1
|
Renert-Yuval Y, Gonzalez J, Garcet S, Williams SC, Moreno A, Krueger JG. Eosinophils in hidradenitis suppurativa patients exhibit pro-inflammatory traits, implicating a potential pathogenic role in the disease. Exp Dermatol 2024; 33:e15129. [PMID: 38984465 DOI: 10.1111/exd.15129] [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: 04/06/2024] [Revised: 06/08/2024] [Accepted: 06/16/2024] [Indexed: 07/11/2024]
Abstract
Hidradenitis suppurativa (HS) is an inflammatory skin disease characterized by painful nodules, abscesses and purulent secretions in intertriginous regions. Intense pruritus frequently accompanies HS lesions, adding further discomfort for patients. While Th17 pathway activation is implicated in HS pathogenesis, disease mechanisms are still not fully understood, and therapeutics are lacking. Previous reports raise a potential role for eosinophils in HS, showing a strong association of eosinophil levels with disease severity. To investigate eosinophils in HS, we recruited patients and matched healthy controls and then performed flow-cytometry studies, eosinophil stimulation assays, and lesional skin staining for eosinophils. We found that HS patients reported similar levels of pain and itch. Compared to matched controls, HS blood exhibited decreased mature eosinophils and increased numbers of immature eosinophils, coupled with a significant increase in dermal eosinophilic infiltrates. Additionally, IL-17RA+ eosinophils were highly and significantly correlated with multiple HS-related clinical scores. In both stimulated and unstimulated conditions, HS eosinophils showed an inflammatory phenotype versus controls, including an increase in costimulatory T- and B-cell markers (e.g. CD5 and CD40) following all stimulations (TNFα/IL-17A/IL-17F). These findings highlight the significance of pruritus in HS and suggest a higher turnover of eosinophils in HS blood, potentially due to the consumption of eosinophils in skin lesions. Our data delineate the features and functions of eosinophils in HS and suggest that eosinophils participate in disease pathogenesis, advancing Th17-related inflammation. Further studies are needed to investigate eosinophils' response to current HS treatments and their potential as a therapeutic target in the disease.
Collapse
Affiliation(s)
- Yael Renert-Yuval
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA
- Pediatric Dermatology Unit, Schneider Children's Medical Center of Israel, Petah Tikva, Israel and Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Juana Gonzalez
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA
| | - Sandra Garcet
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA
| | - Samuel C Williams
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA
- Weill Cornell-Sloan Memorial Sloan Kettering-Rockefeller University, New York, New York, USA
| | - Ariana Moreno
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA
| | - James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA
| |
Collapse
|
2
|
Farhangnia P, Ghomi SM, Mollazadehghomi S, Nickho H, Akbarpour M, Delbandi AA. SLAM-family receptors come of age as a potential molecular target in cancer immunotherapy. Front Immunol 2023; 14:1174138. [PMID: 37251372 PMCID: PMC10213746 DOI: 10.3389/fimmu.2023.1174138] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
The signaling lymphocytic activation molecule (SLAM) family receptors were discovered in immune cells for the first time. The SLAM-family receptors are a significant player in cytotoxicity, humoral immune responses, autoimmune diseases, lymphocyte development, cell survival, and cell adhesion. There is growing evidence that SLAM-family receptors have been involved in cancer progression and heralded as a novel immune checkpoint on T cells. Previous studies have reported the role of SLAMs in tumor immunity in various cancers, including chronic lymphocytic leukemia, lymphoma, multiple myeloma, acute myeloid leukemia, hepatocellular carcinoma, head and neck squamous cell carcinoma, pancreas, lung, and melanoma. Evidence has deciphered that the SLAM-family receptors may be targeted for cancer immunotherapy. However, our understanding in this regard is not complete. This review will discuss the role of SLAM-family receptors in cancer immunotherapy. It will also provide an update on recent advances in SLAM-based targeted immunotherapies.
Collapse
Affiliation(s)
- Pooya Farhangnia
- Immunology Research Center, Institute of Immunology and Infectious Disease, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Shamim Mollazadeh Ghomi
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Shabnam Mollazadehghomi
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hamid Nickho
- Immunology Research Center, Institute of Immunology and Infectious Disease, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahzad Akbarpour
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Advanced Cellular Therapeutics Facility (ACTF), Hematopoietic Cellular Therapy Program, Section of Hematology & Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, United States
| | - Ali-Akbar Delbandi
- Immunology Research Center, Institute of Immunology and Infectious Disease, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Sheu KM, Guru AA, Hoffmann A. Quantifying stimulus-response specificity to probe the functional state of macrophages. Cell Syst 2023; 14:180-195.e5. [PMID: 36657439 PMCID: PMC10023480 DOI: 10.1016/j.cels.2022.12.012] [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: 06/01/2022] [Revised: 10/05/2022] [Accepted: 12/22/2022] [Indexed: 01/19/2023]
Abstract
Immune sentinel macrophages initiate responses to pathogens via hundreds of immune response genes. Each immune threat demands a tailored response, suggesting that the capacity for stimulus-specific gene expression is a key functional hallmark of healthy macrophages. To quantify this property, termed "stimulus-response specificity" (SRS), we developed a single-cell experimental workflow and analytical approaches based on information theory and machine learning. We found that the response specificity of macrophages is driven by combinations of specific immune genes that show low cell-to-cell heterogeneity and are targets of separate signaling pathways. The "response specificity profile," a systematic comparison of multiple stimulus-response distributions, was distinctly altered by polarizing cytokines, and it enabled an assessment of the functional state of macrophages. Indeed, the response specificity profile of peritoneal macrophages from old and obese mice showed characteristic differences, suggesting that SRS may be a basis for measuring the functional state of innate immune cells. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Katherine M Sheu
- Department of Microbiology, Immunology, and Molecular Genetics, and Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, 611 Charles E. Young Dr S, Los Angeles, CA 90093, USA
| | - Aditya A Guru
- Department of Microbiology, Immunology, and Molecular Genetics, and Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, 611 Charles E. Young Dr S, Los Angeles, CA 90093, USA
| | - Alexander Hoffmann
- Department of Microbiology, Immunology, and Molecular Genetics, and Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, 611 Charles E. Young Dr S, Los Angeles, CA 90093, USA.
| |
Collapse
|
4
|
Carenza C, Franzese S, Castagna A, Terzoli S, Simonelli M, Persico P, Bello L, Nibali MC, Pessina F, Kunderfranco P, Peano C, Balin S, Mikulak J, Calcaterra F, Bonecchi R, Savino B, Locati M, Della Bella S, Mavilio D. Perioperative corticosteroid treatment impairs tumor-infiltrating dendritic cells in patients with newly diagnosed adult-type diffuse gliomas. Front Immunol 2023; 13:1074762. [PMID: 36703985 PMCID: PMC9872516 DOI: 10.3389/fimmu.2022.1074762] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction Adult-type diffuse gliomas are malignant primary brain tumors characterized by very poor prognosis. Dendritic cells (DCs) are key in priming antitumor effector functions in cancer, but their role in gliomas remains poorly understood. Methods In this study, we characterized tumor-infiltrating DCs (TIDCs) in adult patients with newly diagnosed diffuse gliomas by using multi-parametric flow cytometry and single-cell RNA sequencing. Results We demonstrated that different subsets of DCs are present in the glioma microenvironment, whereas they are absent in cancer-free brain parenchyma. The largest cluster of TIDCs was characterized by a transcriptomic profile suggestive of severe functional impairment. Patients undergoing perioperative corticosteroid treatment showed a significant reduction of conventional DC1s, the DC subset with key functions in antitumor immunity. They also showed phenotypic and transcriptional evidence of a more severe functional impairment of TIDCs. Discussion Overall, the results of this study indicate that functionally impaired DCs are recruited in the glioma microenvironment. They are severely affected by dexamethasone administration, suggesting that the detrimental effects of corticosteroids on DCs may represent one of the mechanisms contributing to the already reported negative prognostic impact of steroids on glioma patient survival.
Collapse
Affiliation(s)
- Claudia Carenza
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy,Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Sara Franzese
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy,Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Alessandra Castagna
- Laboratory of Leukocyte Biology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Sara Terzoli
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Matteo Simonelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy,Department of Medical Oncology and Hematology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Pasquale Persico
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy,Department of Medical Oncology and Hematology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Lorenzo Bello
- Unit of Oncological Neurosurgery, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Marco Conti Nibali
- Unit of Oncological Neurosurgery, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Federico Pessina
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy,Department of Neurosurgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Paolo Kunderfranco
- Bioinformatics Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Clelia Peano
- Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, Rozzano, Milan, Italy
| | - Simone Balin
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy,Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Joanna Mikulak
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Francesca Calcaterra
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy,Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Raffaella Bonecchi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy,Laboratory of Chemokine Biology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Benedetta Savino
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy,Laboratory of Leukocyte Biology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Massimo Locati
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy,Laboratory of Leukocyte Biology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Silvia Della Bella
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy,Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy,*Correspondence: Silvia Della Bella, ; Domenico Mavilio,
| | - Domenico Mavilio
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy,Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy,*Correspondence: Silvia Della Bella, ; Domenico Mavilio,
| |
Collapse
|
5
|
Dong S, Zhang S, Zhao P, Lin G, Ma X, Xu J, Zhang H, Hu J, Zou C. A combined analysis of bulk and single-cell sequencing data reveals that depleted extracellular matrix and enhanced immune processes co-contribute to fluorouracil beneficial responses in gastric cancer. Front Immunol 2022; 13:999551. [PMID: 36189263 PMCID: PMC9520597 DOI: 10.3389/fimmu.2022.999551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Fluorouracil, also known as 5-FU, is one of the most commonly used chemotherapy drugs in the treatment of advanced gastric cancer (GC). Whereas, the presence of innate or acquired resistance largely limits its survival benefit in GC patients. Although accumulated studies have demonstrated the involvement of tumor microenvironments (TMEs) in chemo-resistance induction, so far little is known about the relevance of GC TMEs in 5-FU resistance. To this end, in this study, we investigated the relationship between TME features and 5-FU responses in GC patients using a combined analysis involving both bulk sequencing data from the TCGA database and single-cell RNA sequencing data from the GEO database. We found that depleted extracellular matrix (ECM) components such as capillary/stroma cells and enhanced immune processes such as increased number of M1 polarized macrophages/Memory T cells/Natural Killer T cells/B cells and decreased number of regulatory T cells are two important features relating to 5-FU beneficial responses in GC patients, especially in diffuse-type patients. We further validated these two features in the tumor tissues of 5-FU-benefit GC patients using immunofluorescence staining experiments. Based on this finding, we also established a Pro (63 genes) and Con (199 genes) gene cohort that could predict 5-FU responses in GC with an AUC (area under curve) score of 0.90 in diffuse-type GC patients, and further proved the partial applicability of this gene panel pan-cancer-wide. Moreover, we identified possible communications mediated by heparanase and galectin-1 which could regulate ECM remodeling and tumor immune microenvironment (TIME) reshaping. Altogether, these findings deciphered the relationship between GC TMEs and 5-FU resistance for the first time, as well as provided potential therapeutic targets and predicting rationale to overcome this chemo-resistance, which could shed some light on developing novel precision treatment strategies in clinical practice.
Collapse
Affiliation(s)
- Shaowei Dong
- The Second Clinical Medical College, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
- School of Medicine, Life and Health Sciences, The Chinese University of Hong Kong (Shenzhen), Shenzhen, China
| | - Siyu Zhang
- School of Medicine, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Pan Zhao
- School of Medicine, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Guanchuan Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Xiaoshi Ma
- The Second Clinical Medical College, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
| | - Jing Xu
- The Second Clinical Medical College, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
| | - Hao Zhang
- Institute of Precision Cancer Medicine and Pathology, Jinan University Medical College, Guangzhou, China
| | - Jiliang Hu
- The Second Clinical Medical College, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
- Guangdong Engineering Technological Research Center for Nervous Anatomy and Related Clinical Applications, Shenzhen, China
| | - Chang Zou
- The Second Clinical Medical College, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
- School of Medicine, Life and Health Sciences, The Chinese University of Hong Kong (Shenzhen), Shenzhen, China
| |
Collapse
|
6
|
Karaky M, Boucher G, Mola S, Foisy S, Beauchamp C, Rivard ME, Burnette M, Gosselin H, Bitton A, Charron G, Goyette P, Rioux JD. Prostaglandins and calprotectin are genetically and functionally linked to the Inflammatory Bowel Diseases. PLoS Genet 2022; 18:e1010189. [PMID: 36155972 PMCID: PMC9536535 DOI: 10.1371/journal.pgen.1010189] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 10/06/2022] [Accepted: 08/31/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Genome wide association studies (GWAS) have identified and validated more than 200 genomic loci associated with the inflammatory bowel disease (IBD), although for most the causal gene remains unknown. Given the importance of myeloid cells in IBD pathogenesis, the current study aimed to uncover the role of genes within IBD genetic loci that are endogenously expressed in this cell lineage. METHODS The open reading frames (ORF) of 42 genes from IBD-associated loci were expressed via lentiviral transfer in the THP-1 model of human monocytes and the impact of each of these on the cell's transcriptome was analyzed using a RNA sequencing-based approach. We used a combination of genetic and pharmacologic approaches to validate our findings in the THP-1 line with further validation in human induced pluripotent stem cell (hiPSC)-derived-monocytes. RESULTS This functional genomics screen provided evidence that genes in four IBD GWAS loci (PTGIR, ZBTB40, SLC39A11 and NFKB1) are involved in controlling S100A8 and S100A9 gene expression, which encode the two subunits of calprotectin (CP). We demonstrated that increasing PTGIR expression and/or stimulating PTGIR signaling resulted in increased CP expression in THP-1. This was further validated in hiPSC-derived monocytes. Conversely, knocking-down PTGIR endogenous expression and/or inhibiting PTGIR signaling led to decreased CP expression. These analyses were extended to the known IBD gene PTGER4, whereby its specific agonist also led to increased CP expression. Furthermore, we demonstrated that the PTGIR and PTGER4 mediated control of CP expression was dependent on signaling via adenylate cyclase and STAT3. Finally, we demonstrated that LPS-mediated increases in CP expression could be potentiated by agonists of PTGIR and PTGER4, and diminished by their antagonists. CONCLUSION Our results support a causal role for the PTGIR, PTGER4, ZBTB40, SLC39A11 and NFKB1 genes in IBD, with all five genes regulating the expression of CP in myeloid cells, as well as potential roles for the prostacyclin/prostaglandin biogenesis and signaling pathways in IBD susceptibility and pathogenesis.
Collapse
Affiliation(s)
- Mohamad Karaky
- Montreal Heart Institute Research Center, Montreal, Quebec, Canada
| | | | - Saraï Mola
- Montreal Heart Institute Research Center, Montreal, Quebec, Canada
| | - Sylvain Foisy
- Montreal Heart Institute Research Center, Montreal, Quebec, Canada
| | | | - Marie-Eve Rivard
- Montreal Heart Institute Research Center, Montreal, Quebec, Canada
| | - Melanie Burnette
- Montreal Heart Institute Research Center, Montreal, Quebec, Canada
| | - Hugues Gosselin
- Montreal Heart Institute Research Center, Montreal, Quebec, Canada
| | - iGenoMed Consortium
- A complete list of members and their affiliations can be found at the end of the manuscript
| | - Alain Bitton
- McGill University Health Centre, Division of Gastroenterology, Montreal, Quebec, Canada
| | - Guy Charron
- Montreal Heart Institute Research Center, Montreal, Quebec, Canada
| | - Philippe Goyette
- Montreal Heart Institute Research Center, Montreal, Quebec, Canada
| | - John D. Rioux
- Montreal Heart Institute Research Center, Montreal, Quebec, Canada
- Université de Montréal, Faculty of Medicine, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Magari M, Nishioka M, Hari T, Ogawa S, Takahashi K, Hatano N, Kanayama N, Futami J, Tokumitsu H. The immunoreceptor SLAMF8 promotes the differentiation of follicular dendritic cell-dependent monocytic cells with B cell-activating ability. FEBS Lett 2022; 596:2659-2667. [PMID: 35953458 DOI: 10.1002/1873-3468.14468] [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: 05/02/2022] [Revised: 06/22/2022] [Accepted: 07/30/2022] [Indexed: 11/06/2022]
Abstract
Follicular dendritic cells (FDCs) play a crucial role in generating high-affinity antibody-producing B cells during the germinal center (GC) reaction. Herein, we analyzed the altered gene expression profile of a mouse FDC line, FL-Y, following lymphotoxin β receptor stimulation, and observed increased Slam-family member 8 (Slamf8) mRNA expression. Forced Slamf8 expression and SLAMF8-Fc addition enhanced the ability of FL-Y cells to induce FDC-induced monocytic cell (FDMC) differentiation. FDMCs accelerated GC-phenotype proliferation in cultured B cells, suggesting that they are capable of promoting GC responses. Furthermore, a pulldown assay showed that SLAMF8-Fc could bind to SLAMF8-His. Overall, the homophilic interaction of SLAMF8 promotes FDMC differentiation and SLAMF8 might act as a novel regulator of GC responses by regulating FDMC differentiation.
Collapse
Affiliation(s)
- Masaki Magari
- Applied Cell Biology, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan.,Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama, 700-8530, Japan
| | - Miku Nishioka
- Applied Cell Biology, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Tomomi Hari
- Applied Cell Biology, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Sayaka Ogawa
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama, 700-8530, Japan
| | - Kaho Takahashi
- Applied Cell Biology, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Naoya Hatano
- Applied Cell Biology, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Naoki Kanayama
- Applied Cell Biology, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan.,Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama, 700-8530, Japan
| | - Junichiro Futami
- Medical Protein Engineering, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Hiroshi Tokumitsu
- Applied Cell Biology, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan.,Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama, 700-8530, Japan
| |
Collapse
|
8
|
Massimini M, Bachetti B, Dalle Vedove E, Benvenga A, Di Pierro F, Bernabò N. A Set of Dysregulated Target Genes to Reduce Neuroinflammation at Molecular Level. Int J Mol Sci 2022; 23:ijms23137175. [PMID: 35806178 PMCID: PMC9266409 DOI: 10.3390/ijms23137175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 01/27/2023] Open
Abstract
Increasing evidence links chronic neurodegenerative diseases with neuroinflammation; it is known that neuroprotective agents are capable of modulating the inflammatory processes, that occur with the onset of neurodegeneration pathologies. Here, with the intention of providing a means for active compounds’ screening, a dysregulation of neuronal inflammatory marker genes was induced and subjected to neuroprotective active principles, with the aim of selecting a set of inflammatory marker genes linked to neurodegenerative diseases. Considering the important role of microglia in neurodegeneration, a murine co-culture of hippocampal cells and inflamed microglia cells was set up. The evaluation of differentially expressed genes and subsequent in silico analysis showed the main dysregulated genes in both cells and the principal inflammatory processes involved in the model. Among the identified genes, a well-defined set was chosen, selecting those in which a role in human neurodegenerative progression in vivo was already defined in literature, matched with the rate of prediction derived from the Principal Component Analysis (PCA) of in vitro treatment-affected genes variation. The obtained panel of dysregulated target genes, including Cxcl9 (Chemokine (C-X-C motif) ligand 9), C4b (Complement Component 4B), Stc1 (Stanniocalcin 1), Abcb1a (ATP Binding Cassette Subfamily B Member 1), Hp (Haptoglobin) and Adm (Adrenomedullin), can be considered an in vitro tool to select old and new active compounds directed to neuroinflammation.
Collapse
Affiliation(s)
- Marcella Massimini
- Faculty of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy
- Correspondence:
| | - Benedetta Bachetti
- R&D Division, C.I.A.M. Srl, 63100 Ascoli Piceno, Italy; (B.B.); (E.D.V.); (A.B.)
| | - Elena Dalle Vedove
- R&D Division, C.I.A.M. Srl, 63100 Ascoli Piceno, Italy; (B.B.); (E.D.V.); (A.B.)
| | - Alessia Benvenga
- R&D Division, C.I.A.M. Srl, 63100 Ascoli Piceno, Italy; (B.B.); (E.D.V.); (A.B.)
| | - Francesco Di Pierro
- Velleja Research, 20125 Milan, Italy;
- Digestive Endoscopy Unit and Gastroenterology, Fondazione Poliambulanza, 25124 Brescia, Italy
| | - Nicola Bernabò
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy;
| |
Collapse
|
9
|
Qin W, Rong X, Yu C, Jia P, Yang J, Zhou G. Knockout of SLAMF8 attenuates collagen-induced rheumatoid arthritis in mice through inhibiting TLR4/NF-κB signaling pathway. Int Immunopharmacol 2022; 107:108644. [DOI: 10.1016/j.intimp.2022.108644] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 12/16/2022]
|
10
|
Yang J, Zhang Q, Zhang J, Ouyang Y, Sun Z, Liu X, Qaio F, Xu LQ, Niu Y, Li J. Exploring the Change of Host and Microorganism in Chronic Obstructive Pulmonary Disease Patients Based on Metagenomic and Metatranscriptomic Sequencing. Front Microbiol 2022; 13:818281. [PMID: 35369515 PMCID: PMC8966909 DOI: 10.3389/fmicb.2022.818281] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a universal respiratory disease resulting from the complex interactions between genes and environmental conditions. The process of COPD is deteriorated by repeated episodes of exacerbations, which are the primary reason for COPD-related morbidity and mortality. Bacterial pathogens are commonly identified in patients’ respiratory tracts both in the stable state and during acute exacerbations, with significant changes in the prevalence of airway bacteria occurring during acute exacerbation of chronic obstructive pulmonary disease (AECOPD). Therefore, the changes in microbial composition and host inflammatory responses will be necessary to investigate the mechanistic link between the airway microbiome and chronic pulmonary inflammation in COPD patients. Methods We performed metatranscriptomic and metagenomic sequencing on sputum samples for twelve AECOPD patients before treatment and for four of them stable COPD (stabilization of AECOPD patients after treatment). Sequencing reads were classified by Kraken2, and the host gene expression was analyzed by Hisat2 and HTseq. The correlation between genes was obtained by the Spearman correlation coefficient. Mann–Whitney U-test was applied to identify microbes that exhibit significantly different distribution in two groups. Results At the phyla level, the top 5 dominant phyla were Firmicutes, Actinobacteria, Proteobacteria, Bacteroidetes, and Fusobacteria. The proportion of dominant gates in metagenomic data was similar in metatranscriptomic data. There were significant differences in the abundance of specific microorganisms at the class level between the two methods. No significant difference between AECOPD and stable COPD was found. However, the different expression levels of 5 host genes were significantly increased in stable COPD and were involved in immune response and inflammatory pathways, which were associated with macrophages. Conclusion Our study may provide a clue to investigate the mechanism of COPD and potential biomarkers in clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Jing Yang
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Qiang Zhang
- Department of Respirology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jun Zhang
- Department of Respirology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | - Zepeng Sun
- China Mobile (Chengdu) Industrial Research Institute, Chengdu, China
| | - Xinlong Liu
- China Mobile (Chengdu) Industrial Research Institute, Chengdu, China
| | - Feng Qaio
- China Mobile (Chengdu) Industrial Research Institute, Chengdu, China
| | - Li-Qun Xu
- China Mobile (Chengdu) Industrial Research Institute, Chengdu, China
| | | | - Jian Li
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Sciences and Technology, Southeast University, Nanjing, China
| |
Collapse
|
11
|
Romero-Pinedo S, Barros DIR, Ruiz-Magaña MJ, Maganto-García E, Moreno de Lara L, Abadía-Molina F, Terhorst C, Abadía-Molina AC. SLAMF8 Downregulates Mouse Macrophage Microbicidal Mechanisms via PI3K Pathways. Front Immunol 2022; 13:910112. [PMID: 35837407 PMCID: PMC9273976 DOI: 10.3389/fimmu.2022.910112] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
Signaling lymphocytic activation molecule family 8 (SLAMF8) is involved in the negative modulation of NADPH oxidase activation. However, the impact of SLAMF8 downregulation on macrophage functionality and the microbicide mechanism remains elusive. To study this in depth, we first analyzed NADPH oxidase activation pathways in wild-type and SLAMF8-deficient macrophages upon different stimulus. Herein, we describe increased phosphorylation of the Erk1/2 and p38 MAP kinases, as well as increased phosphorylation of NADPH oxidase subunits in SLAMF8-deficient macrophages. Furthermore, using specific inhibitors, we observed that specific PI3K inhibition decreased the differences observed between wild-type and SLAMF8-deficient macrophages, stimulated with either PMA, LPS, or Salmonella typhimurium infection. Consequently, SLAMF8-deficient macrophages also showed increased recruitment of small GTPases such as Rab5 and Rab7, and the p47phox subunit to cytoplasmic Salmonella, suggesting an impairment of Salmonella-containing vacuole (SCV) progression in SLAMF8-deficient macrophages. Enhanced iNOS activation, NO production, and IL-6 expression were also observed in the absence of SLAMF8 upon Salmonella infection, either in vivo or in vitro, while overexpression of SLAMF8 in RAW264.7 macrophages showed the opposite phenotype. In addition, SLAMF8-deficient macrophages showed increased activation of Src kinases and reduced SHP-1 phosphate levels upon IFNγ and Salmonella stimuli in comparison to wild-type macrophages. In agreement with in vitro results, Salmonella clearance was augmented in SLAMF8-deficient mice compared to that in wild-type mice. Therefore, in conclusion, SLAMF8 intervention upon bacterial infection downregulates mouse macrophage activation, and confirmed that SLAMF8 receptor could be a potential therapeutic target for the treatment of severe or unresolved inflammatory conditions.
Collapse
Affiliation(s)
- Salvador Romero-Pinedo
- Unidad de Inmunología, Instituto de Biopatología y Medicina Regenerativa (IBIMER), Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - Domingo I Rojas Barros
- Unidad de Inmunología, Instituto de Biopatología y Medicina Regenerativa (IBIMER), Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - María José Ruiz-Magaña
- Unidad de Inmunología, Instituto de Biopatología y Medicina Regenerativa (IBIMER), Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - Elena Maganto-García
- Centro de Biología Molecular "Severo Ochoa" Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Laura Moreno de Lara
- Unidad de Inmunología, Instituto de Biopatología y Medicina Regenerativa (IBIMER), Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - Francisco Abadía-Molina
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain.,Instituto de Nutrición Y Tecnología de los Alimentos "José Mataix", (INYTIA), Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Ana C Abadía-Molina
- Unidad de Inmunología, Instituto de Biopatología y Medicina Regenerativa (IBIMER), Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain.,Departamento de Bioqu´ımica y Biolog´ıa Molecular III e Inmunolog´ıa, Facultad de Medicina, Universidad de Granada, Granada, Spain
| |
Collapse
|
12
|
Shoskes DA, Keslar KS, Gotwald P, Berglund R, Vij S. Neuroinflammatory gene expression in chronic prostatitis/chronic pelvic pain syndrome patients: insights into etiology and phenotype biology. Transl Androl Urol 2021; 10:3340-3347. [PMID: 34532258 PMCID: PMC8421824 DOI: 10.21037/tau-21-387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/15/2021] [Indexed: 01/25/2023] Open
Abstract
Background Chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) has diverse clinical phenotypes and its etiology is multifactorial. Studies to date of gene expression in humans have been limited to small numbers of target genes. NanoString can simultaneously measure hundreds of genes. We wished to study gene expression in blood and urine of CP/CPPS patients compared to controls for neuroinflammatory genes and characterize the results by patient phenotype. Methods Blood and urine were collected from 10 men with CP/CPPS and 7 asymptomatic controls. RNA was isolated from urine pellets using Qiagen RNeasy kits. Whole blood was collected and RNA isolated. 100 ng of RNA was used for gene expression analysis with the 770-gene NanoString Human Neuroinflammation gene panel. Data was imported into Rosalind (OnRamp Bioinformatics) for normalization, calculation of fold-changes and P values, and identification of enriched pathways. Gene expression was considered significantly different if there was a greater than 1.5× change compared to controls and corrected P was <0.05. Results Mean patient age was 42.2 years, median symptom duration was 15.5 months, median UPOINT domains was 3 and mean total National Institute of Health-Chronic Prostatitis Symptom Index Score was 28.8. In blood, there were 5 genes with significantly different expression to controls, the largest differences found in FOS1 (neuropathic pain control), PROS1 (blood clotting) and DDX58 (antiviral innate immunity). Gene set analysis showed differences in inflammation, angiogenesis and cytokine signaling. In urine there were 48 genes with significantly different expression including SLAMF8 (lymphocyte activation) and LAIR1 (inhibits B and T cell function). Gene set analysis showed differences in carbohydrate metabolism, neurons and neurotransmission, adaptive immunity and inflammatory signaling. Subgroup analysis by UPOINT domain showed unique gene expression in the Organ Specific and Neurologic/Systemic domains in both blood and urine for neurogenic pain and cytokine signaling associated genes Conclusions Men with CP/CPPS have a diverse set of neuroinflammatory genes with differential expression compared to controls. Clinical phenotypes have distinct patterns of gene expression. These findings could lead to novel biomarker development, emphasize the importance of multimodal therapy targeting diverse pathways and further validate the biologic basic of clinical phenotyping.
Collapse
Affiliation(s)
- Daniel A Shoskes
- Glickman Urological and Kidney Institute, The Cleveland Clinic, Cleveland, OH, USA
| | - Karen S Keslar
- Glickman Urological and Kidney Institute, The Cleveland Clinic, Cleveland, OH, USA
| | - Paige Gotwald
- Glickman Urological and Kidney Institute, The Cleveland Clinic, Cleveland, OH, USA
| | - Ryan Berglund
- Glickman Urological and Kidney Institute, The Cleveland Clinic, Cleveland, OH, USA
| | - Sarah Vij
- Glickman Urological and Kidney Institute, The Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
13
|
Mangiola S, McCoy P, Modrak M, Souza-Fonseca-Guimaraes F, Blashki D, Stuchbery R, Keam SP, Kerger M, Chow K, Nasa C, Le Page M, Lister N, Monard S, Peters J, Dundee P, Williams SG, Costello AJ, Neeson PJ, Pal B, Huntington ND, Corcoran NM, Papenfuss AT, Hovens CM. Transcriptome sequencing and multi-plex imaging of prostate cancer microenvironment reveals a dominant role for monocytic cells in progression. BMC Cancer 2021; 21:846. [PMID: 34294073 PMCID: PMC8296706 DOI: 10.1186/s12885-021-08529-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/23/2021] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Prostate cancer is caused by genomic aberrations in normal epithelial cells, however clinical translation of findings from analyses of cancer cells alone has been very limited. A deeper understanding of the tumour microenvironment is needed to identify the key drivers of disease progression and reveal novel therapeutic opportunities. RESULTS In this study, the experimental enrichment of selected cell-types, the development of a Bayesian inference model for continuous differential transcript abundance, and multiplex immunohistochemistry permitted us to define the transcriptional landscape of the prostate cancer microenvironment along the disease progression axis. An important role of monocytes and macrophages in prostate cancer progression and disease recurrence was uncovered, supported by both transcriptional landscape findings and by differential tissue composition analyses. These findings were corroborated and validated by spatial analyses at the single-cell level using multiplex immunohistochemistry. CONCLUSIONS This study advances our knowledge concerning the role of monocyte-derived recruitment in primary prostate cancer, and supports their key role in disease progression, patient survival and prostate microenvironment immune modulation.
Collapse
Affiliation(s)
- Stefano Mangiola
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Patrick McCoy
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Martin Modrak
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Fernando Souza-Fonseca-Guimaraes
- University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Daniel Blashki
- The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Ryan Stuchbery
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Simon P Keam
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Michael Kerger
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Ken Chow
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Chayanica Nasa
- Flow Cytometry Facility, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Melanie Le Page
- Flow Cytometry Facility, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Natalie Lister
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Simon Monard
- Flow Cytometry Facility, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Justin Peters
- Epworth Center of Cancer Research, Clayton, Victoria, Australia
| | - Phil Dundee
- Epworth Center of Cancer Research, Clayton, Victoria, Australia
| | - Scott G Williams
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Anthony J Costello
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Paul J Neeson
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Bhupinder Pal
- The Olivia Newton-John Cancer Research Institute, Heidelberg, Melbourne, Australia
| | - Nicholas D Huntington
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Niall M Corcoran
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Urology, Frankston Hospital, Frankston, Victoria, Australia
| | - Anthony T Papenfuss
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- School of Mathematics and Statistics, University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Christopher M Hovens
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| |
Collapse
|
14
|
Kaczanowska S, Beury DW, Gopalan V, Tycko AK, Qin H, Clements ME, Drake J, Nwanze C, Murgai M, Rae Z, Ju W, Alexander KA, Kline J, Contreras CF, Wessel KM, Patel S, Hannenhalli S, Kelly MC, Kaplan RN. Genetically engineered myeloid cells rebalance the core immune suppression program in metastasis. Cell 2021; 184:2033-2052.e21. [PMID: 33765443 PMCID: PMC8344805 DOI: 10.1016/j.cell.2021.02.048] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 09/08/2020] [Accepted: 02/22/2021] [Indexed: 02/07/2023]
Abstract
Metastasis is the leading cause of cancer-related deaths, and greater knowledge of the metastatic microenvironment is necessary to effectively target this process. Microenvironmental changes occur at distant sites prior to clinically detectable metastatic disease; however, the key niche regulatory signals during metastatic progression remain poorly characterized. Here, we identify a core immune suppression gene signature in pre-metastatic niche formation that is expressed predominantly by myeloid cells. We target this immune suppression program by utilizing genetically engineered myeloid cells (GEMys) to deliver IL-12 to modulate the metastatic microenvironment. Our data demonstrate that IL12-GEMy treatment reverses immune suppression in the pre-metastatic niche by activating antigen presentation and T cell activation, resulting in reduced metastatic and primary tumor burden and improved survival of tumor-bearing mice. We demonstrate that IL12-GEMys can functionally modulate the core program of immune suppression in the pre-metastatic niche to successfully rebalance the dysregulated metastatic microenvironment in cancer.
Collapse
Affiliation(s)
- Sabina Kaczanowska
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Daniel W Beury
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Vishaka Gopalan
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Arielle K Tycko
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Haiying Qin
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Miranda E Clements
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Justin Drake
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Chiadika Nwanze
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Meera Murgai
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Zachary Rae
- Single Cell Analysis Facility, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Wei Ju
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Katherine A Alexander
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Jessica Kline
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Cristina F Contreras
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Kristin M Wessel
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Shil Patel
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Sridhar Hannenhalli
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Michael C Kelly
- Single Cell Analysis Facility, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Rosandra N Kaplan
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
15
|
Poveda C, Herreros-Cabello A, Callejas-Hernández F, Osuna-Pérez J, Maza MC, Chillón-Marinas C, Calderón J, Stamatakis K, Fresno M, Gironès N. Interaction of Signaling Lymphocytic Activation Molecule Family 1 (SLAMF1) receptor with Trypanosoma cruzi is strain-dependent and affects NADPH oxidase expression and activity. PLoS Negl Trop Dis 2020; 14:e0008608. [PMID: 32925918 PMCID: PMC7515593 DOI: 10.1371/journal.pntd.0008608] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/24/2020] [Accepted: 07/15/2020] [Indexed: 12/20/2022] Open
Abstract
The receptor Signaling Lymphocyte-Activation Molecule Family 1 (SLAMF1) controls susceptibility to Infection by the lethal Trypanosoma cruzi Y strain. To elucidate whether genetic diversity of the parasite was related with disease susceptibility, we further analyzed the role of SLAMF1 using 6 different Trypanosoma cruzi strains including Y. The interaction of SLAMF1 receptor with T. cruzi was evidenced by fluorescence microscopy, flow cytometry and quantitative PCR. All the strains, except VFRA, showed a decrease in parasite load in infected macrophages in Slamf1-/- compared to BALB/c. In macrophages gene expression NADPH oxidase (NOX2), and reactive oxygen species (ROS) production increased in Slamf1-/- compared to BALB/c in 5 out of 6 strains. However, Slamf1-/-macrophages infected with VFRA strain exhibited a divergent behavior, with higher parasite load, lower NOX2 expression and ROS production compared to BALB/c. Parasitological and immunological studies in vivo with Y strain showed that in the absence of SLAMF1 the immune response protected mice from the otherwise lethal Y infection favoring a proinflammatory response likely involving CD4, CD8, dendritic cells and classically activated macrophages. In the case of VFRA, no major changes were observed in the absence of SLAMF1. Thus, the results suggest that the T. cruzi affects SLAMF1-dependent ROS production, controlling parasite replication in macrophages and affecting survival in mice in a strain-dependent manner. Further studies will focus in the identification of parasite molecules involved in SLAMF1 interaction to explain the immunopathogenesis of the disease.
Collapse
Affiliation(s)
- Cristina Poveda
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Alfonso Herreros-Cabello
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Francisco Callejas-Hernández
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Jesús Osuna-Pérez
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - María C. Maza
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Carlos Chillón-Marinas
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Jossela Calderón
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Konstantinos Stamatakis
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Manuel Fresno
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
- Instituto Sanitario de Investigación Princesa, Madrid, Spain
| | - Núria Gironès
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
- Instituto Sanitario de Investigación Princesa, Madrid, Spain
| |
Collapse
|
16
|
Zhao G, Luo X, Han X, Liu Z. Combining bioinformatics and biological detection to identify novel biomarkers for diagnosis and prognosis of pulmonary tuberculosis. Saudi Med J 2020; 41:351-360. [PMID: 32291421 PMCID: PMC7841615 DOI: 10.15537/smj.2020.4.24989] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 02/03/2020] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVES To identify the novel and promising indicators for pulmonary tuberculosis (PTB) patients. METHODS The study was carried out between June 2016 and June 2019. Three RNA sequencing or microarray datasets of TB infection were used to identify the potential genes showing a common expression trend. The expression level of screened targets was determined by reverse transcription polymerase chain reaction and ELISA using samples of whole blood and peripheral blood mononuclear cells (PBMCs) isolated from 69 PTB patients and 69 healthy volunteers. The potential of the identified targets to predict the treatment outcomes was further studied. RESULTS Bioinformatics analysis demonstrated that a total of 91 genes were up-regulated in all the 3 datasets; among them, the expression of SLAMF8, LILRB4, and IL-10Ra was significantly increased at both the mRNA and protein levels in whole blood and PBMC samples of PTB patients compared with the healthy controls. The mortality rate increased significantly in SLAMF8 or LILRB4 high expression group compared with SLAMF8 or LILRB4 low expression group. Further, the decrease rate of bacteria in patients with SLAMF8 or LILRB4 high expression was slower than that in patients with SLAMF8 or LILRB4 low expression. CONCLUSION This study provides a promising way to identify novel indicators for PTB. Moreover, the LILRB4 expression may play a role in predicting the outcome of treatments on PTB patients.
Collapse
Affiliation(s)
- Guanren Zhao
- Eighth Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China. E-mail.
| | | | | | | |
Collapse
|
17
|
Wilson TJ, Clare S, Mikulin J, Johnson CM, Harcourt K, Lyons PA, Dougan G, Smith KGC. Signalling lymphocyte activation molecule family member 9 is found on select subsets of antigen-presenting cells and promotes resistance to Salmonella infection. Immunology 2020; 159:393-403. [PMID: 31880316 PMCID: PMC7078004 DOI: 10.1111/imm.13169] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/16/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022] Open
Abstract
Signalling lymphocyte activation molecule family member 9 (SLAMF9) is an orphan receptor of the CD2/SLAM family of leucocyte surface proteins. Examination of SLAMF9 expression and function indicates that SLAMF9 promotes inflammation by specialized subsets of antigen‐presenting cells. Within healthy liver and circulating mouse peripheral blood mononuclear cells, SLAMF9 is expressed on CD11b+, Ly6C−, CD11clow, F4/80low, MHC‐II+, CX3CR1+ mononuclear phagocytes as well as plasmacytoid dendritic cells. In addition, SLAMF9 can be found on peritoneal B1 cells and small (F4/80low), but not large (F4/80high), peritoneal macrophages. Upon systemic challenge with Salmonella enterica Typhimurium, Slamf9−/− mice were impaired in their ability to clear the infection from the liver. In humans, SLAMF9 is up‐regulated upon differentiation of monocytes into macrophages, and lipopolysaccharide stimulation of PMA‐differentiated, SLAMF9 knockdown THP‐1 cells showed an essential role of SLAMF9 in production of granulocyte–macrophage colony‐stimulating factor, tumour necrosis factor‐α, and interleukin‐1β. Taken together, these data implicate SLAMF9 in the initiation of inflammation and clearance of bacterial infection.
Collapse
Affiliation(s)
- Timothy J Wilson
- Department of Microbiology, Miami University, Oxford, OH, USA.,Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Simon Clare
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Joseph Mikulin
- Department of Microbiology, Miami University, Oxford, OH, USA
| | | | | | - Paul A Lyons
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK.,Cambridge Institute for Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Gordon Dougan
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK.,Cambridge Institute for Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Kenneth G C Smith
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK.,Cambridge Institute for Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
18
|
O’Connell P, Amalfitano A, Aldhamen YA. SLAM Family Receptor Signaling in Viral Infections: HIV and Beyond. Vaccines (Basel) 2019; 7:E184. [PMID: 31744090 PMCID: PMC6963180 DOI: 10.3390/vaccines7040184] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/04/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
The signaling lymphocytic activation molecule (SLAM) family of receptors are expressed on the majority of immune cells. These receptors often serve as self-ligands, and play important roles in cellular communication and adhesion, thus modulating immune responses. SLAM family receptor signaling is differentially regulated in various immune cell types, with responses generally being determined by the presence or absence of two SLAM family adaptor proteins-Ewing's sarcoma-associated transcript 2 (EAT-2) and SLAM-associated adaptor protein (SAP). In addition to serving as direct regulators of the immune system, certain SLAM family members have also been identified as direct targets for specific microbes and viruses. Here, we will discuss the known roles for these receptors in the setting of viral infection, with special emphasis placed on HIV infection. Because HIV causes such complex dysregulation of the immune system, studies of the roles for SLAM family receptors in this context are particularly exciting.
Collapse
Affiliation(s)
- Patrick O’Connell
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA, (A.A.)
| | - Andrea Amalfitano
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA, (A.A.)
- Department of Pediatrics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Yasser A. Aldhamen
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA, (A.A.)
| |
Collapse
|
19
|
Blohmke CJ, Muller J, Gibani MM, Dobinson H, Shrestha S, Perinparajah S, Jin C, Hughes H, Blackwell L, Dongol S, Karkey A, Schreiber F, Pickard D, Basnyat B, Dougan G, Baker S, Pollard AJ, Darton TC. Diagnostic host gene signature for distinguishing enteric fever from other febrile diseases. EMBO Mol Med 2019; 11:e10431. [PMID: 31468702 PMCID: PMC6783646 DOI: 10.15252/emmm.201910431] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 07/30/2019] [Accepted: 08/09/2019] [Indexed: 12/19/2022] Open
Abstract
Misdiagnosis of enteric fever is a major global health problem, resulting in patient mismanagement, antimicrobial misuse and inaccurate disease burden estimates. Applying a machine learning algorithm to host gene expression profiles, we identified a diagnostic signature, which could distinguish culture-confirmed enteric fever cases from other febrile illnesses (area under receiver operating characteristic curve > 95%). Applying this signature to a culture-negative suspected enteric fever cohort in Nepal identified a further 12.6% as likely true cases. Our analysis highlights the power of data-driven approaches to identify host response patterns for the diagnosis of febrile illnesses. Expression signatures were validated using qPCR, highlighting their utility as PCR-based diagnostics for use in endemic settings.
Collapse
Affiliation(s)
- Christoph J Blohmke
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | | | - Malick M Gibani
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | - Hazel Dobinson
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | - Sonu Shrestha
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | - Soumya Perinparajah
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | - Celina Jin
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | - Harri Hughes
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | - Luke Blackwell
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | - Sabina Dongol
- Patan Academy of Healthy SciencesOxford University Clinical Research UnitKathmanduNepal
| | - Abhilasha Karkey
- Patan Academy of Healthy SciencesOxford University Clinical Research UnitKathmanduNepal
| | | | - Derek Pickard
- Infection Genomics ProgramThe Wellcome Trust Sanger InstituteHinxtonUK
| | - Buddha Basnyat
- Patan Academy of Healthy SciencesOxford University Clinical Research UnitKathmanduNepal
| | - Gordon Dougan
- Infection Genomics ProgramThe Wellcome Trust Sanger InstituteHinxtonUK
| | - Stephen Baker
- The Hospital for Tropical DiseasesWellcome Trust Major Overseas ProgrammeOxford University Clinical Research UnitHo Chi Minh CityVietnam
| | - Andrew J Pollard
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | - Thomas C Darton
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
- The Hospital for Tropical DiseasesWellcome Trust Major Overseas ProgrammeOxford University Clinical Research UnitHo Chi Minh CityVietnam
- Department of Infection, Immunity and Cardiovascular DiseaseUniversity of SheffieldSheffieldUK
| |
Collapse
|
20
|
Sugimoto A, Kataoka TR, Ueshima C, Takei Y, Kitamura K, Hirata M, Nomura T, Haga H. SLAM family member 8 is involved in oncogenic KIT-mediated signalling in human mastocytosis. Exp Dermatol 2019; 27:641-646. [PMID: 29498772 DOI: 10.1111/exd.13523] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2018] [Indexed: 12/26/2022]
Abstract
The signalling lymphocytic activation molecule family member 8 (SLAMF8)/CD353 is a member of the CD2 family of proteins. Its ligand has not been identified. SLAMF8 is expressed by macrophages and suppresses cellular functions. No study has yet explored SLAMF8 expression or function in human mastocytosis, which features oncogenic KIT-mediated proliferation of human mast cells. SLAMF8 protein was expressed in human mastocytosis cells, immunohistochemically. SLAMF8 expression was also evident in the human mast cell lines, HMC1.2 (expressing oncogenic KIT) and LAD2 (expressing wild-type KIT) cells. SLAMF8 knock-down significantly reduced the KIT-mediated growth of HMC1.2 cells but not that of LAD2 cells. SLAMF8 knock-down HMC1.2 cells exhibited significant attenuation of SHP-2 activation and oncogenic KIT-mediated RAS-RAF-ERK signalling. An interaction between SLAMF8 and SHP-2 was confirmed in HMC1.2 cells and all pathological mastocytosis specimens examined (19 of 19 cases, 100%). Thus, SLAMF8 is involved in oncogenic KIT-mediated RAS-RAF-ERK signalling and the subsequent growth of human neoplastic mast cells mediated by SHP-2. SLAMF8 is a possible therapeutic target in human mastocytosis patients.
Collapse
Affiliation(s)
- Akihiko Sugimoto
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Tatsuki R Kataoka
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Chiyuki Ueshima
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Yusuke Takei
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Kyohei Kitamura
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Masahiro Hirata
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Takashi Nomura
- Department of Dermatology, Kyoto University Hospital, Kyoto, Japan
| | - Hironori Haga
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
21
|
Wang X, Lin M, Kang Y. Engineering Porous β-Tricalcium Phosphate (β-TCP) Scaffolds with Multiple Channels to Promote Cell Migration, Proliferation, and Angiogenesis. ACS APPLIED MATERIALS & INTERFACES 2019; 11:9223-9232. [PMID: 30758175 DOI: 10.1021/acsami.8b22041] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Inadequate oxygen and nutrient diffusion in a porous scaffold often resulted in insufficient formation of branched vasculatures, which hindered bone regeneration. In this study, interconnected porous β-tricalcium phosphate (β-TCP) scaffolds with different geometric designs of channels were fabricated and compared to discover the functionality of structure on facilitating nutrient diffusion for angiogenesis. In vitro fluid transportation and degradation of the scaffolds were performed. Cell infiltration, migration, and proliferation of human umbilical vein endothelial cells (HUVECs) on the scaffolds were carried out under both static and dynamic culture conditions. A computational simulation model and a series of immunofluorescent staining were implemented to understand the mechanism of cell behavior in response to different types of scaffolds. Results showed that geometry with multiple channels significantly accelerated the release of Ca2+ and increased the fluid diffusion efficiency. Moreover, multiple channels promoted HUVECs' infiltration and migration in vitro. The ex vivo implantation results showed that the channels promoted cells from the rats' calvarial bone explants to infiltrate into the implanted scaffold. Multiple channels also stimulated HUVECs' proliferation prominently at both static and dynamic culturing conditions. The expression of both cell migration-related protein α5 and angiogenesis-related protein CD31 on multiple-channeled scaffolds was upregulated compared to that on the other two types of scaffolds, implying that multiple channels reinforced cell migration and angiogenesis. All the findings suggested that the geometric design of multiple channels in the porous β-TCP scaffold has promising potential to promote cell infiltration, migration, and further vascularization when implanted in vivo.
Collapse
Affiliation(s)
| | | | - Yunqing Kang
- Integrative Biology PhD Program, Department of Biological Science, College of Science , Florida Atlantic University , Boca Raton , Florida 33431 , United States
| |
Collapse
|
22
|
Zeng X, Liu G, Peng W, He J, Cai C, Xiong W, Chen S, Yang M, Dong Z. Combined deficiency of SLAMF8 and SLAMF9 prevents endotoxin-induced liver inflammation by downregulating TLR4 expression on macrophages. Cell Mol Immunol 2018; 17:153-162. [PMID: 30552382 DOI: 10.1038/s41423-018-0191-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 11/15/2018] [Indexed: 11/09/2022] Open
Abstract
Classical signaling lymphocyte activating molecule (SLAM) family receptors are abundant within many types of immune cells, whereas the nonclassical SLAM family receptors SLAMF8 and SLAMF9, which uniquely lack cytoplasmic signaling motifs, are highly expressed by myeloid cells. Due to the potential redundancy, whether these two receptors regulate macrophage function remains largely unknown. Here, we show that SLAMF8 and SLAMF9 co-regulate macrophage-mediated liver inflammation. To overcome the redundancy, we generated mice that simultaneously lacked SLAMF8 and SLAMF9 using CRISPR-Cas9 technology. Although macrophage differentiation was not altered by the combined deficiency of SLAMF8 and SLAMF9, the loss of these two receptors significantly protected against lipopolysaccharide (LPS)-induced liver injury. SLAMF8 and SLAMF9 double-deficient mice had a prolonged survival rate and less infiltration of inflammatory cells. The depletion of macrophages using clodronate liposomes abolished the effects of SLAMF8 and SLAMF9 deficiencies on LPS-induced liver injury, which demonstrates that these receptors are required for macrophage activation following LPS challenge. Moreover, the deficiency of SLAMF8 and SLAMF9 suppressed the secretion of inflammatory cytokines by downregulating the expression of Toll-like receptor-4 (TLR4), a receptor that specifically binds LPS, which led to decreased mitogen-activated protein kinases (MAPK) signaling activation. Notably, combined injections of truncated extracellular SLAMF8 and SLAMF9 proteins significantly alleviated LPS-induced liver injury. Thus, our findings provide insights into the role of SLAMF8 and SLAMF9 in endotoxin-induced liver injury and suggest that SLAMF8 and SLAMF9 are potential therapeutic targets for acute hepatic injury.
Collapse
Affiliation(s)
- Xiaokang Zeng
- The First Affiliated Hospital, Biomedical Translational Research Institute, Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Guangao Liu
- The First Affiliated Hospital, Biomedical Translational Research Institute, Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Wanwen Peng
- The First Affiliated Hospital, Biomedical Translational Research Institute, Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Junming He
- The First Affiliated Hospital, Biomedical Translational Research Institute, Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Chenxu Cai
- Institute for Immunology and School of Medicine, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, 100086, Beijing, China
| | - Wei Xiong
- Institute for Immunology and School of Medicine, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, 100086, Beijing, China
| | - Shasha Chen
- Institute for Immunology and School of Medicine, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, 100086, Beijing, China
| | - Meixiang Yang
- The First Affiliated Hospital, Biomedical Translational Research Institute, Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China.
| | - Zhongjun Dong
- Institute for Immunology and School of Medicine, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, 100086, Beijing, China.
| |
Collapse
|
23
|
Dollt C, Michel J, Kloss L, Melchers S, Schledzewski K, Becker K, Sauer A, Krewer A, Koll F, Schmieder A. The novel immunoglobulin super family receptor SLAMF9 identified in TAM of murine and human melanoma influences pro-inflammatory cytokine secretion and migration. Cell Death Dis 2018; 9:939. [PMID: 30232321 PMCID: PMC6145869 DOI: 10.1038/s41419-018-1011-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 06/20/2018] [Accepted: 06/22/2018] [Indexed: 01/06/2023]
Abstract
Melanoma is a highly immunogenic tumor with a good response to treatment with immune checkpoint inhibitors. Tumor-associated macrophages (TAMs) play an important immunosuppressive role in such tumors and have therefore been identified as possible future therapeutic targets in oncology. The aim of this study was to identify novel immunoregulatory receptors specifically expressed on TAM. Expression of Slamf9, a member of the signaling lymphocytic-activating molecule (Slam) immunoreceptor family, was found to be upregulated in a gene expression analysis of murine bone marrow-derived macrophages (BMDM) stimulated with tumor-conditioned medium of B16F1 melanoma cells. SLAMF9+ macrophages were identified in human and murine melanomas by using self-generated antibodies against human and murine SLAMF9. A comprehensive immunohistochemical analysis of tissue microarrays detected SLAMF9+ TAM in 73.3% of human melanomas, but also in 95.5% of naevi of melanoma patients and in 50% of naevi from healthy controls. In addition, 20% of melanomas and 2.3% of naevi from melanoma patients displayed a positive SLAMF9 expression also in melanocytic cells. No SLAMF9 expression was detected in naevus cells of healthy donors. Although SLAMF9 has no intracellular signaling motif, a comprehensive functional analysis revealed that the molecule was able to significantly enhance TNF-α secretion after LPS-stimulation. In addition, SLAMF9 delayed the wound closure of RAW 264.7 cells in a scratch assay, while proliferation and cell death were not affected. Taken together, SLAMF9 is a novel type-I-transmembrane receptor with immunomodulatory properties in macrophages. Further studies are required to evaluate whether SLAMF9 classifies as a promising future therapeutic target in melanoma.
Collapse
Affiliation(s)
- Claudia Dollt
- Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Theodor-Kutzer Ufer 1-3, 68167, Mannheim, Germany
| | - Julia Michel
- Institute for Transfusion Medicine and Immunology, Medical Faculty Mannheim, University of Heidelberg, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany.
| | - Loreen Kloss
- Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Theodor-Kutzer Ufer 1-3, 68167, Mannheim, Germany
| | - Susanne Melchers
- Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Theodor-Kutzer Ufer 1-3, 68167, Mannheim, Germany
| | - Kai Schledzewski
- Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Theodor-Kutzer Ufer 1-3, 68167, Mannheim, Germany
| | - Kathrin Becker
- Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Theodor-Kutzer Ufer 1-3, 68167, Mannheim, Germany
| | - Andrea Sauer
- Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Theodor-Kutzer Ufer 1-3, 68167, Mannheim, Germany
| | - Andreas Krewer
- Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Theodor-Kutzer Ufer 1-3, 68167, Mannheim, Germany
| | - Franziska Koll
- Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Theodor-Kutzer Ufer 1-3, 68167, Mannheim, Germany
| | - Astrid Schmieder
- Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Theodor-Kutzer Ufer 1-3, 68167, Mannheim, Germany
| |
Collapse
|
24
|
Radhakrishnan SV, Bhardwaj N, Luetkens T, Atanackovic D. Novel anti-myeloma immunotherapies targeting the SLAM family of receptors. Oncoimmunology 2017. [PMID: 28638731 DOI: 10.1080/2162402x.2017.1308618] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Treatment for multiple myeloma (MM) has significantly advanced in the last decade with the introduction of proteasome inhibitors and immunomodulatory therapies. Unfortunately, MM continues to cause significant morbidity and most patients eventually succumb to the disease. As in other areas of cancer, immunotherapy in MM has also evolved and holds promise to deliver long-lasting remissions or even cure. The signaling lymphocyte activation molecules (SLAM) family of surface proteins represents a group of potential targets for immunotherapy in MM as some of the family members are expressed consistently on plasma cells and also on myeloma propagating pre-plasma cells. Here, we review the SLAM family members in detail, describe their tissue distribution, biologic pathways, as well as relevant pre-clinical studies and clinical trials in MM. Our review demonstrates the value of SLAM family receptors as potential targets for anti-myeloma immunotherapies and outlines how immunotherapeutic approaches can be developed.
Collapse
Affiliation(s)
- Sabarinath Venniyil Radhakrishnan
- Multiple Myeloma Program & Cancer Immunotherapy, Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Neelam Bhardwaj
- Multiple Myeloma Program & Cancer Immunotherapy, Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Tim Luetkens
- Multiple Myeloma Program & Cancer Immunotherapy, Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Djordje Atanackovic
- Multiple Myeloma Program & Cancer Immunotherapy, Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, USA
| |
Collapse
|
25
|
de Lange KM, Moutsianas L, Lee JC, Lamb CA, Luo Y, Kennedy NA, Jostins L, Rice DL, Gutierrez-Achury J, Ji SG, Heap G, Nimmo ER, Edwards C, Henderson P, Mowat C, Sanderson J, Satsangi J, Simmons A, Wilson DC, Tremelling M, Hart A, Mathew CG, Newman WG, Parkes M, Lees CW, Uhlig H, Hawkey C, Prescott NJ, Ahmad T, Mansfield JC, Anderson CA, Barrett JC. Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease. Nat Genet 2017; 49:256-261. [PMID: 28067908 PMCID: PMC5289481 DOI: 10.1038/ng.3760] [Citation(s) in RCA: 838] [Impact Index Per Article: 119.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 12/07/2016] [Indexed: 02/07/2023]
Abstract
Genetic association studies have identified 215 risk loci for inflammatory bowel disease, thereby uncovering fundamental aspects of its molecular biology. We performed a genome-wide association study of 25,305 individuals and conducted a meta-analysis with published summary statistics, yielding a total sample size of 59,957 subjects. We identified 25 new susceptibility loci, 3 of which contain integrin genes that encode proteins in pathways that have been identified as important therapeutic targets in inflammatory bowel disease. The associated variants are correlated with expression changes in response to immune stimulus at two of these genes (ITGA4 and ITGB8) and at previously implicated loci (ITGAL and ICAM1). In all four cases, the expression-increasing allele also increases disease risk. We also identified likely causal missense variants in a gene implicated in primary immune deficiency, PLCG2, and a negative regulator of inflammation, SLAMF8. Our results demonstrate that new associations at common variants continue to identify genes relevant to therapeutic target identification and prioritization.
Collapse
Affiliation(s)
| | - Loukas Moutsianas
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - James C. Lee
- Inflammatory Bowel Disease Research Group, Addenbrooke's Hospital, Cambridge, UK
| | | | - Yang Luo
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
- Division of Genetics and Rheumatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Nicholas A. Kennedy
- Precision Medicine Exeter, University of Exeter, Exeter, UK
- IBD Pharmacogenetics, Royal Devon and Exeter Foundation Trust, Exeter, UK
| | - Luke Jostins
- Wellcome Trust Centre for Human Genetics, University of Oxford, Headington, UK
- Christ Church, University of Oxford, St Aldates, UK
| | - Daniel L. Rice
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | | | - Sun-Gou Ji
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Graham Heap
- Precision Medicine Exeter, University of Exeter, Exeter, UK
- IBD Pharmacogenetics, Royal Devon and Exeter Foundation Trust, Exeter, UK
| | - Elaine R. Nimmo
- Gastrointestinal Unit, Wester General Hospital University of Edinburgh, Edinburgh, UK
| | - Cathryn Edwards
- Department of Gastroenterology, Torbay Hospital, Torbay, Devon, UK
| | - Paul Henderson
- Department of Child Life and Health, University of Edinburgh, Edinburgh, UK
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Sick Children,Edinburgh, UK
| | - Craig Mowat
- Department of Medicine, Ninewells Hospital and Medical School, Dundee, UK
| | - Jeremy Sanderson
- Guy’s & St Thomas’ NHS Foundation Trust, St Thomas’ Hospital, Department of Gastroenterology, London, UK
| | - Jack Satsangi
- Gastrointestinal Unit, Wester General Hospital University of Edinburgh, Edinburgh, UK
| | - Alison Simmons
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
- Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - David C. Wilson
- Paediatric Gastroenterology and Nutrition, Royal Hospital for Sick Children, Edinburgh, UK
- Child Life and Health, University of Edinburgh, Edinburgh, Scotland, UK
| | - Mark Tremelling
- Gastroenterology & General Medicine, Norfolk and Norwich University Hospital, Norwich, UK
| | - Ailsa Hart
- Department of Medicine, St Mark's Hospital, Harrow, Middlesex, UK
| | - Christopher G. Mathew
- Department of Medical and Molecular Genetics, Faculty of Life Science and Medicine, King's College London, Guy's Hospital, London, UK
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of Witwatersrand, South Africa
| | - William G. Newman
- Genetic Medicine, Manchester Academic Health Science Centre, Manchester, UK
- The Manchester Centre for Genomic Medicine, University of Manchester, Manchester, UK
| | - Miles Parkes
- Inflammatory Bowel Disease Research Group, Addenbrooke's Hospital, Cambridge, UK
| | - Charlie W. Lees
- Gastrointestinal Unit, Wester General Hospital University of Edinburgh, Edinburgh, UK
| | - Holm Uhlig
- Translational Gastroenterology Unit and the Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Chris Hawkey
- Nottingham Digestive Diseases Centre, Queens Medical Centre, Nottingham, UK
| | - Natalie J. Prescott
- Department of Medical and Molecular Genetics, Faculty of Life Science and Medicine, King's College London, Guy's Hospital, London, UK
| | - Tariq Ahmad
- Precision Medicine Exeter, University of Exeter, Exeter, UK
- IBD Pharmacogenetics, Royal Devon and Exeter Foundation Trust, Exeter, UK
| | - John C. Mansfield
- Institute of Human Genetics, Newcastle University, Newcastle upon Tyne, UK
| | - Carl A. Anderson
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Jeffrey C. Barrett
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| |
Collapse
|
26
|
Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease. Nat Genet 2017. [PMID: 28067908 DOI: 10.1038/ng.3760.] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Genetic association studies have identified 215 risk loci for inflammatory bowel disease, thereby uncovering fundamental aspects of its molecular biology. We performed a genome-wide association study of 25,305 individuals and conducted a meta-analysis with published summary statistics, yielding a total sample size of 59,957 subjects. We identified 25 new susceptibility loci, 3 of which contain integrin genes that encode proteins in pathways that have been identified as important therapeutic targets in inflammatory bowel disease. The associated variants are correlated with expression changes in response to immune stimulus at two of these genes (ITGA4 and ITGB8) and at previously implicated loci (ITGAL and ICAM1). In all four cases, the expression-increasing allele also increases disease risk. We also identified likely causal missense variants in a gene implicated in primary immune deficiency, PLCG2, and a negative regulator of inflammation, SLAMF8. Our results demonstrate that new associations at common variants continue to identify genes relevant to therapeutic target identification and prioritization.
Collapse
|
27
|
Lewis MD, Kelly JM. Putting Infection Dynamics at the Heart of Chagas Disease. Trends Parasitol 2016; 32:899-911. [PMID: 27612651 PMCID: PMC5086431 DOI: 10.1016/j.pt.2016.08.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/15/2016] [Accepted: 08/16/2016] [Indexed: 12/12/2022]
Abstract
In chronic Trypanosoma cruzi infections, parasite burden is controlled by effective, but nonsterilising immune responses. Infected cells are difficult to detect because they are scarce and focally distributed in multiple sites. However, advances in detection technologies have established a link between parasite persistence and the pathogenesis of Chagas heart disease. Long-term persistence likely involves episodic reinvasion as well as continuous infection, to an extent that varies between tissues. The primary reservoir sites in humans are not definitively known, but analysis of murine models has identified the gastrointestinal tract. Here, we highlight that quantitative, spatial, and temporal aspects of T. cruzi infection are central to a fuller understanding of the association between persistence, pathogenesis, and immunity, and for optimising treatment.
Collapse
Affiliation(s)
- Michael D Lewis
- Department of Pathogen Molecular Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK; Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA.
| | - John M Kelly
- Department of Pathogen Molecular Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| |
Collapse
|
28
|
Lewis MD, Francisco AF, Taylor MC, Jayawardhana S, Kelly JM. Host and parasite genetics shape a link between Trypanosoma cruzi infection dynamics and chronic cardiomyopathy. Cell Microbiol 2016; 18:1429-43. [PMID: 26918803 PMCID: PMC5031194 DOI: 10.1111/cmi.12584] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 02/21/2016] [Indexed: 12/15/2022]
Abstract
Host and parasite diversity are suspected to be key factors in Chagas disease pathogenesis. Experimental investigation of underlying mechanisms is hampered by a lack of tools to detect scarce, pleiotropic infection foci. We developed sensitive imaging models to track Trypanosoma cruzi infection dynamics and quantify tissue-specific parasite loads, with minimal sampling bias. We used this technology to investigate cardiomyopathy caused by highly divergent parasite strains in BALB/c, C3H/HeN and C57BL/6 mice. The gastrointestinal tract was unexpectedly found to be the primary site of chronic infection in all models. Immunosuppression induced expansion of parasite loads in the gut and was followed by widespread dissemination. These data indicate that differential immune control of T. cruzi occurs between tissues and shows that the large intestine and stomach provide permissive niches for active infection. The end-point frequency of heart-specific infections ranged from 0% in TcVI-CLBR-infected C57BL/6 to 88% in TcI-JR-infected C3H/HeN mice. Nevertheless, infection led to fibrotic cardiac pathology in all models. Heart disease severity was associated with the model-dependent frequency of dissemination outside the gut and inferred cumulative heart-specific parasite loads. We propose a model of cardiac pathogenesis driven by periodic trafficking of parasites into the heart, occurring at a frequency determined by host and parasite genetics.
Collapse
Affiliation(s)
- Michael D Lewis
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK.
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, 20892, USA.
| | - Amanda Fortes Francisco
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Martin C Taylor
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Shiromani Jayawardhana
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - John M Kelly
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| |
Collapse
|
29
|
Haslund-Vinding J, McBean G, Jaquet V, Vilhardt F. NADPH oxidases in oxidant production by microglia: activating receptors, pharmacology and association with disease. Br J Pharmacol 2016; 174:1733-1749. [PMID: 26750203 DOI: 10.1111/bph.13425] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 12/15/2015] [Accepted: 01/07/2016] [Indexed: 12/26/2022] Open
Abstract
Microglia are the resident immune cells of the CNS and constitute a self-sustaining population of CNS-adapted tissue macrophages. As mononuclear phagocytic cells, they express high levels of superoxide-producing NADPH oxidases (NOX). The sole function of the members of the NOX family is to generate reactive oxygen species (ROS) that are believed to be important in CNS host defence and in the redox signalling circuits that shape the different activation phenotypes of microglia. NOX are also important in pathological conditions, where over-generation of ROS contributes to neuronal loss via direct oxidative tissue damage or disruption of redox signalling circuits. In this review, we assess the evidence for involvement of NOX in CNS physiopathology, with particular emphasis on the most important surface receptors that lead to generation of NOX-derived ROS. We evaluate the potential significance of the subcellular distribution of NOX isoforms for redox signalling or release of ROS to the extracellular medium. Inhibitory mechanisms that have been reported to restrain NOX activity in microglia and macrophages in vivo are also discussed. We provide a critical appraisal of frequently used and recently developed NOX inhibitors. Finally, we review the recent literature on NOX and other sources of ROS that are involved in activation of the inflammasome and discuss the potential influence of microglia-derived oxidants on neurogenesis, neural differentiation and culling of surplus progenitor cells. The degree to which excessive, badly timed or misplaced NOX activation in microglia may affect neuronal homeostasis in physiological or pathological conditions certainly merits further investigation. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- J Haslund-Vinding
- Institute of Cellular and Molecular Medicine, Copenhagen University, Copenhagen, Denmark.,Department of Pathology and Immunology, Centre Médical Universitaire, Geneva, Switzerland
| | - G McBean
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Ireland
| | - V Jaquet
- Department of Pathology and Immunology, Centre Médical Universitaire, Geneva, Switzerland
| | - F Vilhardt
- Institute of Cellular and Molecular Medicine, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
30
|
van Driel BJ, Liao G, Engel P, Terhorst C. Responses to Microbial Challenges by SLAMF Receptors. Front Immunol 2016; 7:4. [PMID: 26834746 PMCID: PMC4718992 DOI: 10.3389/fimmu.2016.00004] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/06/2016] [Indexed: 12/24/2022] Open
Abstract
The SLAMF family (SLAMF) of cell surface glycoproteins is comprised of nine glycoproteins and while SLAMF1, 3, 5, 6, 7, 8, and 9 are self-ligand receptors, SLAMF2 and SLAMF4 interact with each other. Their interactions induce signal transduction networks in trans, thereby shaping immune cell-cell communications. Collectively, these receptors modulate a wide range of functions, such as myeloid cell and lymphocyte development, and T and B cell responses to microbes and parasites. In addition, several SLAMF receptors serve as microbial sensors, which either positively or negatively modulate the function of macrophages, dendritic cells, neutrophils, and NK cells in response to microbial challenges. The SLAMF receptor-microbe interactions contribute both to intracellular microbicidal activity as well as to migration of phagocytes to the site of inflammation. In this review, we describe the current knowledge on how the SLAMF receptors and their specific adapters SLAM-associated protein and EAT-2 regulate innate and adaptive immune responses to microbes.
Collapse
Affiliation(s)
- Boaz Job van Driel
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| | - Gongxian Liao
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| | - Pablo Engel
- Immunology Unit, Department of Cell Biology, Immunology and Neurosciences, Medical School, University of Barcelona , Barcelona , Spain
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
31
|
Bologna C, Buonincontri R, Serra S, Vaisitti T, Audrito V, Brusa D, Pagnani A, Coscia M, D'Arena G, Mereu E, Piva R, Furman RR, Rossi D, Gaidano G, Terhorst C, Deaglio S. SLAMF1 regulation of chemotaxis and autophagy determines CLL patient response. J Clin Invest 2015; 126:181-94. [PMID: 26619119 DOI: 10.1172/jci83013] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 10/29/2015] [Indexed: 01/22/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a variable disease; therefore, markers to identify aggressive forms are essential for patient management. Here, we have shown that expression of the costimulatory molecule and microbial sensor SLAMF1 (also known as CD150) is lost in a subset of patients with an aggressive CLL that associates with a shorter time to first treatment and reduced overall survival. SLAMF1 silencing in CLL-like Mec-1 cells, which constitutively express SLAMF1, modulated pathways related to cell migration, cytoskeletal organization, and intracellular vesicle formation and recirculation. SLAMF1 deficiency associated with increased expression of CXCR4, CD38, and CD44, thereby positively affecting chemotactic responses to CXCL12. SLAMF1 ligation with an agonistic monoclonal antibody increased ROS accumulation and induced phosphorylation of p38, JNK1/2, and BCL2, thereby promoting the autophagic flux. Beclin1 dissociated from BCL2 in response to SLAMF1 ligation, resulting in formation of the autophagy macrocomplex, which contains SLAMF1, beclin1, and the enzyme VPS34. Accordingly, SLAMF1-silenced cells or SLAMF1(lo) primary CLL cells were resistant to autophagy-activating therapeutic agents, such as fludarabine and the BCL2 homology domain 3 mimetic ABT-737. Together, these results indicate that loss of SLAMF1 expression in CLL modulates genetic pathways that regulate chemotaxis and autophagy and that potentially affect drug responses, and suggest that these effects underlie unfavorable clinical outcome experienced by SLAMF1(lo) patients.
Collapse
MESH Headings
- Antigens, CD/physiology
- Autophagy
- Cell Movement
- Chemotaxis
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- MAP Kinase Kinase 4/antagonists & inhibitors
- Reactive Oxygen Species/metabolism
- Receptors, Cell Surface/physiology
- Signaling Lymphocytic Activation Molecule Family Member 1
Collapse
|