51
|
Skurk T, Krämer T, Marcinek P, Malki A, Lang R, Dunkel A, Krautwurst T, Hofmann TF, Krautwurst D. Sweetener System Intervention Shifted Neutrophils from Homeostasis to Priming. Nutrients 2023; 15:nu15051260. [PMID: 36904259 PMCID: PMC10005247 DOI: 10.3390/nu15051260] [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: 01/30/2023] [Revised: 02/27/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Non-nutritive sweeteners (NNS) are part of personalized nutrition strategies supporting healthy glycemic control. In contrast, the consumption of non-nutritive sweeteners has been related to person-specific and microbiome-dependent glycemic impairments. Reports on the effects of NNS on our highly individual cellular immune system are sparse. The recent identification of taste receptor expression in a variety of immune cells, however, suggested their immune-modulatory relevance. METHODS We studied the influence of a beverage-typical NNS system on the transcriptional profiling of sweetener-cognate taste receptors, selected cytokines and their receptors, and on Ca2+ signaling in isolated blood neutrophils. We determined plasma concentrations of saccharin, acesulfame-K, and cyclamate by HPLC-MS/MS, upon ingestion of a soft drink-typical sweetener surrogate. In an open-labeled, randomized intervention study, we determined pre- versus post-intervention transcript levels by RT-qPCR of sweetener-cognate taste receptors and immune factors. RESULTS Here we show that the consumption of a food-typical sweetener system modulated the gene expression of cognate taste receptors and induced the transcriptional regulation signatures of early homeostasis- and late receptor/signaling- and inflammation-related genes in blood neutrophils, shifting their transcriptional profile from homeostasis to priming. Notably, sweeteners at postprandial plasma concentrations facilitated fMLF (N-formyl-Met-Leu-Phe)-induced Ca2+ signaling. CONCLUSIONS Our results support the notion of sweeteners priming neutrophils to higher alertness towards their adequate stimuli.
Collapse
Affiliation(s)
- Thomas Skurk
- ZIEL Institute for Food and Health, Core Facility Human Studies, TUM School for Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | - Tamara Krämer
- Leibniz Institute for Food Systems Biology, Technical University of Munich, 85354 Freising, Germany
| | - Patrick Marcinek
- Leibniz Institute for Food Systems Biology, Technical University of Munich, 85354 Freising, Germany
| | - Agne Malki
- Leibniz Institute for Food Systems Biology, Technical University of Munich, 85354 Freising, Germany
| | - Roman Lang
- Leibniz Institute for Food Systems Biology, Technical University of Munich, 85354 Freising, Germany
| | - Andreas Dunkel
- Leibniz Institute for Food Systems Biology, Technical University of Munich, 85354 Freising, Germany
| | - Tiffany Krautwurst
- TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Thomas F. Hofmann
- Chair of Food Chemistry and Molecular Sensory Science, Technical University of Munich, 85354 Freising, Germany
| | - Dietmar Krautwurst
- Leibniz Institute for Food Systems Biology, Technical University of Munich, 85354 Freising, Germany
- Correspondence:
| |
Collapse
|
52
|
Goto T, Michiue T, Shibuya H. ccl19 and ccl21 affect cell movements and differentiation in early Xenopus development. Dev Growth Differ 2023; 65:175-189. [PMID: 36861303 DOI: 10.1111/dgd.12847] [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: 01/16/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023]
Abstract
We characterized Xenopus laevis C-C motif chemokine ligand 19.L (ccl19.L) and C-C motif chemokine ligand 21.L (ccl21.L) during early Xenopus embryogenesis. The temporal and spatial expression patterns of ccl19.L and ccl21.L tended to show an inverse correlation, except that the expression level was higher in the dorsal side at the gastrula stage. For example, even at the dorsal sector of the gastrulae, ccl19.L was expressed in the axial region and ccl21.L was expressed in the paraxial region. Dorsal overexpression of ccl19.L and ccl21.L and knockdown of Ccl19.L and Ccl21.L inhibited gastrulation, but their functions were different in cell behaviors during morphogenesis. Observation of Keller sandwich explants revealed that overexpression of both ccl19.L and ccl21.L and knockdown of Ccl21.L inhibited the convergent extension movements, while knockdown of Ccl19.L did not. ccl19.L-overexpressing explants attracted cells at a distance and ccl21.L-overexpressing explants attracted neighboring cells. Ventral overexpression of ccl19.L and ccl21.L induced secondary axis-like structures and chrd.1 expression at the ventral side. Upregulation of chrd.1 was induced by ligand mRNAs through ccr7.S. Knockdown of Ccl19.L and Ccl21.L inhibited gastrulation and downregulated chrd.1 expression at the dorsal side. The collective findings indicate that ccl19.L and ccl21.L might play important roles in morphogenesis and dorsal-ventral patterning during early embryogenesis in Xenopus.
Collapse
Affiliation(s)
- Toshiyasu Goto
- Department of Molecular Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tatsuo Michiue
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Shibuya
- Department of Molecular Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
53
|
Xue S, Su XM, Ke LN, Huang YG. CXCL9 correlates with antitumor immunity and is predictive of a favorable prognosis in uterine corpus endometrial carcinoma. Front Oncol 2023; 13:1077780. [PMID: 36845675 PMCID: PMC9945585 DOI: 10.3389/fonc.2023.1077780] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 01/17/2023] [Indexed: 02/11/2023] Open
Abstract
Background The C-X-C motif chemokine ligand-9 (CXCL9) is related to the progression of multiple neoplasms. Yet, its biological functions in uterine corpus endometrioid carcinoma (UCEC) remain shrouded in confusion. Here, we assessed the prognostic significance and potential mechanism of CXCL9 in UCEC. Methods Firstly, bioinformatics analysis of the public cancer database, including the Cancer Genome Atlas / the Genotype-Tissue Expression project (TCGA+ GTEx, n=552) and Gene Expression Omnibus (GEO): GSE63678 (n=7), were utilized for the CXCL9 expression-related analysis in UCEC. Then, the survival analysis of TCGA-UCEC was performed. Futher, the gene set enrichment analysis (GSEA) was carried out to reveal the potential molecular signaling pathway in UCEC associated with CXCL9 expression. Moreover, the immunohistochemistry (IHC) assay of our validation cohort (n=124) from human specimens were used to demonstrate the latent significance of CXCL9 in UCEC. Results The bioinformatics analysis suggested that CXCL9 expression was significantly upregulated in UCEC patients; and hyper-expression of CXCL9 was related to prolonged survival. the GSEA enrichment analysis showed various immune response-related pathways, including T/NK cell, lymphocyte activation, cytokine-cytokine receptor interaction network, and chemokine signaling pathway, mediated by CXCL9. In addition, the cytotoxic molecules (IFNG, SLAMF7, JCHAIN, NKG7, GBP5, LYZ, GZMA, GZMB, and TNF3F9) and the immunosuppressive genes (including PD-L1) were positively related to the expression of CXCL9. Further, the IHC assay indicated that the CXCL9 protein expression was mainly located in intertumoral and significantly upregulated in the UCEC patients; UCEC with high intertumoral CXCL9 cell abundance harbored an improved prognosis; a higher ratio of anti-tumor immune cells (CD4+, CD8+, and CD56+ cell) and PD-L1 was found in UCEC with CXCL9 high expression. Conclusion Overexpressed CXCL9 correlates with antitumor immunity and is predictive of a favorable prognosis in UCEC. It hinted that CXCL9 may serve as an independent prognostic biomarker or therapeutic target in UCEC patients, which augmented anti-tumor immune effects to furnish survival benefits.
Collapse
Affiliation(s)
- Shen Xue
- Department of obstetrics and gynecology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiao-min Su
- Department of Immunology, Nankai University School of Medicine, Tianjin, China
| | - Li-na Ke
- Department of obstetrics and gynecology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China,*Correspondence: Yu-gang Huang, ; Li-na Ke,
| | - Yu-gang Huang
- Department of Pathology, Taihe Hospital, Hubei University of Medicine, Shiyan, China,*Correspondence: Yu-gang Huang, ; Li-na Ke,
| |
Collapse
|
54
|
Cousin VN, Perez GF, Payne KJ, Voll RE, Rizzi M, Mueller CG, Warnatz K. Lymphoid stromal cells - potential implications for the pathogenesis of CVID. Front Immunol 2023; 14:1122905. [PMID: 36875120 PMCID: PMC9982092 DOI: 10.3389/fimmu.2023.1122905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/03/2023] [Indexed: 02/19/2023] Open
Abstract
Non-hematopoietic lymphoid stromal cells (LSC) maintain lymph node architecture and form niches allowing the migration, activation, and survival of immune cells. Depending on their localization in the lymph node, these cells display heterogeneous properties and secrete various factors supporting the different activities of the adaptive immune response. LSCs participate in the transport of antigen from the afferent lymph as well as in its delivery into the T and B cell zones and organize cell migration via niche-specific chemokines. While marginal reticular cells (MRC) are equipped for initial B-cell priming and T zone reticular cells (TRC) provide the matrix for T cell-dendritic cell interactions within the paracortex, germinal centers (GC) only form when both T- and B cells successfully interact at the T-B border and migrate within the B-cell follicle containing the follicular dendritic cell (FDC) network. Unlike most other LSCs, FDCs are capable of presenting antigen via complement receptors to B cells, which then differentiate within this niche and in proximity to T follicular helper (TFH) cells into memory and plasma cells. LSCs are also implicated in maintenance of peripheral immune tolerance. In mice, TRCs induce the alternative induction of regulatory T cells instead of TFH cells by presenting tissue-restricted self-antigens to naïve CD4 T cells via MHC-II expression. This review explores potential implications of our current knowledge of LSC populations regarding the pathogenesis of humoral immunodeficiency and autoimmunity in patients with autoimmune disorders or common variable immunodeficiency (CVID), the most common form of primary immunodeficiency in humans.
Collapse
Affiliation(s)
- Victoria N Cousin
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,University of Freiburg, Faculty of Biology, Freiburg, Germany.,Freiburg Spemann Graduate School of Biology and Medicine (SGBM), Albert Ludwigs University Freiburg, Faculty of Biology, Freiburg, Germany
| | - Guillermo F Perez
- Immunologie, Immunopathologie et Chimie Thérapeutique, CNRS UPR3572, Strasbourg, France.,Faculty of Life Science, University of Strasbourg, Strasbourg, France
| | - Kathryn J Payne
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,University of Freiburg, Faculty of Biology, Freiburg, Germany
| | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marta Rizzi
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Division of Clinical and Experimental Immunology, Institute of Immunology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Christopher G Mueller
- Immunologie, Immunopathologie et Chimie Thérapeutique, CNRS UPR3572, Strasbourg, France.,Faculty of Life Science, University of Strasbourg, Strasbourg, France
| | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
55
|
Wang Z, Liu H, Gong Y, Cheng Y. Establishment and validation of an aging-related risk signature associated with prognosis and tumor immune microenvironment in breast cancer. Eur J Med Res 2022; 27:317. [PMID: 36581948 PMCID: PMC9798726 DOI: 10.1186/s40001-022-00924-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/01/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is a highly malignant and heterogeneous tumor which is currently the cancer with the highest incidence and seriously endangers the survival and prognosis of patients. Aging, as a research hotspot in recent years, is widely considered to be involved in the occurrence and development of a variety of tumors. However, the relationship between aging-related genes (ARGs) and BC has not yet been fully elucidated. MATERIALS AND METHODS The expression profiles and clinicopathological data were acquired in the Cancer Genome Atlas (TCGA) and the gene expression omnibus (GEO) database. Firstly, the differentially expressed ARGs in BC and normal breast tissues were investigated. Based on these differential genes, a risk model was constructed composed of 11 ARGs via univariate and multivariate Cox analysis. Subsequently, survival analysis, independent prognostic analysis, time-dependent receiver operating characteristic (ROC) analysis and nomogram were performed to assess its ability to sensitively and specifically predict the survival and prognosis of patients, which was also verified in the validation set. In addition, functional enrichment analysis and immune infiltration analysis were applied to reveal the relationship between the risk scores and tumor immune microenvironment, immune status and immunotherapy. Finally, multiple datasets and real-time polymerase chain reaction (RT-PCR) were utilized to verify the expression level of the key genes. RESULTS An 11-gene signature (including FABP7, IGHD, SPIB, CTSW, IGKC, SEZ6, S100B, CXCL1, IGLV6-57, CPLX2 and CCL19) was established to predict the survival of BC patients, which was validated by the GEO cohort. Based on the risk model, the BC patients were divided into high- and low-risk groups, and the high-risk patients showed worse survival. Stepwise ROC analysis and Cox analyses demonstrated the good performance and independence of the model. Moreover, a nomogram combined with the risk score and clinical parameters was built for prognostic prediction. Functional enrichment analysis revealed the robust relationship between the risk model with immune-related functions and pathways. Subsequent immune microenvironment analysis, immunotherapy, etc., indicated that the immune status of patients in the high-risk group decreased, and the anti-tumor immune function was impaired, which was significantly different with those in the low-risk group. Eventually, the expression level of FABP7, IGHD, SPIB, CTSW, IGKC, SEZ6, S100B, CXCL1, IGLV6-57 and CCL19 was identified as down-regulated in tumor cell line, while CPLX2 up-regulated, which was mostly similar with the results in TCGA and Human Protein Atlas (HPA) via RT-PCR. CONCLUSIONS In summary, our study constructed a risk model composed of ARGs, which could be used as a solid model for predicting the survival and prognosis of BC patients. Moreover, this model also played an important role in tumor immunity, providing a new direction for patient immune status assessment and immunotherapy selection.
Collapse
Affiliation(s)
- Zitao Wang
- grid.412632.00000 0004 1758 2270Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei China
| | - Hua Liu
- grid.412632.00000 0004 1758 2270Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei China
| | - Yiping Gong
- grid.412632.00000 0004 1758 2270Department of Breast Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei China
| | - Yanxiang Cheng
- grid.412632.00000 0004 1758 2270Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei China
| |
Collapse
|
56
|
Seyran M, Melanie S, Philip S, Amiq G, Fabian B. Allies or enemies? The effect of regulatory T cells and related T lymphocytes on the profibrotic environment in bleomycin-injured lung mouse models. Clin Exp Med 2022:10.1007/s10238-022-00945-7. [PMID: 36403186 PMCID: PMC10390389 DOI: 10.1007/s10238-022-00945-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/07/2022] [Indexed: 11/21/2022]
Abstract
AbstractIdiopathic pulmonary fibrosis (IPF) is characterized by permanent scarring of lung tissue and declining lung function, and is an incurable disease with increase in prevalence over the past decade. The current consensus is that aberrant wound healing following repeated injuries to the pulmonary epithelium is the most probable cause of IPF, with various immune inflammatory pathways having been reported to impact disease pathogenesis. While the role of immune cells, specifically T lymphocytes and regulatory T cells (Treg), in IPF pathogenesis has been reported and discussed recently, the pathogenic or beneficial roles of these cells in inducing or preventing lung fibrosis is still debated. This lack of understanding could be due in part to the difficulty in obtaining diseased human lung tissue for research purposes. For this reason, many animal models have been developed over the years to attempt to mimic the main clinical hallmarks of IPF: among these, inducing lung injury in rodents with the anti-cancer agent bleomycin has now become the most commonly studied animal model of IPF. Pulmonary fibrosis is the major side effect when bleomycin is administered for cancer treatment in human patients, and a similar effect can be observed after intra-tracheal administration of bleomycin to rodents. Despite many pathophysiological pathways of lung fibrosis having been investigated in bleomycin-injured animal models, one central facet still remains controversial, namely the involvement of specific T lymphocyte subsets, and in particular Treg, in disease pathogenesis. This review aims to summarize the major findings and conclusions regarding the involvement of immune cells and their receptors in the pathogenesis of IPF, and to elaborate on important parallels between animal models and the human disease. A more detailed understanding of the role of Treg and other immune cell subsets in lung injury and fibrosis derived from animal models is a critical basis for translating this knowledge to the development of new immune-based therapies for the treatment of human IPF.
Collapse
|
57
|
Whyte CE, Tumes DJ, Liston A, Burton OT. Do more with Less: Improving High Parameter Cytometry Through Overnight Staining. Curr Protoc 2022; 2:e589. [PMID: 36373983 PMCID: PMC9827877 DOI: 10.1002/cpz1.589] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Recent advances in flow cytometry have allowed high-dimensional characterization of biological phenomena, enabling breakthroughs in a multitude of fields. Despite the appreciation of the unique properties of antigens and fluorophores in high-parameter panel design, staining conditions are often standardized for short surface stains, regardless of antibody affinity or antigen accessibility. Here, we demonstrate how increasing antibody incubation times can lead to substantial improvements in sensitivity, maintaining specificity, and reducing background, while also significantly reducing the costs of high-parameter cytometry panels. Furthermore, overnight staining reduces the influence of interexperimental variability, assisting accurate pooling over experiments over extended time courses. We provide guidance on how to optimize staining conditions for diverse antigens, including how different fixation strategies can affect epitope accessibility. Overnight staining can thus substantially improve the resolution, repeatability, and cost-effectiveness of high-parameter cytometry. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Carly E. Whyte
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideAustralia
| | - Damon J. Tumes
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideAustralia
| | - Adrian Liston
- Immunology ProgrammeBabraham InstituteCambridgeUnited Kingdom
| | | |
Collapse
|
58
|
Thomas H, McCloskey E, Rider V. Pregnancy preparation: redistribution of CCR7-positive cells in the rat uterus. Reproduction 2022; 164:183-193. [PMID: 35960551 PMCID: PMC10531295 DOI: 10.1530/rep-22-0074] [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: 03/12/2022] [Accepted: 08/12/2022] [Indexed: 11/08/2022]
Abstract
In brief Changes in the endometrium prior to implantation may be critical in predicting pregnancy outcomes. This study shows that the endocrine system directs positional changes in CCR7+ cells before implantation, which may be critical for developing maternal tolerance. Abstract Suppression of the maternal immune system is vital for the implantation of the semi-allogeneic embryo. Although progress in understanding the dialogue between mother and embryo has been made, key interactions between maternal immune cells, hormones, and chemokines remain elusive. Uterine expression of the C-C chemokine receptor type 7 (CCR7) could recruit T regulatory cells and facilitate localized immune suppression. To test this concept, Ccr7 mRNA and protein were assessed in uterine tissue. Ccr7 mRNA expression peaked at day 4 in pregnant rat uteri and then declined at days 5 and 6. CCR7 protein showed similar quantitative changes. To test if female sex steroids affected the spatial distribution of CCR7-expressing cells, uteri from ovariectomized rats, progesterone-pretreated rats (2 mg daily), and progesterone-pretreated rats injected with estradiol (0.2 µg) were analyzed. Progesterone increased CCR7-positive (+) cells in the antimesometrial stroma. Progesterone and estradiol increased CCR7+ cells in the mesometrial stroma. Estradiol increased the density of cluster of differentiation 4 (CD4) positive cells in the mesometrial stromal region over progesterone alone. The density of cells expressing the T regulatory cell marker, forkhead box protein 3 (FOXP3), increased in the antimesometrial stroma in response to progesterone alone. Progesterone and estradiol increased FOXP3+ cells in the antimesometrial region of the stroma. Co-localization of CCR7, CD4, and FOXP3 in the stroma suggests CCR7+ cells are T regulatory cells. Polarization of CCR7+ cells in the endometrial stroma was an intrinsic response regulated by sex steroids and did not require the presence of an embryo.
Collapse
Affiliation(s)
- Hannah Thomas
- 1Department of Biology, Pittsburg State University, Pittsburg, Kansas, USA
| | - Erick McCloskey
- 1Department of Biology, Pittsburg State University, Pittsburg, Kansas, USA
| | - Virginia Rider
- 1Department of Biology, Pittsburg State University, Pittsburg, Kansas, USA
| |
Collapse
|
59
|
Didriksen H, Molberg Ø, Mehta A, Jordan S, Palchevskiy V, Fretheim H, Gude E, Ueland T, Brunborg C, Garen T, Midtvedt Ø, Andreassen AK, Lund-Johansen F, Distler O, Belperio J, Hoffmann-Vold AM. Target organ expression and biomarker characterization of chemokine CCL21 in systemic sclerosis associated pulmonary arterial hypertension. Front Immunol 2022; 13:991743. [PMID: 36211384 PMCID: PMC9541617 DOI: 10.3389/fimmu.2022.991743] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Systemic sclerosis (SSc) is a heterogenous disorder that appears to result from interplay between vascular pathologies, tissue fibrosis and immune processes, with evidence for deregulation of chemokines, which normally control immune trafficking. We recently identified altered levels of chemokine CCL21 in SSc associated pulmonary arterial hypertension (PAH). Here, we aimed to define target organ expression and biomarker characteristics of CCL21. Materials and methods To investigate target organ expression of CCL21, we performed immunohistochemistry (IHC) on explanted lung tissues from SSc-PAH patients. We assessed serum levels of CCL21 by ELISA and Luminex in two well-characterized SSc cohorts from Oslo (OUH, n=552) and Zurich (n=93) University hospitals and in 168 healthy controls. For detection of anti-CCl21 antibodies, we performed protein array analysis applying serum samples from SSc patients (n=300) and healthy controls. To characterize circulating CCL21 in SSc, we applied immunoprecipitation (IP) with antibodies detecting both full length and tailless and a custom-made antibody detecting only the C-terminal of CCL21. IP products were analyzed by SDS-PAGE/western blot and Mass spectrometry (MS). Results By IHC, we found that CCL21 was mainly expressed in the airway epithelial cells of SSc patients with PAH. In the analysis of serum levels of CCL21 we found weak correlation between Luminex and ELISA (r=0.515, p<0.001). Serum levels of anti-CCL21 antibodies were higher in SSc patients than in healthy controls (p<0.001), but only 5% of the SSc population were positive for anti-CCL21 antibodies in SSc, and we found no correlation between anti-CCl21 and serum levels of CCL21. By MS, we only identified peptides located within amino acid (aa) 23-102 of CCL21, indicating that CCL21 in SSc circulate as a truncated protein without the C-terminal tail. Conclusion This study demonstrates expression of CCL21 in epithelial lung tissue from SSc patients with PAH, and indicate that CCL21 in SSc circulates as a truncated protein. We extend previous observations indicating biomarker potential of CCL21, but find that Luminex is not suitable as platform for biomarker analyses. Finally, in vivo generated anti-CCL21 antibodies exist in SSc, but do not appear to modify serum CCL21 levels in patients with SSc-PAH.
Collapse
Affiliation(s)
- Henriette Didriksen
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Øyvind Molberg
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Adi Mehta
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Suzana Jordan
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Vyacheslav Palchevskiy
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Håvard Fretheim
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Einar Gude
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital – Rikshospitalet, Oslo, Norway
| | - Cathrine Brunborg
- Oslo Centre for Biostatistics and Epidemiology, Research Support Services, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Torhild Garen
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Øyvind Midtvedt
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Arne K. Andreassen
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | | | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - John Belperio
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Anna-Maria Hoffmann-Vold
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- *Correspondence: Anna-Maria Hoffmann-Vold,
| |
Collapse
|
60
|
Yamaguchi T, Iijima H, Yoshihara T, Tani M, Otake Y, Iwatani S, Amano T, Tashiro T, Kurahashi T, Inoue T, Tsujii Y, Hayashi Y, Inoue T, Motooka D, Nakamura S, Shinzaki S, Takehara T. Exacerbation of non-steroidal anti-inflammatory drug-induced enteropathy in C-C chemokine receptor type 7-deficient mice. J Gastroenterol Hepatol 2022; 37:1561-1570. [PMID: 35435994 DOI: 10.1111/jgh.15868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 02/10/2022] [Accepted: 04/08/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND AND AIM Non-steroidal anti-inflammatory drugs (NSAIDs) induce intestinal enteropathy and the pathophysiology is related to immune-mediated mechanisms. We aimed to investigate the role of C-C chemokine receptor type 7 (CCR7) which regulates immune cell migration in NSAID-induced enteropathy. METHODS Injury of the small intestine was evaluated 24 h after the subcutaneous injection of indomethacin in CCR7-deficient (Ccr7-/- ) and wild-type (WT) mice. The cellular profile and cytokine production in intestinal cells were analyzed. Indomethacin-induced enteropathy was evaluated in mice adoptively transferred with CD103+ dendritic cells (DCs) from Ccr7-/- or WT mice. RESULTS Indomethacin induced more severe intestinal injury in Ccr7-/- mice than in WT mice. The major inflammatory cytokines were not increased and the proportion of regulatory T cells following indomethacin injection was not decreased in Ccr7-/- mice compared with WT mice. The expression of interleukin (IL)-22 binding protein (IL-22BP), which inhibits IL-22 activity, was significantly higher in CD103+ DCs from Ccr7-/- mice than those from WT mice. Mice adoptively transferred with CD103+ DCs isolated from Ccr7-/- mice exhibited more severe intestinal injury following indomethacin injection compared with those adoptively transferred with CD103+ DCs of WT mice. Ccr7-/- mice injected with indomethacin showed a significant reduction in regenerating islet-derived 1 (Reg1) mRNA expression, which is regulated by IL-22, in intestinal epithelial cells. CONCLUSIONS C-C chemokine receptor type 7 deficiency exacerbated NSAID-induced enteropathy in association with an altered phenotype of CD103+ DCs that produces IL-22BP. CCR7 contributes to protect the small intestine from NSAID-induced mucosal injury.
Collapse
Affiliation(s)
- Toshio Yamaguchi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan.,Department of Gastroenterology, Osaka Rosai Hospital, Osaka, Japan
| | - Hideki Iijima
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takeo Yoshihara
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Mizuki Tani
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuriko Otake
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shuko Iwatani
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takahiro Amano
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Taku Tashiro
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomohide Kurahashi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takanori Inoue
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshiki Tsujii
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshito Hayashi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takahiro Inoue
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Daisuke Motooka
- Department of Infection Metagenomics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Shota Nakamura
- Department of Infection Metagenomics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Shinichiro Shinzaki
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
61
|
Zhou WH, Wang Y, Yan C, Du WD, Al-Aroomi MA, Zheng L, Lin SF, Gao JX, Jiang S, Wang ZX, Sun CF, Liu FY. CC chemokine receptor 7 promotes macrophage recruitment and induces M2-polarization through CC chemokine ligand 19&21 in oral squamous cell carcinoma. Discov Oncol 2022; 13:67. [PMID: 35904690 PMCID: PMC9338204 DOI: 10.1007/s12672-022-00533-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/13/2022] [Indexed: 12/14/2022] Open
Abstract
PURPOSE This study aimed to investigate the impact of CC chemokine receptor 7 (CCR7) on the recruitment and polarization of tumor-associated macrophages (TAMs) in oral squamous cell carcinoma (OSCC). METHODS We analyzed CCR7 expression pattern, clinicopathological significance, and its association with M2 macrophage infiltration in OSCC by bioinformatic methods. Small interfering RNA (siRNA) was utilized to silence CCR7 in OSCC cells. Conditioned media (CM) was harvested from transfected OSCC cells to establish a co-culture model of THP-1 derived macrophages and OSCC cells. Transwell assay and cell adhesion assay were performed to examine the effect of CCR7 on macrophages recruitment and adhesion. Cytoskeleton was labelled by phalloidin to observe macrophage morphological changes. Moreover, phenotypic alteration of macrophages was measured using quantitative real-time PCR (qRT-PCR), flow cytometry, and immunofluorescence (IF) staining. Ultimately, recombinant human CCL19 and CCL21 were added into the medium of THP-1 derived macrophages to explore their effects on polarization in vitro. RESULTS In OSCC patients, the overexpression of CCR7 positively correlated with lymph node metastasis and M2 macrophage infiltration. Macrophage not only exhibited enhanced migration, invasion and adhesion abilities, but also appeared more spindle and branched in vitro when treated with CM from OSCC cells. However, these phenomena were abrogated with knockdown of CCR7. We also discovered that inhibition of CCR7 in OSCC cells suppressed TAMs polarization to an M2 phenotype. In addition, recombinant human CCL19 and CCL21 promoted macrophage M2-polarization in vitro. CONCLUSION CCR7 in OSCC cells promoted recruitment and M2-polarization of THP-1 derived macrophages in vitro by regulating production of CCL19 and CCL21.
Collapse
Affiliation(s)
- Wan-Hang Zhou
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Yao Wang
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Cong Yan
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Wei-Dong Du
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Maged Ali Al-Aroomi
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Li Zheng
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Shan-Feng Lin
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Jia-Xing Gao
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Sheng Jiang
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Zeng-Xu Wang
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Chang-Fu Sun
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China
| | - Fa-Yu Liu
- Department of Oral Maxillofacial-Head and Neck Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, 117 Nanjing North Road, Heping District, Shenyang, 110000, Liaoning, China.
| |
Collapse
|
62
|
Wang Q, Chu F, Zhang X, Hu H, Lu L, Wang F, Yu Y, Zhang Y, Ma J, Xu Z, Eldemery F, Ou C, Liu X. Infectious bursal disease virus replication is inhibited by avain T cell chemoattractant chemokine CCL19. Front Microbiol 2022; 13:912908. [PMID: 35935208 PMCID: PMC9355407 DOI: 10.3389/fmicb.2022.912908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Chemokine CCL19, together with its receptor CCR7, is one of the most important factors recruiting immune cells into target organ during virus infection. Our previous study has shown that CCL19 played a vital role in the process of T cell trafficking into bursae during bursal disease virus (IBDV) infection. In this study, we hypothesized that CCL19 could exert direct influences on IBDV replication other than recruiting immune cells. A eukaryotic expression vector of pEGFP-N1/CCL19 was successfully constructed and identified by PCR, double enzymes digestion, and sequencing. Different concentrations of pEGFP-N1/CCL19 plasmids were transfected into DF1 cells and CCL19 protein was highly expressed. Then, DF1 cells were infected with IBDV B87 strain post-transfection. Based on PCR and Western blot results, CCL19 could obviously decrease the gene levels of VP1 and VP2 and the protein levels of VP2 and VP3. When CCL19 was knocked down, the gene levels of VP1 and VP2 were significantly upregulated. Moreover, indirect immunostaining revealed that the IBDV content was largely decreased after CCL19 overexpression. Additionally, CCL19 inhibitory effects might rely on activation of the JNK signal pathway. Taken together, chemokine CCL19 directly blocks IBDV replication in DF1 cells, indicating that CCL19 could play crucial functions other than recruiting T cells during the pathogenesis of IBDV.
Collapse
Affiliation(s)
- Qiuxia Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Fuming Chu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Xin Zhang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Huilong Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Lang Lu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Fang Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Yan Yu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Yanhong Zhang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Jinyou Ma
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Zhiyong Xu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Fatma Eldemery
- Department of Hygiene and Zoonoses, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Changbo Ou
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
- College of Animal Science and Technology, Guangxi University, Nanning, China
- *Correspondence: Changbo Ou
| | - Xingyou Liu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
- College of Life Science, Xinxiang University, Xinxiang, China
- Xingyou Liu
| |
Collapse
|
63
|
Mesquida-Veny F, Martínez-Torres S, Del Rio JA, Hervera A. Nociception-Dependent CCL21 Induces Dorsal Root Ganglia Axonal Growth via CCR7-ERK Activation. Front Immunol 2022; 13:880647. [PMID: 35911704 PMCID: PMC9331658 DOI: 10.3389/fimmu.2022.880647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/25/2022] [Indexed: 11/30/2022] Open
Abstract
While chemokines were originally described for their ability to induce cell migration, many studies show how these proteins also take part in many other cell functions, acting as adaptable messengers in the communication between a diversity of cell types. In the nervous system, chemokines participate both in physiological and pathological processes, and while their expression is often described on glial and immune cells, growing evidence describes the expression of chemokines and their receptors in neurons, highlighting their potential in auto- and paracrine signalling. In this study we analysed the role of nociception in the neuronal chemokinome, and in turn their role in axonal growth. We found that stimulating TRPV1+ nociceptors induces a transient increase in CCL21. Interestingly we also found that CCL21 enhances neurite growth of large diameter proprioceptors in vitro. Consistent with this, we show that proprioceptors express the CCL21 receptor CCR7, and a CCR7 neutralizing antibody dose-dependently attenuates CCL21-induced neurite outgrowth. Mechanistically, we found that CCL21 binds locally to its receptor CCR7 at the growth cone, activating the downstream MEK-ERK pathway, that in turn activates N-WASP, triggering actin filament ramification in the growth cone, resulting in increased axonal growth.
Collapse
Affiliation(s)
- Francina Mesquida-Veny
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Sara Martínez-Torres
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Jose Antonio Del Rio
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Arnau Hervera
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
- *Correspondence: Arnau Hervera,
| |
Collapse
|
64
|
Chemokines and NSCLC: Emerging role in prognosis, heterogeneity, and therapeutics. Semin Cancer Biol 2022; 86:233-246. [PMID: 35787939 DOI: 10.1016/j.semcancer.2022.06.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/15/2022] [Accepted: 06/24/2022] [Indexed: 12/11/2022]
Abstract
Lung cancer persists to contribute to one-quarter of cancer-associated deaths. Among the different histologies, non-small cell lung cancer (NSCLC) alone accounts for 85% of the cases. The development of therapies involving immune checkpoint inhibitors and angiogenesis inhibitors has increased patients' survival probability and reduced mortality rates. Developing targeted therapies against essential genetic alterations also translates to better treatment strategies. But the benefits still seem farfetched due to the development of drug resistance and refractory tumors. In this review, we have highlighted the interplay of different tumor microenvironment components, essentially discussing the chemokine families (CC, CXC, C, and CX3C) that regulate the tumor biology in NSCLC and promote tumor growth, metastasis, and associated heterogeneity. The development of therapeutics and prognostic markers is a complex and multipronged approach. However, some essential chemokines can act as critical players for being considered potential prognostic markers and therapeutic targets.
Collapse
|
65
|
Xu J, Li JQ, Chen QL, Shestakova EA, Misyurin VA, Pokrovsky VS, Tchevkina EM, Chen HB, Song H, Zhang JY. Advances in Research on the Effects and Mechanisms of Chemokines and Their Receptors in Cancer. Front Pharmacol 2022; 13:920779. [PMID: 35770088 PMCID: PMC9235028 DOI: 10.3389/fphar.2022.920779] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/05/2022] [Indexed: 01/10/2023] Open
Abstract
Cancer is a common and intractable disease that seriously affects quality of life of patients and imposes heavy economic burden on families and the entire society. Current medications and intervention strategies for cancer have respective shortcomings. In recent years, it has been increasingly spotlighted that chemokines and their receptors play vital roles in the pathophysiology of cancer. Chemokines are a class of structurally similar short-chain secreted proteins that initiate intracellular signaling pathways through the activation of corresponding G protein-coupled receptors and participate in physiological and pathological processes such as cell migration and proliferation. Studies have shown that chemokines and their receptors have close relationships with cancer epigenetic regulation, growth, progression, invasion, metastasis, and angiogenesis. Chemokines and their receptors may also serve as potential targets for cancer treatment. We herein summarize recent research progresses on anti-tumor effects and mechanisms of chemokines and their receptors, suggesting avenues for future studies. Perspectives for upcoming explorations, such as development of multi-targeted chemokine-based anti-tumor drugs, are also discussed in the present review.
Collapse
Affiliation(s)
- Jing Xu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Jing-quan Li
- The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Qi-lei Chen
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Elena A. Shestakova
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vsevolod A. Misyurin
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vadim S. Pokrovsky
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Biochemistry, People’s Friendship University, Moscow, Russia
| | - Elena M. Tchevkina
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Hu-biao Chen
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Hang Song
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Jian-ye Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
66
|
Plasminogen and plasmin can bind to human T cells and generate truncated CCL21 that increases dendritic cell chemotactic responses. J Biol Chem 2022; 298:102112. [PMID: 35690148 PMCID: PMC9270246 DOI: 10.1016/j.jbc.2022.102112] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 11/22/2022] Open
Abstract
Plasmin is a broad-spectrum protease and therefore needs to be tightly regulated. Active plasmin is formed from plasminogen, which is found in high concentrations in the blood and is converted by the plasminogen activators. In the circulation, high levels of α2-antiplasmin rapidly and efficiently inhibit plasmin activity. Certain myeloid immune cells have been shown to bind plasmin and plasminogen on their cell surface via proteins that bind to the plasmin(ogen) kringle domains. Our earlier work showed that T cells can activate plasmin, but that they do not themselves express plasminogen. Here, we demonstrate that T cells express several known plasminogen receptors, and that they bind plasminogen on their cell surface. We show T cell-bound plasminogen was converted to plasmin by plasminogen activators upon T cell activation. To examine functional consequences of plasmin generation by activated T cells, we investigated its effect on the chemokine, C-C Motif Chemokine Ligand 21 (CCL21). Video microscopy and western blotting confirmed that plasmin bound by human T cells cleaves CCL21 and increases the chemotactic response of monocyte-derived dendritic cells towards higher CCL21 concentrations along the concentration gradient by increasing their directional migration and track straightness. These results demonstrate how migrating T cells and potentially other activated immune cells may co-opt a powerful proteolytic system from the plasma towards immune processes in the peripheral tissues, where α2-antiplasmin is more likely to be absent. We propose that plasminogen bound to migrating immune cells may strongly modulate chemokine responses in peripheral tissues.
Collapse
|
67
|
McCright J, Naiknavare R, Yarmovsky J, Maisel K. Targeting Lymphatics for Nanoparticle Drug Delivery. Front Pharmacol 2022; 13:887402. [PMID: 35721179 PMCID: PMC9203826 DOI: 10.3389/fphar.2022.887402] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/16/2022] [Indexed: 12/25/2022] Open
Abstract
The lymphatics transport material from peripheral tissues to lymph nodes, where immune responses are formed, before being transported into systemic circulation. With key roles in transport and fluid homeostasis, lymphatic dysregulation is linked to diseases, including lymphedema. Fluid within the interstitium passes into initial lymphatic vessels where a valve system prevents fluid backflow. Additionally, lymphatic endothelial cells produce key chemokines, such as CCL21, that direct the migration of dendritic cells and lymphocytes. As a result, lymphatics are an attractive delivery route for transporting immune modulatory treatments to lymph nodes where immunotherapies are potentiated in addition to being an alternative method of reaching systemic circulation. In this review, we discuss the physiology of lymphatic vessels and mechanisms used in the transport of materials from peripheral tissues to lymph nodes. We then summarize nanomaterial-based strategies to take advantage of lymphatic transport functions for delivering therapeutics to lymph nodes or systemic circulation. We also describe opportunities for targeting lymphatic endothelial cells to modulate transport and immune functions.
Collapse
Affiliation(s)
| | | | | | - Katharina Maisel
- Department of Bioengineering, University of Maryland College Park, College Park, MD, United States
| |
Collapse
|
68
|
Kim DH, Lee YH, Sayed AEDH, Choi IY, Lee JS. Genome-wide identification of 194 G protein-coupled receptor (GPCR) genes from the water flea Daphnia magna. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2022; 42:100983. [PMID: 35367896 DOI: 10.1016/j.cbd.2022.100983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/07/2023]
Abstract
In crustaceans, G protein-coupled receptors (GPCRs) are the largest transmembrane receptor family and function by mediating various environmental stimuli in cells. Understanding GPCR signaling is crucial to better understanding of crustacean endocrinology. GPCRs evolved from early eukaryotes, and genome-wide identification of GPCRs in a particular taxon can provide insight into evolutionary tendencies and adaptive strategies of GPCR response to environmental stimuli. Here, we identified 194 full-length GPCR genes in the water flea Daphnia magna that can be divided into five distinct classes (A, B, C, F, and other). A strong orthologous relationship for amine, neuropeptide, and opsin receptors was found in the phylogenetic comparison of D. magna GPCRs to those of humans and two well-known insects (Drosophila melanogaster and Solenopsis invicta). Our results based on phylogenetic relationships suggest that most GPCRs subfamilies have undergone sporadic evolutionary processes for adaptation to environmental pressures. Despite the dynamics of GPCR evolution, some GPCRs are highly conserved between species. This study provides a better understanding of the evolution of GPCRs and expands our knowledge of the potential physiological mechanisms in D. magna in response to various environmental stimuli.
Collapse
Affiliation(s)
- Duck-Hyun Kim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Young Hwan Lee
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Alaa El-Din H Sayed
- Department of Zoology, Faculty of Science, Assiut University, 71516 Assiut, Egypt
| | - Ik-Young Choi
- Department of Agricultural Life Industry, College of Lifelong Learning, Kangwon National University, Chuncheon 24341, South Korea.
| | - Jae-Seong Lee
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
69
|
Wang J, Qin D, Ye L, Wan L, Wang F, Yang Y, Ma Y, Yang H, Yang Z, Chen M, Jiang W, Zhang Q. CCL19 has potential to be a potential prognostic biomarker and a modulator of tumor immune microenvironment (TIME) of breast cancer: a comprehensive analysis based on TCGA database. Aging (Albany NY) 2022; 14:4158-4175. [PMID: 35550569 PMCID: PMC9134962 DOI: 10.18632/aging.204081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/02/2022] [Indexed: 11/25/2022]
Abstract
The development of cancer was determined by not only the intrinsic properties of cancer cells, but also the communication between cancer cells and tumor microenvironment (TME). We applied ESTIMATE and CIBERSORT algorithms to calculate the immune/stromal component and tumor-infiltrating immune cells (TICs) in TME of BC. The results showed that immune component in TME predicted patients’ survival and associated with progression of BC. Differentially expressed genes (DEGs) were primarily enriched in immune-related activities. Finally, CCL19 was acquired which shared the leading nodes in PPI network and was associated with patients’ survival. High expression of CCL19 predicted better prognosis and participated in progression of BC. Genes in CCL19 up-regulated group were enriched in immune-related activities and these functions might depend on the communications between CCL19 and multiple TICs in TIME. In conclusion, CCL19 functioned as a potential prognostic biomarker and a modulator of TIME in BC through communicating with various TICs.
Collapse
Affiliation(s)
- Jinyan Wang
- Department of Oncology, Nanjing Jiangning Hospital, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Dongmei Qin
- Department of Pathology, Nanjing Jiangning Hospital, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Lingling Ye
- Department of Oncology, Nanjing Jiangning Hospital, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Li Wan
- Department of Oncology, The Second Hospital of Nanjing, Nanjing, China
| | - Fen Wang
- Department of Oncology, Nanjing Jiangning Hospital, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Yang
- Department of Oncology, Nanjing Jiangning Hospital, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Yajun Ma
- Department of Oncology, Nanjing Jiangning Hospital, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Hui Yang
- Department of Oncology, Nanjing Jiangning Hospital, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Zhaohui Yang
- Department of Oncology, Nanjing Jiangning Hospital, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Meili Chen
- Department of Oncology, Nanjing Jiangning Hospital, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Wen Jiang
- Department of Oncology, Nanjing Jiangning Hospital, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Quan'an Zhang
- Department of Oncology, Nanjing Jiangning Hospital, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
70
|
Shifting CCR7 towards Its Monomeric Form Augments CCL19 Binding and Uptake. Cells 2022; 11:cells11091444. [PMID: 35563750 PMCID: PMC9101108 DOI: 10.3390/cells11091444] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/12/2022] [Accepted: 04/21/2022] [Indexed: 01/20/2023] Open
Abstract
The chemokine receptor CCR7, together with its ligands, is responsible for the migration and positioning of adaptive immune cells, and hence critical for launching adaptive immune responses. CCR7 is also induced on certain cancer cells and contributes to metastasis formation. Thus, CCR7 expression and signalling must be tightly regulated for proper function. CCR7, like many other members of the G-protein coupled receptor superfamily, can form homodimers and oligomers. Notably, danger signals associated with pathogen encounter promote oligomerisation of CCR7 and is considered as one layer of regulating its function. Here, we assessed the dimerisation of human CCR7 and several single point mutations using split-luciferase complementation assays. We demonstrate that dimerisation-defective CCR7 mutants can be transported to the cell surface and elicit normal chemokine-driven G-protein activation. By contrast, we discovered that CCR7 mutants whose expression are shifted towards monomers significantly augment their capacities to bind and internalise fluorescently labelled CCL19. Modeling of the receptor suggests that dimerisation-defective CCR7 mutants render the extracellular loops more flexible and less structured, such that the chemokine recognition site located in the binding pocket might become more accessible to its ligand. Overall, we provide new insights into how the dimerisation state of CCR7 affects CCL19 binding and receptor trafficking.
Collapse
|
71
|
Saxena V, Piao W, Li L, Paluskievicz C, Xiong Y, Simon T, Lakhan R, Brinkman CC, Walden S, Hippen KL, WillsonShirkey M, Lee YS, Wagner C, Blazar BR, Bromberg JS. Treg tissue stability depends on lymphotoxin beta-receptor- and adenosine-receptor-driven lymphatic endothelial cell responses. Cell Rep 2022; 39:110727. [PMID: 35443187 PMCID: PMC9093052 DOI: 10.1016/j.celrep.2022.110727] [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: 12/14/2021] [Revised: 02/23/2022] [Accepted: 03/30/2022] [Indexed: 02/03/2023] Open
Abstract
Regulatory T cell (Treg) lymphatic migration is required for resolving inflammation and prolonging allograft survival. Focusing on Treg interactions with lymphatic endothelial cells (LECs), we dissect mechanisms and functional consequences of Treg transendothelial migration (TEM). Using three genetic mouse models of pancreatic islet transplantation, we show that Treg lymphotoxin (LT) αβ and LEC LTβ receptor (LTβR) signaling are required for efficient Treg migration and suppressive function to prolong allograft survival. Inhibition of LT signaling increases Treg conversion to Foxp3loCD25lo exTregs. In a transwell-based model of TEM across polarized LECs, non-migrated Tregs become exTregs. Such conversion is regulated by LTβR nuclear factor κB (NF-κB) signaling in LECs, which increases interleukin-6 (IL-6) production and drives exTreg conversion. Migrating Tregs are ectonucleotidase CD39hi and resist exTreg conversion in an adenosine-receptor-2A-dependent fashion. Human Tregs migrating across human LECs behave similarly. These molecular interactions can be targeted for therapeutic manipulation of immunity and suppression.
Collapse
Affiliation(s)
- Vikas Saxena
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Wenji Piao
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Lushen Li
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Christina Paluskievicz
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yanbao Xiong
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Thomas Simon
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Ram Lakhan
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - C Colin Brinkman
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sarah Walden
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Pediatrics, University of Minnesota Cancer Center, Minneapolis, MN 55455, USA
| | - Keli L Hippen
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Pediatrics, University of Minnesota Cancer Center, Minneapolis, MN 55455, USA
| | - Marina WillsonShirkey
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Young S Lee
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Chelsea Wagner
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Pediatrics, University of Minnesota Cancer Center, Minneapolis, MN 55455, USA
| | - Jonathan S Bromberg
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
72
|
Mayberry CL, Logan NA, Wilson JJ, Chang CH. Providing a Helping Hand: Metabolic Regulation of T Follicular Helper Cells and Their Association With Disease. Front Immunol 2022; 13:864949. [PMID: 35493515 PMCID: PMC9047778 DOI: 10.3389/fimmu.2022.864949] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/25/2022] [Indexed: 01/02/2023] Open
Abstract
T follicular helper (Tfh) cells provide support to B cells upon arrival in the germinal center, and thus are critical for the generation of a robust adaptive immune response. Tfh express specific transcription factors and cellular receptors including Bcl6, CXCR5, PD-1, and ICOS, which are critical for homing and overall function. Generally, the induction of an immune response is tightly regulated. However, deviation during this process can result in harmful autoimmunity or the inability to successfully clear pathogens. Recently, it has been shown that Tfh differentiation, activation, and proliferation may be linked with the cellular metabolic state. In this review we will highlight recent discoveries in Tfh differentiation and explore how these cells contribute to functional immunity in disease, including autoimmune-related disorders, cancer, and of particular emphasis, during infection.
Collapse
Affiliation(s)
| | | | | | - Chih-Hao Chang
- The Jackson Laboratory, Bar Harbor, ME, United States
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
- *Correspondence: Chih-Hao Chang,
| |
Collapse
|
73
|
Osman HM, Omar GM, Elameen NF, Abdel-Nasser AM. CCL21 and IP10 as serum biomarkers for pulmonary involvement in systemic lupus erythematosus. Lupus 2022; 31:706-715. [PMID: 35380893 DOI: 10.1177/09612033221093493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Although the significance of inflammatory cytokines and chemokines in the pathogenesis of SLE is well established, the findings showed diversity and implied that combining different biomarkers could be useful in monitoring disease activity or organ involvement. Despite the potentially high prevalence of lung involvement in SLE, only a few studies have investigated for lung biomarkers. OBJECTIVE The aim of this study was to assess the value of Chemokine Ligand 21 (CCL 21) and Interferon gamma-induced protein 10 (IP10) as serum biomarkers for pulmonary involvement in SLE and their correlation with disease activity, organ involvement, pulmonary function tests (PFTs), and chest CT findings. MATERIALS AND METHODS Sixty SLE patients and 30 age- and sex-matched controls were enrolled into this study. All patients underwent serological tests, PFTs, and chest CT examination. The serum levels of CCL21 and IP10 were analyzed, and their correlations with PFTs and CT were explored. RESULTS SLE patients with pulmonary involvement had higher serum CCL21 and IP10 levels compared to those without pulmonary involvement which in turn had higher levels than the controls. There were strong negative correlations between CCL21 and IP10 and FEV1, FVC, and DLCO. There were also strong correlations between both biomarkers and HRCT and pulmonary damage, but no correlation with other disease manifestations. Serum level of 2095 pg/mL for CCL21 and 7185 pg/mL for IP10 could detect pulmonary involvement in SLE with a sensitivity of 83.7% and a specificity of 94.1%. Both biomarkers performed equally well in detecting SLE pulmonary involvement with a strong agreement between them (κ = 0.86, p < .001), but CCL21 was better correlated with PFT abnormalities. CONCLUSION Both CCL21 and IP10 are serum biomarkers to detect pulmonary involvement in SLE with high sensitivity and specificity. CCL21 correlates better with PFT abnormalities.
Collapse
Affiliation(s)
- Haidy M Osman
- Department of Rheumatology and Rehabilitation, 68877Minia University, El Minia, Egypt
| | - Gihan M Omar
- Department of Rheumatology and Rehabilitation, 68877Minia University, El Minia, Egypt
| | - Nadia F Elameen
- Department of Radiology, 68877Minia University, El Minia, Egypt
| | - Ahmed M Abdel-Nasser
- Department of Rheumatology and Rehabilitation, 68877Minia University, El Minia, Egypt
| |
Collapse
|
74
|
CCR7 expression in CD19 chimeric antigen receptor-engineered natural killer cells improves migration toward CCL19-expressing lymphoma cells and increases tumor control in mice with human lymphoma. Cytotherapy 2022; 24:827-834. [DOI: 10.1016/j.jcyt.2022.02.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/10/2022] [Accepted: 02/13/2022] [Indexed: 12/11/2022]
|
75
|
Huang Y, Zhou L, Zhang H, Zhang L, Xi X, Sun Y. BMDCs induce the generation of the CD103+CD8+ tissue-resident memory T cell subtype, which amplifies local tumor control in the genital tract. Cell Immunol 2022; 374:104502. [DOI: 10.1016/j.cellimm.2022.104502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 02/21/2022] [Accepted: 03/06/2022] [Indexed: 12/15/2022]
|
76
|
Li K, Lai C, Liu C, Li Z, Guo K, Xu K. WGCNA and molecular docking reveal key hub genes and potential natural inhibitor in interstitial cystitis/bladder pain syndrome. Int Urogynecol J 2022; 33:2241-2249. [PMID: 35333927 DOI: 10.1007/s00192-022-05113-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/27/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION AND HYPOTHESIS The etiology and treatment of interstitial cystitis/bladder pain syndrome are still controversial. The purpose of this study is to determine the key genes and specific regulatory pathways related to it and to find potential drug-active components through integrated bioinformatics. METHODS The data set GSE11783 was downloaded from GEO database. The modules significantly related to interstitial cystitis/bladder pain syndrome were identified by weighted correlation network analysis. The genes in the key modules were analyzed by functional enrichment and protein interaction by Cytoscape software, and finally the core hub genes were screened. Furthermore, the molecular docking verification of active components and key proteins was carried out by using AutoDock Vin software. RESULTS Among the 14 modules derived from WGCNA, turquoise module had the highest correlation with IC/BPS (r = 0.85, P < 0.001). The genes in the module were mainly enriched in the biological processes such as the interaction between cytokines and cytokine receptors and chemokine signaling pathway. The genes in the related modules of differentially expressed genes and WGCNA traits were intersected to obtain the core hub genes. Protein-protein interaction network analysis showed that the key genes were upregulated genes CCR7 and CCL19. In terms of molecular docking, triptolide, the active component in the traditional anti-inflammatory drug Tripterygium wilfordii, can form effective molecular binding with both core hub genes. CONCLUSIONS Our study identified the core hub genes CCR7 and CCL19, which acted as essential components in interstitial cystitis/bladder pain syndrome. Furthermore, CCR7 and CCL19 can form effective binding with triptolide, which will provide new insights into the development of new therapies for interstitial cystitis/bladder pain syndrome.
Collapse
Affiliation(s)
- Kuiqing Li
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yan Jiang West Road, Guangzhou, 510120, China
| | - Cong Lai
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yan Jiang West Road, Guangzhou, 510120, China
| | - Cheng Liu
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yan Jiang West Road, Guangzhou, 510120, China
| | - Zhuohang Li
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yan Jiang West Road, Guangzhou, 510120, China
| | - Kaixuan Guo
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yan Jiang West Road, Guangzhou, 510120, China
| | - Kewei Xu
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yan Jiang West Road, Guangzhou, 510120, China.
| |
Collapse
|
77
|
Abstract
CAR-T cell therapy has been heralded as a breakthrough in the field of immunotherapy, but to date, this success has been limited to hematological malignancies. By harnessing the chemokine system and taking into consideration the chemokine expression profile in the tumor microenvironment, CAR-T cells may be homed into tumors to facilitate direct tumor cell cytolysis and overcome a major hurdle in generating effective CAR-T cell responses to solid cancers.
Collapse
Affiliation(s)
- Jade Foeng
- Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
- Carina Biotech, Innovation and Collaboration Centre, The University of South Australia, Adelaide, SA 5000, Australia
| | - Iain Comerford
- Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Shaun R. McColl
- Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
- Carina Biotech, Innovation and Collaboration Centre, The University of South Australia, Adelaide, SA 5000, Australia
- Corresponding author
| |
Collapse
|
78
|
Hong W, Yang B, He Q, Wang J, Weng Q. New Insights of CCR7 Signaling in Dendritic Cell Migration and Inflammatory Diseases. Front Pharmacol 2022; 13:841687. [PMID: 35281921 PMCID: PMC8914285 DOI: 10.3389/fphar.2022.841687] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/07/2022] [Indexed: 12/14/2022] Open
Abstract
CCR7, collaborated with its ligands CCL19 and CCL21, controls extensive migratory events in the immune system. CCR7-bearing dendritic cells can swarm into T-cell zones in lymph nodes, initiating the antigen presentation and T-cell response. Abnormal expression of CCR7 in dendritic cells will cause a series of inflammatory diseases due to the chaotic dendritic cell trafficking. In this review, we take an in-depth look at the structural–functional domains of CCR7 and CCR7-bearing dendritic cell trajectory to lymph nodes. Then, we summarize the regulatory network of CCR7, including transcriptional regulation, translational and posttranslational regulation, internalization, desensitization, and recycling. Furthermore, the potential strategies of targeting the CCR7 network to regulate dendritic cell migration and to deal with inflammatory diseases are integrated, which not only emphasizes the possibility of CCR7 to be a potential target of immunotherapy but also has an implication on the homing of dendritic cells to benefit inflammatory diseases.
Collapse
Affiliation(s)
- Wenxiang Hong
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Bo Yang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Jiajia Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- *Correspondence: Qinjie Weng, ; Jiajia Wang,
| | - Qinjie Weng
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Qinjie Weng, ; Jiajia Wang,
| |
Collapse
|
79
|
C-C Chemokine Receptor 7 in Cancer. Cells 2022; 11:cells11040656. [PMID: 35203305 PMCID: PMC8870371 DOI: 10.3390/cells11040656] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/01/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
C-C chemokine receptor 7 (CCR7) was one of the first two chemokine receptors that were found to be upregulated in breast cancers. Chemokine receptors promote chemotaxis of cells and tissue organization. Since under homeostatic conditions, CCR7 promotes migration of immune cells to lymph nodes, questions immediately arose regarding the ability of CCR7 to direct migration of cancer cells to lymph nodes. The literature since 2000 was examined to determine to what extent the expression of CCR7 in malignant tumors promoted migration to the lymph nodes. The data indicated that in different cancers, CCR7 plays distinct roles in directing cells to lymph nodes, the skin or to the central nervous system. In certain tumors, it may even serve a protective role. Future studies should focus on defining mechanisms that differentially regulate the unfavorable or beneficial role that CCR7 plays in cancer pathophysiology, to be able to improve outcomes in patients who harbor CCR7-positive cancers.
Collapse
|
80
|
Chen H, Zhang Z, Zhou L, Cai T, Liu B, Wang L, Yang J. Identification of CCL19 as a Novel Immune-Related Biomarker in Diabetic Nephropathy. Front Genet 2022; 13:830437. [PMID: 35222545 PMCID: PMC8864156 DOI: 10.3389/fgene.2022.830437] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/24/2022] [Indexed: 01/07/2023] Open
Abstract
Diabetic nephropathy (DN) is one of the major microvascular complications in diabetic patients and the leading cause of end-stage renal disease (ESRD). Previous studies found that immune-related genes and immune cell infiltration play important roles in the pathogenesis and development of DN. Therefore, this study aimed to explore immune-related biomarkers in DN. In this research, three microarray datasets that included 18 DN and 28 healthy tubule samples were downloaded and integrated as the training set to identify differentially expressed immune-related genes (DEIGs). A total of 63 DEIGs were identified, and most upregulated DEIGs were primarily involved in the inflammatory response and chemokine-mediated signaling pathways. The Microenvironment Cell Populations-counter (MCP-counter) algorithm was then used to estimate the abundance of infiltrated immune and stromal cell populations. According to DEIG, weighted gene coexpression network and protein–protein network analyses, CCL19 was identified as the hub immune-related biomarker. Moreover, the upregulated level of CCL19 was confirmed in other independent datasets as well as in in vitro experiments with high glucose. In summary, this study provides novel insights into the pathogenesis of diabetic nephropathy and identifies CCL19 as a potential critical gene of DN.
Collapse
Affiliation(s)
- Hanzhi Chen
- Center for Kidney Disease, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Nephrology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Zhijian Zhang
- Department of Nephrology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Leting Zhou
- Department of Nephrology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Ting Cai
- Department of Nephrology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Bin Liu
- Department of Nephrology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Liang Wang
- Department of Nephrology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
- *Correspondence: Liang Wang, ; Junwei Yang,
| | - Junwei Yang
- Center for Kidney Disease, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Liang Wang, ; Junwei Yang,
| |
Collapse
|
81
|
The Significance of the Alter miR let-7a and miR-335 Expression Level Regulating the CCR7/CCL19 Axis as Potential Biomarkers of Tumor Progression in NSCLC. J Clin Med 2022; 11:jcm11030655. [PMID: 35160116 PMCID: PMC8836798 DOI: 10.3390/jcm11030655] [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: 11/26/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
The chemokine receptor 7/C-C ligand 19 chemokine (CCR7/CCL19) has been implicated in the development and progression of NSCLC. Its expression is regulated by various epigenetic factors including miRNAs. The aim of this study was to assess the expression of CCR7/CCL19 in cancer tissue in relation to that of miRNAs (miR-let-7a, miR-335) as transcriptional regulators. The expression of the tested miRNAs was also evaluated in serum exosomes. Sixty patients (n = 60) were enrolled in the study. The total expression of the studied mRNA and miRNAs were evaluated using qPCR. Tumor tissue fragments, macroscopically unchanged adjacent tissue, and serum were used as controls. Higher CCR7 and CCL19 mRNA expression levels were observed in tumor tissue compared to control. According to stages of the disease (AJCC tumor staging), the greatest expression level of the studied genes' mRNA was observed in patients with stage III. In NSCLC patients, lower miR let-7a expression level was observed in tumor tissue compared to serum; however, miR-335 expression level was higher (p < 0.05). The expression level of miR-335 positively correlated with tumor size (T features according to pTNM staging) and AJCC tumor staging, while miR let-7a had a negative correlation (p > 0.05) with liquid biopsy. Significantly greater miR-335 expression level and lower miR let-7a expression level in serum were observed in patients with metastases to lymph nodes. Our findings reveal a significant correlation between the expression levels of the mRNA of the studied genes and miRNAs. Changes in miR-335 and miR let-7a expression levels in the serum exosomes of NSCLC patients in relation to lymph node metastases and tumor stage may serve as a non-invasive molecular biomarker of tumor progression.
Collapse
|
82
|
Jasso GJ, Jaiswal A, Varma M, Laszewski T, Grauel A, Omar A, Silva N, Dranoff G, Porter JA, Mansfield K, Cremasco V, Regev A, Xavier RJ, Graham DB. Colon stroma mediates an inflammation-driven fibroblastic response controlling matrix remodeling and healing. PLoS Biol 2022; 20:e3001532. [PMID: 35085231 PMCID: PMC8824371 DOI: 10.1371/journal.pbio.3001532] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 02/08/2022] [Accepted: 01/07/2022] [Indexed: 12/22/2022] Open
Abstract
Chronic inflammation is often associated with the development of tissue fibrosis, but how mesenchymal cell responses dictate pathological fibrosis versus resolution and healing remains unclear. Defining stromal heterogeneity and identifying molecular circuits driving extracellular matrix deposition and remodeling stands to illuminate the relationship between inflammation, fibrosis, and healing. We performed single-cell RNA-sequencing of colon-derived stromal cells and identified distinct classes of fibroblasts with gene signatures that are differentially regulated by chronic inflammation, including IL-11-producing inflammatory fibroblasts. We further identify a transcriptional program associated with trans-differentiation of mucosa-associated fibroblasts and define a functional gene signature associated with matrix deposition and remodeling in the inflamed colon. Our analysis supports a critical role for the metalloprotease Adamdec1 at the interface between tissue remodeling and healing during colitis, demonstrating its requirement for colon epithelial integrity. These findings provide mechanistic insight into how inflammation perturbs stromal cell behaviors to drive fibroblastic responses controlling mucosal matrix remodeling and healing.
Collapse
Affiliation(s)
- Guadalupe J. Jasso
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Alok Jaiswal
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Mukund Varma
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Tyler Laszewski
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States of America
| | - Angelo Grauel
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States of America
| | - Abdifatah Omar
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Nilsa Silva
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States of America
| | - Glenn Dranoff
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States of America
| | - Jeffrey A. Porter
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States of America
| | - Keith Mansfield
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States of America
| | - Viviana Cremasco
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States of America
| | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Howard Hughes Medical Institute and David H. Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Ramnik J. Xavier
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- * E-mail: (RJX); (DBG)
| | - Daniel B. Graham
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- * E-mail: (RJX); (DBG)
| |
Collapse
|
83
|
Bandyopadhyay A, Majumdar K, Mishra V. Non-neoplastic Lesions of the Appendix. SURGICAL PATHOLOGY OF THE GASTROINTESTINAL SYSTEM 2022:481-519. [DOI: 10.1007/978-981-16-6395-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
84
|
Liu Q, Qiao M, Lohinai Z, Mao S, Pan Y, Wang Y, Yang S, Zhou F, Jiang T, Yi X, Ren S, Zhou C, Hirsch FR. CCL19 associates with lymph node metastasis and inferior prognosis in patients with small cell lung cancer. Lung Cancer 2021; 162:194-202. [PMID: 34823893 DOI: 10.1016/j.lungcan.2021.11.003] [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: 07/21/2021] [Revised: 10/25/2021] [Accepted: 11/05/2021] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Small cell lung cancer (SCLC) is a systemic disease and most patients have metastases at diagnosis. Better understanding of the underlying mechanisms of SCLC metastasis may provide potential approach to improve clinical outcome. METHODS HTG Edge-seq was used to identify the differential gene expression between primary SCLC lesions and paired metastatic lymph nodes (LN). Overall survival (OS) analysis was performed in patients with different levels of plasma CCL19 concentration. Invasion, migration, proliferation, apoptosis and angiogenesis ability of SCLC cells and function of CD8 + T cells were evaluated in vitro to investigate the mechanism of CCL19 in promoting metastasis. RESULTS Four chemokines (CCL19, CCL21, CCL8, CCR1) were the most differentially expressed between primary lesions and metastatic LN. CCL19 was further investigated because its mRNA and protein level expression were also validated in four SCLC cell lines (H446, H69, H82, H196). Higher plasma CCL19 was associated with late lymph node (N3) metastasis (training cohort P = 0.044, validation cohort P = 0.020) and shorter OS (training cohort P = 0.040, validation cohort P = 0.047) in SCLC patients. Silencing CCL19 inhibited SCLC cell migration, invasion, proliferation and HUVECs tube formation. Furthermore, we found that CCL19 could decrease percentage of CD8 + Ki67 + and CD8 + GZMB + T cells and increase proportion of CD8 + PD1 + T cells. CONCLUSION CCL19 was associated with LN metastasis and poor prognosis in patients with SCLC. Its expression promoted tumor progression and metastasis and impaired the function of CD8 + T cells, suggesting CCL19 might be a potential target for SCLC.
Collapse
Affiliation(s)
- Qian Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Lung Cancer Institute, Tongji University School of Medicine, Shanghai, China; Department of Oncology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Meng Qiao
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Lung Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Zoltan Lohinai
- National Korányi Institute of Pulmonology, Budapest, Hungary
| | - Shiqi Mao
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Lung Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yingying Pan
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Lung Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yan Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Lung Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Shuo Yang
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Lung Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Fei Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Lung Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Lung Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Xianghua Yi
- Department of Pathology, Tongji University Affiliated Tongji Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Lung Cancer Institute, Tongji University School of Medicine, Shanghai, China.
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital and Lung Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Fred R Hirsch
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Tisch Cancer Institute, Center for Thoracic Oncology, Mount Sinai Health System, New York, NY, USA
| |
Collapse
|
85
|
Hossainey MRH, Yaparla A, Hauser KA, Moore TE, Grayfer L. The Roles of Amphibian ( Xenopus laevis) Macrophages during Chronic Frog Virus 3 Infections. Viruses 2021; 13:v13112299. [PMID: 34835105 PMCID: PMC8621048 DOI: 10.3390/v13112299] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/06/2021] [Accepted: 11/16/2021] [Indexed: 12/27/2022] Open
Abstract
Infections by Frog Virus 3 (FV3) and other ranavirus genus members are significantly contributing to global amphibian decline. The Xenopus laevis frog is an ideal research platform upon which to study the roles of distinct frog leukocyte populations during FV3 infections. Frog macrophages (MΦs) are integrally involved during FV3 infection, as they facilitate viral dissemination and persistence but also participate in immune defense against this pathogen. In turn, MΦ differentiation and functionality depend on the colony-stimulating factor-1 receptor (CSF-1R), which is ligated by CSF-1 and iterleukin-34 (IL-34) cytokines. Our past work indicated that X. laevis CSF-1 and IL-34 give rise to morphologically and functionally distinct frog MΦ subsets, and that these CSF-1- and IL-34-MΦs respectively confer susceptibility and antiviral resistance to FV3. Because FV3 targets the frog kidneys and establishes chronic infections therein, presently we examined the roles of the frog CSF-1- and IL-34-MΦs in seeding and maintaining these chronic kidney infections. Our findings indicate that the frog CSF-1-MΦs result in more prominent kidney FV3 infections, which develop into greater reservoirs of lingering FV3 marked by infiltrating leukocytes, fibrosis, and overall immunosuppressive states. Moreover, the antiviral effects of IL-34-MΦs are short-lived and are lost as FV3 infections progress.
Collapse
|
86
|
Cuesta-Mateos C, Terrón F, Herling M. CCR7 in Blood Cancers - Review of Its Pathophysiological Roles and the Potential as a Therapeutic Target. Front Oncol 2021; 11:736758. [PMID: 34778050 PMCID: PMC8589249 DOI: 10.3389/fonc.2021.736758] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/12/2021] [Indexed: 11/23/2022] Open
Abstract
According to the classical paradigm, CCR7 is a homing chemokine receptor that grants normal lymphocytes access to secondary lymphoid tissues such as lymph nodes or spleen. As such, in most lymphoproliferative disorders, CCR7 expression correlates with nodal or spleen involvement. Nonetheless, recent evidence suggests that CCR7 is more than a facilitator of lymphatic spread of tumor cells. Here, we review published data to catalogue CCR7 expression across blood cancers and appraise which classical and novel roles are attributed to this receptor in the pathogenesis of specific hematologic neoplasms. We outline why novel therapeutic strategies targeting CCR7 might provide clinical benefits to patients with CCR7-positive hematopoietic tumors.
Collapse
Affiliation(s)
- Carlos Cuesta-Mateos
- Immunology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria- Instituto la Princesa (IIS-IP), Madrid, Spain.,Immunological and Medicinal Products (IMMED S.L.), Madrid, Spain.,Catapult Therapeutics BV, Lelystad, Netherlands
| | - Fernando Terrón
- Immunological and Medicinal Products (IMMED S.L.), Madrid, Spain.,Catapult Therapeutics BV, Lelystad, Netherlands
| | - Marco Herling
- Clinic of Hematology and Cellular Therapy, University of Leipzig, Leipzig, Germany
| |
Collapse
|
87
|
Cuevas VD, Simón-Fuentes M, Orta-Zavalza E, Samaniego R, Sánchez-Mateos P, Escribese M, Cimas FJ, Bustos M, Pérez-Diego M, Ocaña A, Domínguez-Soto Á, Vega MA, Corbí ÁL. The Gene Signature of Activated M-CSF-Primed Human Monocyte-Derived Macrophages Is IL-10-Dependent. J Innate Immun 2021; 14:243-256. [PMID: 34670213 DOI: 10.1159/000519305] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 08/30/2021] [Indexed: 11/19/2022] Open
Abstract
During inflammatory responses, monocytes are recruited into inflamed tissues, where they become monocyte-derived macrophages and acquire pro-inflammatory and tissue-damaging effects in response to the surrounding environment. In fact, monocyte-derived macrophage subsets are major pathogenic cells in inflammatory pathologies. Strikingly, the transcriptome of pathogenic monocyte-derived macrophage subsets resembles the gene profile of macrophage colony-stimulating factor (M-CSF)-primed monocyte-derived human macrophages (M-MØ). As M-MØ display a characteristic cytokine profile after activation (IL10high TNFlow IL23low IL6low), we sought to determine the transcriptional signature of M-MØ upon exposure to pathogenic stimuli. Activation of M-MØ led to the acquisition of a distinctive transcriptional profile characterized by the induction of a group of genes (Gene set 1) highly expressed by pathogenic monocyte-derived macrophages in COVID-19 and whose presence in tumor-associated macrophages (TAM) correlates with the expression of macrophage-specific markers (CD163, SPI1) and IL10. Indeed, Gene set 1 expression was primarily dependent on ERK/p38 and STAT3 activation, and transcriptional analysis and neutralization experiments revealed that IL-10 is not only required for the expression of a subset of genes within Gene set 1 but also significantly contributes to the idiosyncratic gene signature of activated M-MØ. Our results indicate that activation of M-CSF-dependent monocyte-derived macrophages induces a distinctive gene expression profile, which is partially dependent on IL-10, and identifies a gene set potentially helpful for macrophage-centered therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Rafael Samaniego
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Laboratorio de Inmuno-Oncología, Madrid, Spain
| | - Paloma Sánchez-Mateos
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Laboratorio de Inmuno-Oncología, Madrid, Spain
| | - María Escribese
- Institute for Applied Molecular Medicine, School of Medicine, Universidad CEU San Pablo, Madrid, Spain
| | - Francisco J Cimas
- Instituto de Investigación Sanitaria (IdISSC) and CIBERONC, Medical Oncology Department, Experimental Therapeutics Unit, Hospital Clínico San Carlos (HCSC), Madrid, Spain
| | - Matilde Bustos
- Institute of Biomedicine in Seville (IBiS), Campus del Hospital "Virgen del Rocío", Sevilla, Spain
| | | | - Alberto Ocaña
- Instituto de Investigación Sanitaria (IdISSC) and CIBERONC, Medical Oncology Department, Experimental Therapeutics Unit, Hospital Clínico San Carlos (HCSC), Madrid, Spain
| | | | - Miguel A Vega
- Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Ángel L Corbí
- Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| |
Collapse
|
88
|
Iida Y, Yoshikawa R, Murata A, Kotani H, Kazuki Y, Oshimura M, Matsuzaki Y, Harada M. Local injection of CCL19-expressing mesenchymal stem cells augments the therapeutic efficacy of anti-PD-L1 antibody by promoting infiltration of immune cells. J Immunother Cancer 2021; 8:jitc-2020-000582. [PMID: 32675195 PMCID: PMC7368496 DOI: 10.1136/jitc-2020-000582] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2020] [Indexed: 12/12/2022] Open
Abstract
Background Mesenchymal stem/stromal cells (MSC) accumulate and reside in tumor sites. Methods Taking advantage of this feature in anticancer therapy, immortalized murine MSC (iMSC) were genetically altered to produce chemokine (C-C motif) ligand 19 (iMSC/CCL19), which attracts dendritic cells (DC) and T lymphocytes. Thereafter, iMSC/CCL19 were examined for their therapeutic efficacy using a syngeneic CT26 colon carcinoma cell line. Results Co-injection of iMSC/CCL19 into mice significantly suppressed the in vivo growth of CT26 cells compared with that of CCL19-expressing immortalized fibroblasts (iFib/CCL19). This anticancer effect was not observed when injected in CT26-bearing nude mice. Co-injected iMSC/CCL19 survived longer than iFib/CCL19 in the tumor sites. In a therapeutic model, local injection of iMSC/CCL19 suppressed the tumor growth, and increased IFN (interferon)-γ+ CD8+ T cells and CCR7+ DC infiltration in tumor site was observed when treated with iMSC/CCL19, but not with iMSC. This antitumor effect was completely negated by depletion of CD4+ cells and partially negated by depletion of CD8+ cells. Furthermore, the antitumor effects induced by local injection of iMSC/CCL19 were augmented by additional therapy with anti-programmed death (PD)-ligand 1 (PD-L1) antibody, but not with anti-PD-1 antibody. This combination therapy cured most of the tumors in CT26-bearing mice. Conclusion These results suggest that local therapy with iMSC/CCL19 can suppress tumor growth via effective recruitment of CCR7+ DC into tumor sites and increase IFN-γ+ CD8+ T cells, and that combination with anti-PD-L1 antibody therapy can be a powerful anticancer therapy.
Collapse
Affiliation(s)
- Yuichi Iida
- Immunology, Shimane University Faculty of Medicine Graduate School of Medicine, Izumo, Japan
| | - Rintaro Yoshikawa
- Life Science, Shimane University Faculty of Medicine Graduate School of Medicine, Izumo, Shimane, Japan
| | - Akihiko Murata
- Molecular and Cellular Biology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Hitoshi Kotani
- Immunology, Shimane University Faculty of Medicine Graduate School of Medicine, Izumo, Japan
| | - Yasuhiro Kazuki
- Biomedical Science, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Mitsuo Oshimura
- Chromosome Engineering Research Center, Tottori University, Yonago, Tottori, Japan
| | - Yumi Matsuzaki
- Life Science, Shimane University Faculty of Medicine Graduate School of Medicine, Izumo, Shimane, Japan
| | - Mamoru Harada
- Immunology, Shimane University Faculty of Medicine Graduate School of Medicine, Izumo, Japan
| |
Collapse
|
89
|
Combination of AAV-CCL19 and GPC3 CAR-T Cells in the Treatment of Hepatocellular Carcinoma. J Immunol Res 2021; 2021:1782728. [PMID: 34527749 PMCID: PMC8437660 DOI: 10.1155/2021/1782728] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/06/2021] [Accepted: 08/17/2021] [Indexed: 01/10/2023] Open
Abstract
Background Chimeric antigen receptor-modified T cell (CAR-T) therapy has great potential for treating malignant tumors, especially hematological malignancies. However, the therapeutic effect of solid tumors is limited. One of the most important factors is the homing of CAR-T cells to tumor tissues in vivo. Method a recombinant adeno-associated virus 2 (AAV2) subtype carrying the CCL19 gene was used to pretreat the tumor before the Glypican-3 (GPC3) CAR-T treatment. The tumor tissue continuously expressed CCL19 and analyzed the tumor-suppressive effect of AAV-CCL19 on GPC3 CAR-T by in vitro and in vivo experiments. Result Under the chemotaxis of CCL19, CAR-T cells had a significant increase in the degree of tumor tissue infiltration; also, the antitumor effect in vitro was significantly enhanced. AAV-CCL19 combined with GPC3 CAR-T significantly increased the survival time of mice. The aforementioned results showed that the combination of AAV-CCL19 and GPC3 CAR-T cells effectively increased the ability of CAR-T cells to go home into the tumor tissue, making the CAR-T cell treatment more effective. Conclusion This study is expected to solve the dilemma in treating CAR-T cell solid tumors and achieve better clinical results.
Collapse
|
90
|
Wu S, Xia Y, Hu Y, Ma G. Bio-mimic particles for the enhanced vaccinations: Lessons learnt from the natural traits and pathogenic invasion. Adv Drug Deliv Rev 2021; 176:113871. [PMID: 34311014 DOI: 10.1016/j.addr.2021.113871] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/30/2021] [Accepted: 07/11/2021] [Indexed: 12/21/2022]
Abstract
In the combat against pathogens, the immune systems were evolved with the immune recognitions against the various danger signals, which responded vigorously upon the pathogen invasions and elicited potent antibodies or T cell engagement against the re-infections. Envisage with the prevailing pandemics and increasing demands for cancer vaccines, bio-mimic particles were developed to imitate the natural traits of the pathogens, which conferred the optimal strategies to stimulate the immune engagement and let to the increased vaccine efficacy. Here, the recent development in bio-mimic particles, as well as the natural cues from the pathogens were discussed. As such, the designing principles that adapted from the physiochemical properties of the pathogens were unfolded as the surface characteristics (hydrophobic, nano-pattern, antigen display, charge), properties (size, shape, softness) and the delivered components (peptide, protein, nuclear acids, toll-like receptor (TLR) agonist, antibody). Additionally, the strategies for the efficient delivery, regarding the biodistribution, internalization and presentation of the antigens were also illustrated. Through reviewing the state-of-art in biomimetic particles, the lesson learnt from the natural traits and pathogenic invasion may shed light on the rational design for the enhanced vaccinations.
Collapse
|
91
|
Andreev-Andrievskiy AA, Zinovkin RA, Mashkin MA, Frolova OY, Kazaishvili YG, Scherbakova VS, Rudoy BA, Nesterenko VG. Gene Expression Pattern of Peyer's Patch Lymphocytes Exposed to Kagocel Suggests Pattern-Recognition Receptors Mediate Its Action. Front Pharmacol 2021; 12:679511. [PMID: 34413772 PMCID: PMC8369352 DOI: 10.3389/fphar.2021.679511] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 07/16/2021] [Indexed: 12/21/2022] Open
Abstract
Kagocel is a synthetic carboxymethylcellulose derivative copolymerized with gossypol. Clinical data evidence its safety and efficiency for the treatment of flu and other viral infections via enhancement of interferon production. The gut-associated lymphoid tissue seems a likely site of kagocel action. The study was aimed to investigate the molecular mechanisms of its action using murine Peyer’s patches lymphocytes as a test system and the cytokines production and gene expression patterns as the primary outcomes. The Peyer’s patches lymphocytes isolated from BALB/c mice were stimulated with concanavalin A, or, to mimic viral infection, with a combination of concanavalin A and TLR3 ligand poly I:C. After 24 h of stimulation the cells were treated with saline, 30, 100, or 300 μg/ml of kagocel, or, as positive controls, 300 μg/ml oats b-D-glucan or 300 μg/ml lentinan. After 24 and 72 h of incubation with these drugs cytokines production was analyzed with ELISA and gene expression pattern was investigated using nCounter Inflammation panel chips followed by bioinformatics analysis. Expression of genes involved in the inflammatory response, antiviral defense, lymphocytes survival and proliferation (C1qa, C2, C3, Ccl21a, Il11, Il1b, Il23a, Il5, Ltb4r2, Alox15, Pla2g4a, Ptger1, Mapkapk5, Hras, Ifna1, Tlr2, Mrc1, Mx2) was upregulated in kagocel-treated Peyer’s patches lymphocytes. A list of plausible transcription factors (CEBPs, IRF, NFκB, RXR, Stat, Tead4, and ZSCAN) and master-regulators has been identified (cIAP, CIKS, dock9, MEKK1, FXR, IKK, IRAK, TRAF, dsRNA:TLR3:TRIF). The changes in gene expression pattern and the outcomes of bioinformatics analysis suggest that pattern recognition receptors, TLRs and dectin-1, are the key mediators of kagocel immunomodulatory action, with the possible involvement of interferon autocrine loop. The genes upregulated with kagocel include diverse components of the innate immune defense system.
Collapse
Affiliation(s)
- Alexander A Andreev-Andrievskiy
- Biology Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia.,MSU Institute for Mitoengineering, Moscow, Russia.,Institute of Biomedical Problems RAS, Moscow, Russia
| | - Roman A Zinovkin
- MSU Institute for Mitoengineering, Moscow, Russia.,A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Mikhail A Mashkin
- MSU Institute for Mitoengineering, Moscow, Russia.,Institute of Biomedical Problems RAS, Moscow, Russia
| | | | - Yuriy G Kazaishvili
- N.F. Gamaleya Federal Research Centre of Epidemiology and Microbiology, Moscow, Russia
| | | | - Boris A Rudoy
- N.F. Gamaleya Federal Research Centre of Epidemiology and Microbiology, Moscow, Russia
| | - Vladimir G Nesterenko
- N.F. Gamaleya Federal Research Centre of Epidemiology and Microbiology, Moscow, Russia
| |
Collapse
|
92
|
Li R, Cheng L, Wang Q, Zhou L. Comparative Transcriptomic Analysis Reveals the Immunosuppressive Targets of Mesalazine in Dextran Sulfate Sodium-Induced Ulcerative Colitis. Front Genet 2021; 12:698983. [PMID: 34456974 PMCID: PMC8386351 DOI: 10.3389/fgene.2021.698983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/03/2021] [Indexed: 11/30/2022] Open
Abstract
Ulcerative colitis (UC) is a complex inflammatory bowel disorder that can induce colonic and rectal dysfunction. Mesalazine, a first-line medicine, is routinely prescribed for UC treatment. However, the pharmacological targets of mesalazine against UC are not detailed in current publications. In the current study, a transcriptomics strategy was applied to reveal the therapeutic targets and molecular mechanisms of mesalazine for treating dextran sulfate sodium (DSS)-induced UC in mice. Compared with the UC group, a total of 1,663 differentially expressed genes were identified in mesalazine-treated mice, of which 262 were upregulated and 1,401 were downregulated. GO and KEGG enrichment analyses indicated that the protective actions of mesalazine for treating UC were related to the functional regulation of immune inflammatory response, such as the regulation of T cells, white blood cells, and cytokine receptor pathways. In addition, ingenuity pathway analysis of the gene network further revealed the inhibitory action of mesalazine on C-C motif chemokine ligands (CCL11 and CCL21) and C-X-C motif chemokine ligands (CXCL3 and CXCR2). Taken together, the current transcriptomic findings revealed anti-UC pharmacological targets, including the newly discovered biotargets CCL11, CCL21, CXCL3, and CXCR2, of mesalazine against DSS-induced intestinal inflammation.
Collapse
Affiliation(s)
| | | | | | - Liming Zhou
- Department of Pharmacology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
93
|
Brandum EP, Jørgensen AS, Rosenkilde MM, Hjortø GM. Dendritic Cells and CCR7 Expression: An Important Factor for Autoimmune Diseases, Chronic Inflammation, and Cancer. Int J Mol Sci 2021; 22:ijms22158340. [PMID: 34361107 PMCID: PMC8348795 DOI: 10.3390/ijms22158340] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 12/21/2022] Open
Abstract
Chemotactic cytokines-chemokines-control immune cell migration in the process of initiation and resolution of inflammatory conditions as part of the body's defense system. Many chemokines also participate in pathological processes leading up to and exacerbating the inflammatory state characterizing chronic inflammatory diseases. In this review, we discuss the role of dendritic cells (DCs) and the central chemokine receptor CCR7 in the initiation and sustainment of selected chronic inflammatory diseases: multiple sclerosis (MS), rheumatoid arthritis (RA), and psoriasis. We revisit the binary role that CCR7 plays in combatting and progressing cancer, and we discuss how CCR7 and DCs can be harnessed for the treatment of cancer. To provide the necessary background, we review the differential roles of the natural ligands of CCR7, CCL19, and CCL21 and how they direct the mobilization of activated DCs to lymphoid organs and control the formation of associated lymphoid tissues (ALTs). We provide an overview of DC subsets and, briefly, elaborate on the different T-cell effector types generated upon DC-T cell priming. In the conclusion, we promote CCR7 as a possible target of future drugs with an antagonistic effect to reduce inflammation in chronic inflammatory diseases and an agonistic effect for boosting the reactivation of the immune system against cancer in cell-based and/or immune checkpoint inhibitor (ICI)-based anti-cancer therapy.
Collapse
|
94
|
Atalis A, Dixon JB, Roy K. Soluble and Microparticle-Based Delivery of TLR4 and TLR9 Agonists Differentially Modulate 3D Chemotaxis of Bone Marrow-Derived Dendritic Cells. Adv Healthc Mater 2021; 10:e2001899. [PMID: 33928762 PMCID: PMC9211062 DOI: 10.1002/adhm.202001899] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/12/2021] [Indexed: 12/30/2022]
Abstract
Vaccines are commonly administered subcutaneously or intramuscularly, and local immune cells, notably dendritic cells (DCs), play a significant role in transporting vaccine antigens and adjuvants to draining lymph nodes. Here, it is compared how soluble and biomaterial-mediated delivery of Toll-like receptor (TLR)-targeted adjuvants, monophosphoryl lipid A (MPLA, TLR4 ligand) and 5'-C-phosphate-G-3' DNA (CpG DNA, TLR9 ligand), modulate 3D chemotaxis of bone marrow-derived dendritic cells (BMDCs) toward lymphatic chemokine gradients. Within microfluidic devices containing 3D collagen-based matrices to mimic tissue conditions, soluble MPLA increases BMDC chemotaxis toward gradients of CCL19 and CCL21, while soluble CpG has no effect. Delivering CpG on poly(lactic-co-glycolic) acid microparticles (MPs) enhances BMDC chemotaxis compared to MPLA-encapsulated MPs, and when co-delivered, MPLA and CpG do not synergistically enhance BMDC migration. It is concluded that supplementing granulocyte-macrophage colony stimulating factor-derived BMDC culture with interleukin-4 is necessary to induce CCR7 expression and chemotaxis of BMDCs. Different cell subsets in BMDC culture upregulate CCR7 in response to soluble versus biomaterial-loaded MPLA and CpG, and CCR7 expression does not consistently correlate with functional migration. The results show both adjuvant type and delivery method influence chemotaxis of DCs, and these findings uncover new directions for the rational design of vaccine formulations.
Collapse
Affiliation(s)
- Alexandra Atalis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - J Brandon Dixon
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Krishnendu Roy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Marcus Center for Therapeutic Cell Characterization and Manufacturing (MC3M), Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
95
|
Mateu-Albero T, Juárez-Sánchez R, Loscertales J, Mol W, Terrón F, Muñoz-Calleja C, Cuesta-Mateos C. Effect of ibrutinib on CCR7 expression and functionality in chronic lymphocytic leukemia and its implication for the activity of CAP-100, a novel therapeutic anti-CCR7 antibody. Cancer Immunol Immunother 2021; 71:627-636. [PMID: 34297159 DOI: 10.1007/s00262-021-03014-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 07/08/2021] [Indexed: 02/01/2023]
Abstract
CAP-100 is a novel therapeutic antibody directed against the ligand binding site of human CCR7. This chemokine receptor is overexpressed in chronic lymphocytic leukemia (CLL) and orchestrates the homing of CLL cells into the lymph node. Previous studies, on a very limited number of samples, hypothesized that the Bruton's tyrosine kinase inhibitor (BTKi) ibrutinib might induce loss of surface CCR7 levels in CLL cells. CAP-100 will be evaluated in clinical trials as a therapy for relapse/refractory CLL patients, who have received at least two systemic therapies (NCT04704323). As nowadays many relapse/refractory CLL patients will have received ibrutinib as a prior therapy, we aimed to investigate in a large cohort of CLL patients the impact of this BTKi on CCR7 expression and functionality as well as on the therapeutic activity of CAP-100. Our data confirm that ibrutinib moderately down-regulates the very high expression of CCR7 in CLL cells but has no apparent effect on CCR7-induced chemotaxis. Moreover, CLL cells are perfectly targetable by CAP-100 which led to a complete inhibition of CCR7-mediated migration and induced strong target cell killing through antibody-dependent cell-mediated cytotoxicity, irrespective of previous or contemporary ibrutinib administration. Together, these results validate the therapeutic utility of CAP-100 as a next-line single-agent therapy for CLL patients who failed to ibrutinib and confirm that CAP-100 and ibrutinib have complementary non-overlapping mechanisms of action, potentially allowing for combination therapy.
Collapse
Affiliation(s)
- Tamara Mateu-Albero
- Immunology Department, Hospital Universitario de La Princesa, IIS-IP, Diego de León 62, 28006, Madrid, Spain
| | - Raquel Juárez-Sánchez
- Immunology Department, Hospital Universitario de La Princesa, IIS-IP, Diego de León 62, 28006, Madrid, Spain.,IMMED S.L., Immunological and Medicinal Products, C/ Velázquez 57, 6º derecha, 28001, Madrid, Spain
| | - Javier Loscertales
- Hematology Department, Hospital Universitario de La Princesa, IIS-IP, Diego de León 62, 28006, Madrid, Spain
| | - Wim Mol
- Catapult Therapeutics, Lelystad, The Netherlands
| | - Fernando Terrón
- IMMED S.L., Immunological and Medicinal Products, C/ Velázquez 57, 6º derecha, 28001, Madrid, Spain.,Catapult Therapeutics, Lelystad, The Netherlands
| | - Cecilia Muñoz-Calleja
- Immunology Department, Hospital Universitario de La Princesa, IIS-IP, Diego de León 62, 28006, Madrid, Spain.,Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlos Cuesta-Mateos
- Immunology Department, Hospital Universitario de La Princesa, IIS-IP, Diego de León 62, 28006, Madrid, Spain. .,IMMED S.L., Immunological and Medicinal Products, C/ Velázquez 57, 6º derecha, 28001, Madrid, Spain. .,Catapult Therapeutics, Lelystad, The Netherlands.
| |
Collapse
|
96
|
Prokop JW, Hartog NL, Chesla D, Faber W, Love CP, Karam R, Abualkheir N, Feldmann B, Teng L, McBride T, Leimanis ML, English BK, Holsworth A, Frisch A, Bauss J, Kalpage N, Derbedrossian A, Pinti RM, Hale N, Mills J, Eby A, VanSickle EA, Pageau SC, Shankar R, Chen B, Carcillo JA, Sanfilippo D, Olivero R, Bupp CP, Rajasekaran S. High-Density Blood Transcriptomics Reveals Precision Immune Signatures of SARS-CoV-2 Infection in Hospitalized Individuals. Front Immunol 2021; 12:694243. [PMID: 34335605 PMCID: PMC8322982 DOI: 10.3389/fimmu.2021.694243] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/30/2021] [Indexed: 12/27/2022] Open
Abstract
The immune response to COVID-19 infection is variable. How COVID-19 influences clinical outcomes in hospitalized patients needs to be understood through readily obtainable biological materials, such as blood. We hypothesized that a high-density analysis of host (and pathogen) blood RNA in hospitalized patients with SARS-CoV-2 would provide mechanistic insights into the heterogeneity of response amongst COVID-19 patients when combined with advanced multidimensional bioinformatics for RNA. We enrolled 36 hospitalized COVID-19 patients (11 died) and 15 controls, collecting 74 blood PAXgene RNA tubes at multiple timepoints, one early and in 23 patients after treatment with various therapies. Total RNAseq was performed at high-density, with >160 million paired-end, 150 base pair reads per sample, representing the most sequenced bases per sample for any publicly deposited blood PAXgene tube study. There are 770 genes significantly altered in the blood of COVID-19 patients associated with antiviral defense, mitotic cell cycle, type I interferon signaling, and severe viral infections. Immune genes activated include those associated with neutrophil mechanisms, secretory granules, and neutrophil extracellular traps (NETs), along with decreased gene expression in lymphocytes and clonal expansion of the acquired immune response. Therapies such as convalescent serum and dexamethasone reduced many of the blood expression signatures of COVID-19. Severely ill or deceased patients are marked by various secondary infections, unique gene patterns, dysregulated innate response, and peripheral organ damage not otherwise found in the cohort. High-density transcriptomic data offers shared gene expression signatures, providing unique insights into the immune system and individualized signatures of patients that could be used to understand the patient's clinical condition. Whole blood transcriptomics provides patient-level insights for immune activation, immune repertoire, and secondary infections that can further guide precision treatment.
Collapse
Affiliation(s)
- Jeremy W. Prokop
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Nicholas L. Hartog
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Allergy & Immunology, Spectrum Health, Grand Rapids, MI, United States
| | - Dave Chesla
- Office of Research, Spectrum Health, Grand Rapids, MI, United States
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - William Faber
- Physical Sciences, Grand Rapids Community College, Grand Rapids, MI, United States
| | - Chanise P. Love
- Office of Research, Spectrum Health, Grand Rapids, MI, United States
| | | | | | | | - Li Teng
- Ambry Genetics, Aliso Viejo, CA, United States
| | | | - Mara L. Leimanis
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Pediatric Intensive Care Unit, Helen DeVos Children’s Hospital, Grand Rapids, MI, United States
| | - B. Keith English
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Amanda Holsworth
- Allergy & Immunology, Spectrum Health, Grand Rapids, MI, United States
| | - Austin Frisch
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Jacob Bauss
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Nathisha Kalpage
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Aram Derbedrossian
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Ryan M. Pinti
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Nicole Hale
- The Department of Chemistry and Biochemistry, Calvin University, Grand Rapids, MI, United States
| | - Joshua Mills
- Department of Biology, Grand Valley State University, Allendale, MI, United States
| | - Alexandra Eby
- Department of Science, Davenport University, Grand Rapids, MI, United States
| | | | - Spencer C. Pageau
- Office of Research, Spectrum Health, Grand Rapids, MI, United States
| | - Rama Shankar
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Bin Chen
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Joseph A. Carcillo
- Department of Critical Care Medicine and Pediatrics, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Dominic Sanfilippo
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Pediatric Intensive Care Unit, Helen DeVos Children’s Hospital, Grand Rapids, MI, United States
| | - Rosemary Olivero
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Infectious Disease, Helen DeVos Children’s Hospital, Grand Rapids, MI, United States
| | - Caleb P. Bupp
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Medical Genetics, Spectrum Health Medical Genetics, Grand Rapids, MI, United States
| | - Surender Rajasekaran
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Office of Research, Spectrum Health, Grand Rapids, MI, United States
- Pediatric Intensive Care Unit, Helen DeVos Children’s Hospital, Grand Rapids, MI, United States
| |
Collapse
|
97
|
Hornburg M, Desbois M, Lu S, Guan Y, Lo AA, Kaufman S, Elrod A, Lotstein A, DesRochers TM, Munoz-Rodriguez JL, Wang X, Giltnane J, Mayba O, Turley SJ, Bourgon R, Daemen A, Wang Y. Single-cell dissection of cellular components and interactions shaping the tumor immune phenotypes in ovarian cancer. Cancer Cell 2021; 39:928-944.e6. [PMID: 33961783 DOI: 10.1016/j.ccell.2021.04.004] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/12/2020] [Accepted: 04/06/2021] [Indexed: 01/06/2023]
Abstract
Distinct T cell infiltration patterns, i.e., immune infiltrated, excluded, and desert, result in different responses to cancer immunotherapies. However, the key determinants and biology underpinning these tumor immune phenotypes remain elusive. Here, we provide a high-resolution dissection of the entire tumor ecosystem through single-cell RNA-sequencing analysis of 15 ovarian tumors. Immune-desert tumors are characterized by unique tumor cell-intrinsic features, including metabolic pathways and low antigen presentation, and an enrichment of monocytes and immature macrophages. Immune-infiltrated and -excluded tumors differ markedly in their T cell composition and fibroblast subsets. Furthermore, our study reveals chemokine receptor-ligand interactions within and across compartments as potential mechanisms mediating immune cell infiltration, exemplified by the tumor cell-T cell cross talk via CXCL16-CXCR6 and stromal-immune cell cross talk via CXCL12/14-CXCR4. Our data highlight potential molecular mechanisms that shape the tumor immune phenotypes and may inform therapeutic strategies to improve clinical benefit from cancer immunotherapies.
Collapse
Affiliation(s)
- Milena Hornburg
- Department of Bioinformatics & Computational Biology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Mélanie Desbois
- Department of Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Shan Lu
- Department of Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Yinghui Guan
- Department of Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Amy A Lo
- Department of Research Pathology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Susan Kaufman
- Department of Biochemical Cellular Pharmacology, Genentech, Inc., South San Francisco, CA 94080, USA
| | | | | | | | | | - Xingwei Wang
- Department of Digital Pathology, Roche Tissue Diagnostics, Santa Clara, CA 95050, USA
| | - Jennifer Giltnane
- Department of Research Pathology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Oleg Mayba
- Department of Bioinformatics & Computational Biology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Shannon J Turley
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Richard Bourgon
- Department of Bioinformatics & Computational Biology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Anneleen Daemen
- Department of Bioinformatics & Computational Biology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Yulei Wang
- Department of Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA 94080, USA.
| |
Collapse
|
98
|
The Entry and Egress of Monocytes in Atherosclerosis: A Biochemical and Biomechanical Driven Process. Cardiovasc Ther 2021; 2021:6642927. [PMID: 34345249 PMCID: PMC8282391 DOI: 10.1155/2021/6642927] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
In accordance with “the response to injury” theory, the entry of monocytes into the intima guided by inflammation signals, taking up cholesterol and transforming into foam cells, and egress from plaques determines the progression of atherosclerosis. Multiple cytokines and receptors have been reported to be involved in monocyte recruitment such as CCL2/CCR2, CCL5/CCR5, and CX3CL1/CX3CR1, and the egress of macrophages from the plaque like CCR7/CCL19/CCL21. Interestingly, some neural guidance molecules such as Netrin-1 and Semaphorin 3E have been demonstrated to show an inhibitory effect on monocyte migration. During the processes of monocytes recruitment and migration, factors affecting the biomechanical properties (e.g., the membrane fluidity, the deformability, and stiffness) of the monocytes, like cholesterol, amyloid-β peptide (Aβ), and lipopolysaccharides (LPS), as well as the biomechanical environment that the monocytes are exposed, like the extracellular matrix stiffness, mechanical stretch, blood flow, and hypertension, were discussed in the latter section. Till now, several small interfering RNAs (siRNAs), monoclonal antibodies, and antagonists for CCR2 have been designed and shown promising efficiency on atherosclerosis therapy. Seeking more possible biochemical factors that are chemotactic or can affect the biomechanical properties of monocytes, and uncovering the underlying mechanism, will be helpful in future studies.
Collapse
|
99
|
Capitano ML, Jaiswal A, Broxmeyer HE, Pride Y, Glover S, Amlashi FG, Kirby A, Srinivasan G, Williamson EA, Mais D, Hromas R. A humanized monoclonal antibody against the endothelial chemokine CCL21 for the diagnosis and treatment of inflammatory bowel disease. PLoS One 2021; 16:e0252805. [PMID: 34197491 PMCID: PMC8248966 DOI: 10.1371/journal.pone.0252805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/21/2021] [Indexed: 12/31/2022] Open
Abstract
Chemokines are small proteins that promote leukocyte migration during development, infection, and inflammation. We and others isolated the unique chemokine CCL21, a potent chemo-attractant for naïve T-cells, naïve B-cells, and immature dendritic cells. CCL21 has a 37 amino acid carboxy terminal extension that is distinct from the rest of the chemokine family, which is thought to anchor it to venule endothelium where the amino terminus can interact with its cognate receptor, CCR7. We and others have reported that venule endothelium expressing CCL21 plays a crucial role in attracting naïve immune cells to sites of antigen presentation. In this study we generated a series of monoclonal antibodies to the amino terminus of CCL21 in an attempt to generate an antibody that blocked the interaction of CCL21 with its receptor CCR7. We found one humanized clone that blocked naïve T-cell migration towards CCL21, while memory effector T-cells were less affected. Using this monoclonal antibody, we also demonstrated that CCL21 is expressed in the mucosal venule endothelium of the large majority of inflammatory bowel diseases (IBD), including Crohn's disease, ulcerative colitis, and also in celiac disease. This expression correlated with active IBD in 5 of 6 cases, whereas none of 6 normal bowel biopsies had CCL21 expression. This study raises the possibility that this monoclonal antibody could be used to diagnose initial or recurrent of IBD. Significantly, this antibody could also be used for therapeutic intervention in IBD by selectively interfering with recruitment of naïve immune effector cells to sites of antigen presentation, without harming overall memory immunity.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Chemokine CCL21/genetics
- Chemokine CCL21/immunology
- Chemokine CCL21/metabolism
- Chemotaxis/drug effects
- Endothelium/metabolism
- Humans
- Inflammatory Bowel Diseases/diagnosis
- Inflammatory Bowel Diseases/drug therapy
- Inflammatory Bowel Diseases/metabolism
- Mice
- Mice, Inbred BALB C
- Protein Binding
- Receptors, CCR7/metabolism
- T-Lymphocytes, Helper-Inducer/cytology
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
Collapse
Affiliation(s)
- Maegan L Capitano
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Aruna Jaiswal
- Department of Medicine and the Mays Cancer Center, University of Texas School Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Yilianys Pride
- Division of Gastroenterology, Department of Medicine, University of Mississippi Medical Center, Jackson, MI, United States of America
| | - Sarah Glover
- Division of Gastroenterology, Department of Medicine, University of Mississippi Medical Center, Jackson, MI, United States of America
| | - Fatemah G Amlashi
- Department of Pathology, University of Texas School Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Austin Kirby
- Department of Medicine and the Mays Cancer Center, University of Texas School Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Gayathri Srinivasan
- Department of Medicine and the Mays Cancer Center, University of Texas School Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Elizabeth A Williamson
- Department of Medicine and the Mays Cancer Center, University of Texas School Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Daniel Mais
- Department of Pathology, University of Texas School Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Robert Hromas
- Department of Medicine and the Mays Cancer Center, University of Texas School Health Science Center at San Antonio, San Antonio, TX, United States of America
| |
Collapse
|
100
|
Marks KE, Flaherty S, Patterson KM, Stratton M, Martinez GJ, Reynolds JM. Toll-like receptor 2 induces pathogenicity in Th17 cells and reveals a role for IPCEF in regulating Th17 cell migration. Cell Rep 2021; 35:109303. [PMID: 34192530 PMCID: PMC8270556 DOI: 10.1016/j.celrep.2021.109303] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 04/02/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023] Open
Abstract
Pathogenic Th17 cells drive inflammation in autoimmune disease, yet the molecular programming underlying Th17 cell pathogenicity remains insufficiently understood. Activation of Toll-like receptor 2 (TLR2) increases Th17 cell inflammatory potential, but little is known regarding the mechanistic outcomes of TLR2 signaling in Th17 cells. Here, we demonstrate that TLR2 is comparable to IL-23 in inducing pathogenicity and increasing the migratory capacity of Th17 cells. We perform RNA sequencing of Th17 cells stimulated though the TLR2 pathway and find differential expression of several genes linked with the Th17 genetic program as well as genes not previously associated with pathogenic Th17 cells, including Ipcef1. Enforced expression of Ipcef1 in Th17 cells abolishes the TLR2-dependent increases in migratory capacity and severely impairs the ability of Th17 cells to induce experimental autoimmune encephalomyelitis. This study establishes the importance of the TLR2 signaling pathway in inducing Th17 cell pathogenicity and driving autoimmune inflammation.
Collapse
Affiliation(s)
- Kathryne E Marks
- Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Stephanie Flaherty
- Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Kristen M Patterson
- Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Matthew Stratton
- Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Gustavo J Martinez
- Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Joseph M Reynolds
- Center for Cancer Cell Biology, Immunology, and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA; Edward Hines Jr. VA Hospital, Hines, IL, USA.
| |
Collapse
|