMPK signaling pathway, the mTOR signaling pathway, and the FcγRI signaling pathway, all of which are indispensable for the normal functioning of porcine alveolar macrophages. CONCLUSIONS Our results provide data on the miRNA profile of porcine alveolar macrophages infected with T. gondii. To our knowledge, this study provides the first demonstration of the relationship between miRNA and macrophages of swine origin. Understanding the functions of these regulated miRNAs will aid the investigation of T. gondii infectious diseases, and the differentially expressed miRNAs might be candidate drug targets for T. gondii infection in pigs.
Collapse
Affiliation(s)
- Senyang Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1, Shizishan Street, Wuhan, Hubei Province, 430070, People's Republic of China
| | - Jing Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1, Shizishan Street, Wuhan, Hubei Province, 430070, People's Republic of China
| | - Luyao Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1, Shizishan Street, Wuhan, Hubei Province, 430070, People's Republic of China
| | - Fen Du
- Hubei Center for Animal Diseases Control and Prevention, Wuhan, Hubei Province, 430070, People's Republic of China
| | - Junlong Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1, Shizishan Street, Wuhan, Hubei Province, 430070, People's Republic of China.
| | - Rui Fang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1, Shizishan Street, Wuhan, Hubei Province, 430070, People's Republic of China.
| |
Collapse
|
50
|
Clarke F, Purvis HA, Sanchez-Blanco C, Gutiérrez-Martinez E, Cornish GH, Zamoyska R, Guermonprez P, Cope AP. The protein tyrosine phosphatase PTPN22 negatively regulates presentation of immune complex derived antigens. Sci Rep 2018; 8:12692. [PMID: 30139951 PMCID: PMC6107551 DOI: 10.1038/s41598-018-31179-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/06/2018] [Indexed: 12/11/2022] Open
Abstract
A C1858T single nucleotide polymorphism within PTPN22 (which encodes PTPN22R620W) is associated with an enhanced susceptibility to multiple autoimmune diseases including type 1 diabetes and rheumatoid arthritis. Many of the associated autoimmune diseases have an autoantibody component to their pathology. Fc receptors (FcRs) recognise autoantibodies when they bind to autoantigens and form immune complexes. After immune complex binding and receptor crosslinking, FcRs signal via Src and Syk family kinases, leading to antigen uptake, presentation and cytokine secretion. Ptpn22 encodes a protein tyrosine phosphatase that negatively regulates Src and Syk family kinases proximal to immunoreceptor signalling cascades. We therefore hypothesised that PTPN22 regulates immune complex stimulated FcR responses in dendritic cells (DCs). Bone marrow derived DCs (BMDCs) from wild type (WT) or Ptpn22-/- mice were pulsed with ovalbumin:anti-ovalbumin immune complexes (ova ICs). Co-culture with WT OT-II T cells revealed that ova IC pulsed Ptpn22-/- BMDCs have an enhanced capability to induce T cell proliferation. This was associated with an increased capability of Ptpn22-/- BMDCs to present immune complex derived antigens and to form ova IC dependent DC-T cell conjugates. These findings highlight PTPN22 as a regulator of FcR mediated responses and provide a link between the association of PTPN22R620W with autoantibody associated autoimmune diseases.
Collapse
Affiliation(s)
- Fiona Clarke
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, United Kingdom.
| | - Harriet A Purvis
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, United Kingdom
| | - Cristina Sanchez-Blanco
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, United Kingdom
| | - Enrique Gutiérrez-Martinez
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, United Kingdom
| | - Georgina H Cornish
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, United Kingdom
| | - Rose Zamoyska
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh, EH9 3FL, United Kingdom
| | - Pierre Guermonprez
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, United Kingdom
| | - Andrew P Cope
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, SE1 1UL, United Kingdom
| |
Collapse
|