101
|
Luan X, Li S, Zhao J, Zhai J, Liu X, Chen ZJ, Li W, Du Y. Down-regulation of CCR7 via AKT pathway and GATA2 inactivation suppressed trophoblast migration and invasion in recurrent spontaneous abortion†. Biol Reprod 2021; 102:424-433. [PMID: 31504210 DOI: 10.1093/biolre/ioz172] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 08/01/2019] [Accepted: 08/29/2019] [Indexed: 11/13/2022] Open
Abstract
The underlying mechanism of the chemokine-C receptor 7 (CCR7) that leads to aberrant trophoblast migration and invasion in recurrent spontaneous abortion (RSA) remains unknown. CCR7 is considered crucial for migration and invasion and has been associated with the risk of miscarriage. However, the functional role of CCR7 in RSA is not fully understood. Our study found that CCR7 mRNA and protein abundance were significantly decreased in the villous from RSA patients compared with healthy controls. Knockdown of CCR7 caused a significant reduction of migration and invasion in JAR and JEG-3 cells. Meanwhile, CCR7 functioned as a positive upstream factor of the AKT pathway contributing to the expression of GATA2, promoting trophoblast migration, and invasion via MMP2. Notably, a decreased abundance of CCR7 was positively correlated with the phosphorylation of AKT and with an abundance of GATA2 and MMP2 in human villous specimens of RSA compared with the control group. CCL19, a ligand of CCR7, could promote trophoblast migration and invasion by activating the deregulation of the CCR7-mediated pathway in RSA. We are convinced that CCR7 and its downstream factors may be possible mechanisms for the pathogenesis of RSA.
Collapse
Affiliation(s)
- Xiaorui Luan
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China.,Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shang Li
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Jun Zhao
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Junyu Zhai
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Xiaojing Liu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory for Reproductive Endocrinology of Ministry of Education, Shandong Provincial Key Laboratory of Reproductive Medicine, Center for Reproductive Medicine, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Weiping Li
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China.,Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanzhi Du
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
102
|
Systematic Assessment of Chemokine Signaling at Chemokine Receptors CCR4, CCR7 and CCR10. Int J Mol Sci 2021; 22:ijms22084232. [PMID: 33921794 PMCID: PMC8073111 DOI: 10.3390/ijms22084232] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 01/14/2023] Open
Abstract
Chemokines interact with chemokine receptors in a promiscuous network, such that each receptor can be activated by multiple chemokines. Moreover, different chemokines have been reported to preferentially activate different signalling pathways via the same receptor, a phenomenon known as biased agonism. The human CC chemokine receptors (CCRs) CCR4, CCR7 and CCR10 play important roles in T cell trafficking and have been reported to display biased agonism. To systematically characterize these effects, we analysed G protein- and β-arrestin-mediated signal transduction resulting from stimulation of these receptors by each of their cognate chemokine ligands within the same cellular background. Although the chemokines did not elicit ligand-biased agonism, the three receptors exhibited different arrays of signaling outcomes. Stimulation of CCR4 by either CC chemokine ligand 17 (CCL17) or CCL22 induced β-arrestin recruitment but not G protein-mediated signaling, suggesting that CCR4 has the potential to act as a scavenger receptor. At CCR7, both CCL19 and CCL21 stimulated G protein signaling and β-arrestin recruitment, with CCL19 consistently displaying higher potency. At CCR10, CCL27 and CCL28(4-108) stimulated both G protein signaling and β-arrestin recruitment, whereas CCL28(1-108) was inactive, suggesting that CCL28(4-108) is the biologically relevant form of this chemokine. These comparisons emphasize the intrinsic abilities of different receptors to couple with different downstream signaling pathways. Comparison of these results with previous studies indicates that differential agonism at these receptors may be highly dependent on the cellular context.
Collapse
|
103
|
Loef EJ, Sheppard HM, Birch NP, Dunbar PR. Live-Cell Microscopy Reveals That Human T Cells Primarily Respond Chemokinetically Within a CCL19 Gradient That Induces Chemotaxis in Dendritic Cells. Front Immunol 2021; 12:628090. [PMID: 33841411 PMCID: PMC8033042 DOI: 10.3389/fimmu.2021.628090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/05/2021] [Indexed: 11/17/2022] Open
Abstract
The ability to study migratory behavior of immune cells is crucial to understanding the dynamic control of the immune system. Migration induced by chemokines is often assumed to be directional (chemotaxis), yet commonly used end-point migration assays are confounded by detecting increased cell migration that lacks directionality (chemokinesis). To distinguish between chemotaxis and chemokinesis we used the classic “under-agarose assay” in combination with video-microscopy to monitor migration of CCR7+ human monocyte-derived dendritic cells and T cells in response to a concentration gradient of CCL19. Formation of the gradients was visualized with a fluorescent marker and lasted several hours. Monocyte-derived dendritic cells migrated chemotactically towards the CCL19 gradient. In contrast, T cells exhibited a biased random walk that was largely driven by increased exploratory chemokinesis towards CCL19. This dominance of chemokinesis over chemotaxis in T cells is consistent with CCR7 ligation optimizing T cell scanning of antigen-presenting cells in lymphoid tissues.
Collapse
Affiliation(s)
- Evert J Loef
- School of Biological Science, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Hilary M Sheppard
- School of Biological Science, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Nigel P Birch
- School of Biological Science, University of Auckland, Auckland, New Zealand.,Centre for Brain Research and Brain Research New Zealand, Rangahau Roro Aotearoa, University of Auckland, Auckland, New Zealand
| | - P Rod Dunbar
- School of Biological Science, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| |
Collapse
|
104
|
Cuesta-Mateos C, Brown JR, Terrón F, Muñoz-Calleja C. Of Lymph Nodes and CLL Cells: Deciphering the Role of CCR7 in the Pathogenesis of CLL and Understanding Its Potential as Therapeutic Target. Front Immunol 2021; 12:662866. [PMID: 33841445 PMCID: PMC8024566 DOI: 10.3389/fimmu.2021.662866] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/09/2021] [Indexed: 01/13/2023] Open
Abstract
The lymph node (LN) is an essential tissue for achieving effective immune responses but it is also critical in the pathogenesis of chronic lymphocytic leukemia (CLL). Within the multitude of signaling pathways aberrantly regulated in CLL the homeostatic axis composed by the chemokine receptor CCR7 and its ligands is the main driver for directing immune cells to home into the LN. In this literature review, we address the roles of CCR7 in the pathophysiology of CLL, and how this chemokine receptor is of critical importance to develop more rational and effective therapies for this malignancy.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/metabolism
- Biomarkers, Tumor
- Chemotaxis/genetics
- Chemotaxis/immunology
- Disease Susceptibility
- Gene Expression
- Humans
- Immune Tolerance
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Ligands
- Lymph Nodes/immunology
- Lymph Nodes/metabolism
- Molecular Targeted Therapy
- Protein Binding
- Receptors, CCR7/antagonists & inhibitors
- Receptors, CCR7/genetics
- Receptors, CCR7/metabolism
- Tumor Microenvironment
Collapse
Affiliation(s)
- Carlos Cuesta-Mateos
- Immunology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria- Instituto de La Princesa (IIS-IP), Madrid, Spain
- IMMED S.L., Immunological and Medicinal Products, Madrid, Spain
- Catapult Therapeutics BV, Lelystad, Netherlands
| | - Jennifer R. Brown
- Chronic Lymphocytic Leukemia (CLL) Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Fernando Terrón
- IMMED S.L., Immunological and Medicinal Products, Madrid, Spain
- Catapult Therapeutics BV, Lelystad, Netherlands
| | - Cecilia Muñoz-Calleja
- Immunology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria- Instituto de La Princesa (IIS-IP), Madrid, Spain
- School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
105
|
A comprehensive analysis of tumor microenvironment-related genes in colon cancer. Clin Transl Oncol 2021; 23:1769-1781. [PMID: 33689097 DOI: 10.1007/s12094-021-02578-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/23/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND The development and progression of colon cancer are significantly affected by the tumor microenvironment, which has attracted much attention. The goal of our study was primarily to find out all possible tumor microenvironment-related genes in colon cancer. METHOD This study quantified the immune and stromal landscape using the ESTIMATION algorithm using the gene expression matrix obtained from the UCSC Xena database. Dysregulated genes were harvested using the limma R package, and relevant pathways and biofunctions were identified using enrichment analysis. A least absolute shrinkage and selection operator (LASSO) regression was used to select the pivotal genes from the DEGs. Then, survival analysis was performed to determine the hub genes and a prognostic model was constructed by these hub genes with (or) TNM stage. Besides, associations between hub gene expressions and immune cell infiltration were assessed. RESULTS A total of 725 DEGs were identified. Most of the results of the enrichment analysis were immune-related items. 13 genes were selected as the hub genes and a moderate-to-strong positive correlation between most hub genes and several immune cells were observed. Besides, the prognostic value of the hub genes were comparable to TNM staging. CONCLUSIONS Our study provides a better understanding of how interactions between the 13 immune-prognostic hub genes and immune cells in the tumor microenvironment affect biological processes in colon cancer. These genes exhibit an equivalent ability to TNM staging in prognosis prediction. They are particularly expected to become novel prognostic biomarkers and targets of immunotherapies for colon cancer.
Collapse
|
106
|
Carnaru M, Hinchcliff M. Soluble Biomarkers for Prediction of Vascular and Gastrointestinal Disease Severity in Patients with Systemic Sclerosis. CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2021; 7:21-38. [PMID: 40270643 PMCID: PMC12017339 DOI: 10.1007/s40674-021-00171-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2021] [Indexed: 10/22/2022]
Abstract
Purpose of Review Disease severity biomarkers in patients with systemic sclerosis (SSc) provide an early and noninvasive screening tool to identify patients at increased risk for internal organ involvement that may impact diagnostic testing and treatment decisions. This review will focus on soluble SSc vascular and gastrointestinal disease biomarkers. Recent Findings Due to high morbidity and mortality associated with SSc pulmonary hypertension, multiple biomarkers are currently under investigation including serum autoantibodies, chemistries [such as N-terminal pro-brain natriuretic peptide (NT-proBNP)], proteins [midkine (MDK) and follistatin-like 3 (FSTL3)], chemokines [C-X-C motif ligand 4 (CXCL4) and C-C motif ligand 21 (CCL21)], plasma growth factors [vascular endothelial growth factor (VEGF) and placental growth factor (PlGF)], cell adhesion molecules [vascular cell adhesion molecule 1 (VCAM-1)], and endothelial microparticles [CD144+ endothelial microparticle (CD144+ EMP)]. A subset of these have also been proposed as SSc digital ulcer biomarkers [anti-endothelin-1 type A receptor (anti-ETAR), PlGF, and NT-proBNP]. A combination of NT-proBNP and high sensitivity cardiac troponins T (hs-cTnT) and I (hs-cTnI) may be useful for assessing primary SSc cardiac involvement. Putative SSc renal disease biomarkers include VEGF and endostatin levels; while anti-U1 and U3 ribonucleoprotein (anti-U1- and anti-U3-RNP) antibodies and fecal-calprotectin (F-calprotectin) are associated with GI involvement. Summary Serum autoantibodies are the mainstay SSc severity biomarkers, but new biomarkers are under investigation.
Collapse
Affiliation(s)
- Miruna Carnaru
- Yale School of Medicine, Department of Medicine, Section of Rheumatology, Allergy & Immunology, New Haven, CT 06519
| | - Monique Hinchcliff
- Yale School of Medicine, Department of Medicine, Section of Rheumatology, Allergy & Immunology, New Haven, CT 06519
| |
Collapse
|
107
|
Immunogenicity Challenges Associated with Subcutaneous Delivery of Therapeutic Proteins. BioDrugs 2021; 35:125-146. [PMID: 33523413 PMCID: PMC7848667 DOI: 10.1007/s40259-020-00465-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 12/12/2022]
Abstract
The subcutaneous route of administration has provided convenient and non-inferior delivery of therapeutic proteins compared to intravenous infusion, but there is potential for enhanced immunogenicity toward subcutaneously administered proteins in a subset of patients. Unwanted anti-drug antibody response toward proteins or monoclonal antibodies upon repeated administration is shown to impact the pharmacokinetics and efficacy of multiple biologics. Unique immunogenicity challenges of the subcutaneous route have been realized through various preclinical and clinical examples, although subcutaneous delivery has often demonstrated comparable immunogenicity to intravenous administration. Beyond route of administration as a treatment-related factor of immunogenicity, certain product-related risk factors are particularly relevant to subcutaneously administered proteins. This review attempts to provide an overview of the mechanism of immune response toward proteins administered subcutaneously (subcutaneous proteins) and comments on product-related risk factors related to protein structure and stability, dosage form, and aggregation. A two-wave mechanism of antigen presentation in the immune response toward subcutaneous proteins is described, and interaction with dynamic antigen-presenting cells possessing high antigen processing efficiency and migratory activity may drive immunogenicity. Mitigation strategies for immunogenicity are discussed, including those in general use clinically and those currently in development. Mechanistic insights along with consideration of risk factors involved inspire theoretical strategies to provide antigen-specific, long-lasting effects for maintaining the safety and efficacy of therapeutic proteins.
Collapse
|
108
|
Drakes ML, Stiff PJ. Ovarian Cancer: Therapeutic Strategies to Overcome Immune Suppression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1330:33-54. [PMID: 34339029 DOI: 10.1007/978-3-030-73359-9_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ovarian cancer generally escapes diagnosis until the advanced stages. High-grade serous ovarian cancer (HGSOC) is the most frequently occurring form of this malaise and is a disease which has the highest mortality rate of gynecologic cancers. Over recent years it has been revealed that the course of such cancers can be significantly influenced by the nature of immune cells in tumors at the time of diagnosis and by immune cells induced by therapy. Numerous investigators have since focused on disease biology to identify biomarkers or therapeutic targets. Yet, while over the past decade there have been significant improvements in state-of-the-art surgery for ovarian cancer as frontline therapy, there have been limited advancements in the development of novel curative or management drugs for this disease. This chapter discusses the major elements of immune suppression in HGSOC from a biological viewpoint, mechanisms of overcoming resistance to therapies, and recent therapy aimed at improving patient care and survival.
Collapse
Affiliation(s)
- Maureen L Drakes
- Department of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL, USA.
| | - Patrick J Stiff
- Department of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
109
|
Anderson JL, Khoury G, Fromentin R, Solomon A, Chomont N, Sinclair E, Milush JM, Hartogensis W, Bacchetti P, Roche M, Tumpach C, Gartner M, Pitman MC, Epling CL, Hoh R, Hecht FM, Somsouk M, Cameron PU, Deeks SG, Lewin SR. Human Immunodeficiency Virus (HIV)-Infected CCR6+ Rectal CD4+ T Cells and HIV Persistence On Antiretroviral Therapy. J Infect Dis 2020; 221:744-755. [PMID: 31796951 PMCID: PMC7026892 DOI: 10.1093/infdis/jiz509] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 12/02/2019] [Indexed: 02/07/2023] Open
Abstract
Background Identifying where human immunodeficiency virus (HIV) persists in people living with HIV and receiving antiretroviral therapy is critical to develop cure strategies. We assessed the relationship of HIV persistence to expression of chemokine receptors and their chemokines in blood (n = 48) and in rectal (n = 20) and lymph node (LN; n = 8) tissue collected from people living with HIV who were receiving suppressive antiretroviral therapy. Methods Cell-associated integrated HIV DNA, unspliced HIV RNA, and chemokine messenger RNA were quantified by quantitative polymerase chain reaction. Chemokine receptor expression on CD4+ T cells was determined using flow cytometry. Results Integrated HIV DNA levels in CD4+ T cells, CCR6+CXCR3+ memory CD4+ T-cell frequency, and CCL20 expression (ligand for CCR6) were highest in rectal tissue, where HIV-infected CCR6+ T cells accounted for nearly all infected cells (median, 89.7%). Conversely in LN tissue, CCR6+ T cells were infrequent, and there was a statistically significant association of cell-associated HIV DNA and RNA with CCL19, CCL21, and CXCL13 chemokines. Conclusions HIV-infected CCR6+ CD4+ T cells accounted for the majority of infected cells in rectal tissue. The different relationships between HIV persistence and T-cell subsets and chemokines in rectal and LN tissue suggest that different tissue-specific strategies may be required to eliminate HIV persistence and that assessment of biomarkers for HIV persistence may not be generalizable between blood and other tissues.
Collapse
Affiliation(s)
- Jenny L Anderson
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Gabriela Khoury
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Rémi Fromentin
- Centre de Recherche du CHUM and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Ajantha Solomon
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Nicolas Chomont
- Centre de Recherche du CHUM and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Elizabeth Sinclair
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jeffrey M Milush
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Wendy Hartogensis
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Peter Bacchetti
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Michael Roche
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia.,School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Carolin Tumpach
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia.,School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Matthew Gartner
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Matthew C Pitman
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Christine Lorrie Epling
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Rebecca Hoh
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Frederick M Hecht
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Ma Somsouk
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Paul U Cameron
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Steven G Deeks
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Sharon R Lewin
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
110
|
Li L, Xu P, Zhou Q, Xu J. The Function of T Follicular Helper Cells in the Autoimmune Liver Diseases. J Immunol Res 2020; 2020:5679254. [PMID: 33294464 PMCID: PMC7691009 DOI: 10.1155/2020/5679254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/29/2020] [Accepted: 10/31/2020] [Indexed: 11/17/2022] Open
Abstract
T follicular helper (TFH) cells are recognized as a subtype of T cells that are involved in the germinal center formation and B cell development. When dysregulated, TFH cells may represent an important mechanism that contributes to a heightened humoral response and autoantibody production in autoimmune liver diseases (AILDs). TFH cells participate in the immune response associated with AILDs by expressing surface receptors such as programmed cell death protein-1, C-X-C motif chemokine receptor 5, and inducible T cell costimulators, as well as cytokines such as interleukin-21. TFH cells also downregulate chemokine (C-C motif) receptor 7 and promote the dysregulation of the T follicular regulatory/TFH axis. This review highlights the importance of TFH cells in AILDs.
Collapse
Affiliation(s)
- Lin Li
- Department of Laboratory Medicine, First Hospital of Jilin University, Changchun 130021, China
| | - Panyang Xu
- Department of Laboratory Medicine, First Hospital of Jilin University, Changchun 130021, China
| | - Qi Zhou
- Department of Pediatrics, First Hospital of Jilin University, Changchun 130021, China
| | - Jiancheng Xu
- Department of Laboratory Medicine, First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
111
|
Huang M, Peng X, Yang L, Yang S, Li X, Tang S, Li B, Jin H, Wu B, Liu J, Li H. Non-coding RNA derived from extracellular vesicles in cancer immune escape: Biological functions and potential clinical applications. Cancer Lett 2020; 501:234-246. [PMID: 33186654 DOI: 10.1016/j.canlet.2020.11.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/23/2020] [Accepted: 11/06/2020] [Indexed: 02/07/2023]
Abstract
The tumor microenvironment represents a dynamically composed matrix into which cancer cells and many other cell types are embedded to form organ-like structures. The tumor immune microenvironment (TIME), composed of immune cells, is an inseparable part of the tumor microenvironment. Extracellular vesicles (EVs) participate in the occurrence and development of tumors by delivering various biologically active molecules between cells; their role in cancer immune escape in particular has been widely proven. EVs can carry a wide array of cargo, such as non-coding RNAs (ncRNAs), including miRNAs, lncRNAs, and circRNAs, which are selectively loaded by EVs, secreted, and transported to participate in the proliferation of immune cells. Hence, strategies to specifically target EV-ncRNAs could be attractive therapeutic options. In this review, we summarize the current research on the role of EV-ncRNAs in cancer immune escape, and discuss the latest research on the function and regulation mechanism of EV-ncRNAs in cancer immune escape, highlighting and elucidating the potential clinical applications of EV-ncRNAs, including in diagnosis and immunotherapy.
Collapse
Affiliation(s)
- Mingyao Huang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Shuo Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Shilei Tang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Bowen Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Hongyuan Jin
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Bo Wu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Jingang Liu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
| |
Collapse
|
112
|
Chen D, Bao C, Zhao F, Yu H, Zhong G, Xu L, Yan S. Exploring Specific miRNA-mRNA Axes With Relationship to Taxanes-Resistance in Breast Cancer. Front Oncol 2020; 10:1397. [PMID: 32974144 PMCID: PMC7473300 DOI: 10.3389/fonc.2020.01397] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 07/02/2020] [Indexed: 12/23/2022] Open
Abstract
Breast cancer is the most prevalent type of malignancy in women worldwide. Taxanes (paclitaxel and docetaxel) are widely applied as first-line chemotherapeutic agents, while the therapeutic effect is seriously limited by the development of drug resistance. In the present study, we screened out several miRNAs dysregulated in taxanes-resistant breast cancer samples and confirmed that two miRNAs (miR-335-5p and let-7c-5p) played a major role in cell proliferation, apoptosis, and chemo-resistance. In addition, the weighted gene co-expression network analysis (WGCNA) for potential target genes of miR-335-5p and let-7c-5p identified three hub genes (CXCL9, CCR7, and SOCS1) with a positive relationship to taxanes-sensitivity. Further, target relationships between miR-335-5p and CXCL9, let-7c-5p and CCR7/SOCS1 were confirmed by dual-luciferase reporter assays. Importantly, the regulatory functions of CXCL9, CCR7, and SOCS1 on proliferation and chemoresistance were validated. In conclusion, our study shed light on clinical theragnostic relationships between miR-335-5p/CXCL9, let-7c-5p/CCR7/SOCS1 axes, and taxanes-resistance in breast cancer.
Collapse
Affiliation(s)
- Danni Chen
- Department of Radiation Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Chang Bao
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Organ Transplantation, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou, China
| | - Feng Zhao
- Department of Radiation Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Haogang Yu
- Department of Radiation Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Guansheng Zhong
- Breast Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Liang Xu
- Clinical Research Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Senxiang Yan
- Department of Radiation Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
113
|
Cui Q, Tang J, Zhang D, Kong D, Liao X, Ren J, Gong Y, Xie C, Wu G. A prognostic eight-gene expression signature for patients with breast cancer receiving adjuvant chemotherapy. J Cell Biochem 2020; 121:3923-3934. [PMID: 31692061 DOI: 10.1002/jcb.29550] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 10/10/2019] [Indexed: 01/24/2023]
Abstract
Breast cancer is a popularly diagnosed malignant tumor. Genomic profiling studies suggest that breast cancer is a disease with heterogeneity. Chemotherapy is one of the chief means to treat breast cancer, while its responses and clinical outcomes vary largely due to the conventional clinicopathological factors and inherent chemosensitivity of breast cancer. Using the least absolute shrinkage and selection operator (LASSO) Cox regression model, our study established a multi-mRNA-based signature model and constructed a relative nomogram in predicting distant-recurrence-free survival for patients receiving surgery and following chemotherapy. We constructed a signature of eight mRNAs (IPCEF1, SYNDIG1, TIGIT, SPESP1, C2CD4A, CLCA2, RLN2, and CCL19) with the LASSO model, which was employed to separate subjects into groups with high- and low-risk scores. Obvious differences of distant-recurrence-free survival were found between these two groups. This eight-mRNA-based signature was independently associated with the prognosis and had better prognostic value than classical clinicopathologic factors according to multivariate Cox regression results. Receiver operating characteristic results demonstrated excellent performance in diagnosing 3-year distant-recurrence by the eight-mRNA signature. A nomogram that combined both the eight-mRNA-based signature and clinicopathological risk factors was constructed. Comparing with an ideal model, the nomograms worked well both in the training and validation sets. Through the results that the eight-mRNA signature effectively classified patients into low- and high-risk of distant recurrence, we concluded that this eight-mRNA-based signature played a promising predictive role in prognosis and could be clinically applied in breast cancer patients receiving adjuvant chemotherapy.
Collapse
Affiliation(s)
- Qiuxia Cui
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jianing Tang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dan Zhang
- Department of Thyroid and Breast Surgery, Tongji Hospital of Huazhong University of Science and Technology, Wuhan, China
| | - Deguang Kong
- Department of General Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xing Liao
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiangbo Ren
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Gong
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Gaosong Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
114
|
Karpus WJ. Cytokines and Chemokines in the Pathogenesis of Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2020; 204:316-326. [PMID: 31907274 DOI: 10.4049/jimmunol.1900914] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/13/2019] [Indexed: 12/25/2022]
Abstract
Experimental autoimmune encephalomyelitis is a CD4+ T cell-mediated demyelinating disease of the CNS that serves as a model for multiple sclerosis. Cytokines and chemokines shape Th1 and Th17 effector responses as well as regulate migration of leukocytes to the CNS during disease. The CNS cellular infiltrate consists of Ag-specific and nonspecific CD4+ and CD8+ T cells, neutrophils, B cells, monocytes, macrophages, and dendritic cells. The mechanism of immune-mediated inflammation in experimental autoimmune encephalomyelitis has been extensively studied in an effort to develop therapeutic modalities for multiple sclerosis and, indeed, has provided insight in modern drug discovery. The present Brief Review highlights critical pathogenic aspects of cytokines and chemokines involved in generation of effector T cell responses and migration of inflammatory cells to the CNS. Select cytokines and chemokines are certainly important in the regulatory response, which involves T regulatory, B regulatory, and myeloid-derived suppressor cells. However, that discussion is beyond the scope of this brief review.
Collapse
Affiliation(s)
- William J Karpus
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
115
|
MicroRNA miR-155 is required for expansion of regulatory T cells to mediate robust pregnancy tolerance in mice. Mucosal Immunol 2020; 13:609-625. [PMID: 31988469 DOI: 10.1038/s41385-020-0255-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 12/11/2019] [Accepted: 01/08/2020] [Indexed: 02/04/2023]
Abstract
The immune-regulatory microRNA miR-155 is reduced in recurrent miscarriage, suggesting that miR-155 contributes to immune tolerance in pregnancy. Here we show miR-155 is induced in the uterine mucosa and draining lymph nodes (dLN) during the female immune response to male seminal fluid alloantigens. Mice with null mutation in miR-155 (miR-155-/-) exhibited a reduced CD4+ T cell response after mating, with a disproportionate loss of CD25+FOXP3+ Treg cells. miR-155 deficiency impaired expansion of both peripheral and thymic Treg cells, distinguished by neuropilin-1 (NRP1), and fewer Treg cells expressed Ki67 proliferation marker and suppressive function marker CTLA4. Altered Treg phenotype distribution in miR-155-/- mice was confirmed by t-distributed neighbor embedding (tSNE) analysis. Fewer dendritic cells (DCs) and macrophages trafficked to the dLN of miR-155-/- mice, associated with lower CCR7 on DCs, and reduced uterine Ccl19 expression, implicating compromised antigen presentation in the stunted Treg cell response. miR-155-/- mice exhibited elevated susceptibility to inflammation-induced fetal loss and fetal growth restriction compared with miR-155+/+ controls, but outcomes were restored by transfer of wild-type Tregs. Thus miR-155 is a key regulator of immune adaptation to pregnancy and is necessary for sufficient Tregs to achieve robust pregnancy tolerance and protect against fetal loss.
Collapse
|
116
|
Senécal JL, Hoa S, Yang R, Koenig M. Pathogenic roles of autoantibodies in systemic sclerosis: Current understandings in pathogenesis. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2020; 5:103-129. [PMID: 35382028 PMCID: PMC8922609 DOI: 10.1177/2397198319870667] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/29/2019] [Indexed: 09/12/2023]
Abstract
The potential pathogenic role for autoantibodies in systemic sclerosis has captivated researchers for the past 40 years. This review answers the question whether there is yet sufficient knowledge to conclude that certain serum autoantibodies associated with systemic sclerosis contribute to its pathogenesis. Definitions for pathogenic, pathogenetic and functional autoantibodies are formulated, and the need to differentiate these autoantibodies from natural autoantibodies is emphasized. In addition, seven criteria for the identification of pathogenic autoantibodies are proposed. Experimental evidence is reviewed relevant to the classic systemic sclerosis antinuclear autoantibodies, anti-topoisomerase I and anticentromere, and to functional autoantibodies to endothelin 1 type A receptor, angiotensin II type 1 receptor, muscarinic receptor 3, platelet-derived growth factor receptor, chemokine receptors CXCR3 and CXCR4, estrogen receptor α, and CD22. Pathogenic evidence is also reviewed for anti-matrix metalloproteinases 1 and 3, anti-fibrillin 1, anti-IFI16, anti-eIF2B, anti-ICAM-1, and anti-RuvBL1/RuvBL2 autoantibodies. For each autoantibody, objective evidence for a pathogenic role is scored qualitatively according to the seven pathogenicity criteria. It is concluded that anti-topoisomerase I is the single autoantibody specificity with the most evidence in favor of a pathogenic role in systemic sclerosis, followed by anticentromere. However, these autoantibodies have not been demonstrated yet to fulfill completely the seven proposed criteria for pathogenicity. Their contributory roles to the pathogenesis of systemic sclerosis remain possible but not yet conclusively demonstrated. With respect to functional autoantibodies and other autoantibodies, only a few criteria for pathogenicity are fulfilled. Their common presence in healthy and disease controls suggests that major subsets of these immunoglobulins are natural autoantibodies. While some of these autoantibodies may be pathogenetic in systemic sclerosis, establishing that they are truly pathogenic is a work in progress. Experimental data are difficult to interpret because high serum autoantibody levels may be due to polyclonal B-cell activation. Other limitations in experimental design are the use of total serum immunoglobulin G rather than affinity-purified autoantibodies, the confounding effect of other systemic sclerosis autoantibodies present in total immunoglobulin G and the lack of longitudinal studies to determine if autoantibody titers fluctuate with systemic sclerosis activity and severity. These intriguing new specificities expand the spectrum of autoantibodies observed in systemic sclerosis. Continuing elucidation of their potential mechanistic roles raises hope of a better understanding of systemic sclerosis pathogenesis leading to improved therapies.
Collapse
Affiliation(s)
- Jean-Luc Senécal
- Scleroderma Research Chair, Université de Montréal, Montreal, QC, Canada
- Division of Rheumatology, Centre hospitalier de l’Université de Montréal, Montreal, QC, Canada
- Autoimmunity Research Laboratory, Research Centre of the Centre hospitalier de l’Université de Montréal, Montreal, QC, Canada
| | - Sabrina Hoa
- Division of Rheumatology, Centre hospitalier de l’Université de Montréal, Montreal, QC, Canada
- Autoimmunity Research Laboratory, Research Centre of the Centre hospitalier de l’Université de Montréal, Montreal, QC, Canada
| | - Roger Yang
- Division of Rheumatology, Centre hospitalier de l’Université de Montréal, Montreal, QC, Canada
| | - Martial Koenig
- Autoimmunity Research Laboratory, Research Centre of the Centre hospitalier de l’Université de Montréal, Montreal, QC, Canada
- Division of Internal Medicine, Centre hospitalier de l’Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
117
|
Fu K, March K, Alexaki A, Fabozzi G, Moysi E, Petrovas C. Immunogenicity of Protein Therapeutics: A Lymph Node Perspective. Front Immunol 2020; 11:791. [PMID: 32477334 PMCID: PMC7240201 DOI: 10.3389/fimmu.2020.00791] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 04/07/2020] [Indexed: 12/31/2022] Open
Abstract
The continuous development of molecular biology and protein engineering technologies enables the expansion of the breadth and complexity of protein therapeutics for in vivo administration. However, the immunogenicity and associated in vivo development of antibodies against therapeutics are a major restriction factor for their usage. The B cell follicular and particularly germinal center areas in secondary lymphoid organs are the anatomical sites where the development of antibody responses against pathogens and immunogens takes place. A growing body of data has revealed the importance of the orchestrated function of highly differentiated adaptive immunity cells, including follicular helper CD4 T cells and germinal center B cells, for the optimal generation of these antibody responses. Understanding the cellular and molecular mechanisms mediating the antibody responses against therapeutics could lead to novel strategies to reduce their immunogenicity and increase their efficacy.
Collapse
Affiliation(s)
- Kristy Fu
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Kylie March
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Aikaterini Alexaki
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Giulia Fabozzi
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Eirini Moysi
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Constantinos Petrovas
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
118
|
Rodríguez-Fernández JL, Criado-García O. The Chemokine Receptor CCR7 Uses Distinct Signaling Modules With Biased Functionality to Regulate Dendritic Cells. Front Immunol 2020; 11:528. [PMID: 32351499 PMCID: PMC7174648 DOI: 10.3389/fimmu.2020.00528] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 03/09/2020] [Indexed: 12/22/2022] Open
Abstract
Chemotaxis is a molecular mechanism that confers leukocytes the ability to detect gradients of chemoattractants. Chemokine receptors are well-known regulators of chemotaxis in leukocytes; however, they can regulate several other activities in these cells. This information has been often neglected, probably due to the paramount role of chemotaxis in the immune system and in biology. Therefore, the experimental data available on the mechanisms used by chemokine receptors to regulate other functions of leukocytes is sparse. The results obtained in the study of the chemokine receptor CCR7 in dendritic cells (DCs) provide interesting information on this issue. CCR7 guides the DCs from the peripheral tissues to the lymph nodes, where these cells control T cell activation. CCR7 can regulate DC chemotaxis, survival, migratory speed, cytoarchitecture, and endocytosis. Biochemical and functional analyses show: first, that CCR7 uses in DCs the PI3K/Akt pathway to control survival, the MAPK pathway to control chemotaxis, and the RhoA pathways to regulate actin dynamics, which in turn controls migratory speed, cytoarchitecture, and endocytosis; second, that these three signaling pathways behave as modules with a high degree of independence; and third, that although each one of these routes can regulate several functions in different settings, CCR7 promotes in DCs a functional bias in each pathway. The data uncover an interesting mechanism used by CCR7 to regulate the DCs, entailing multifunctional signaling pathways organized in modules with biased functionality. A similar mechanism could be used by other chemoattractant receptors to regulate the functions of leukocytes.
Collapse
Affiliation(s)
- José Luis Rodríguez-Fernández
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Olga Criado-García
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
119
|
Hutchings CJ. A review of antibody-based therapeutics targeting G protein-coupled receptors: an update. Expert Opin Biol Ther 2020; 20:925-935. [PMID: 32264722 DOI: 10.1080/14712598.2020.1745770] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION G protein-coupled receptors (GPCRs) play key roles in many biological functions and are linked to many diseases across all therapeutic areas. As such, GPCRs represent a significant opportunity for antibody-based therapeutics. AREAS COVERED The structure of the major GPCR families is summarized in the context of choice of antigen source employed in the drug discovery process and receptor biology considerations which may impact on targeting strategies. An overview of the therapeutic GPCR-antibody target landscape and the diversity of current therapeutic programs is provided along with summary case studies for marketed antibody drugs or those in advanced clinical studies. Antibodies in early clinical studies and the emergence of next-generation modalities are also highlighted. EXPERT OPINION The GPCR-antibody pipeline has progressed significantly with a number of technical developments enabling the successful resolution of some of the challenges previously encountered and this has contributed to the growing interest in antibody-based therapeutics addressing this target class.
Collapse
|
120
|
Quebrada Palacio LP, Fernández ER, Hernández-Vásquez Y, Petray PB, Postan M. Circulating T Follicular Helper Cell Abnormalities Associated to Different Clinical Forms of Chronic Chagas Disease. Front Cell Infect Microbiol 2020; 10:126. [PMID: 32296649 PMCID: PMC7136390 DOI: 10.3389/fcimb.2020.00126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 03/06/2020] [Indexed: 12/31/2022] Open
Abstract
Multiple perturbations of the immune response affecting a range of cells have been reported in Trypanosoma cruzi-infected individuals and associated to clinical manifestations of chronic Chagas disease. There is a paucity of knowledge about the role of T follicular helper (Tfh) cells in this infection. Here, we sought to characterize circulating Tfh (cTfh) cells in chronic Chagas disease patients and to identify potential associations with disease severity in humans. cTfh cells were characterized by flow cytometry in freshly isolated PBMCs from 7 T. cruzi-infected asymptomatic patients (ASYMP), 5 patients with chronic chagasic dilated cardiomyopathy (CCC) and 8 healthy controls, using antibodies against chemokine receptors CXCR5, CXCR3, CCR6, and CCR7. Our results showed significant expansion of CD4+CD45RO+CXCR5+CCR6+ cells in ASYMP and CCC patients, along with a contraction of CD4+CD45RO+CXCR5+CXCR3-CCR6- (cTfh2) cells. ASYMP patients further exhibited decreased CD4+CD45RO+CXCR5+CXCR3+CCR6- (cTfh1) cells and expanded CD4+CD45RO+CXCR5+CXCR3-CCR6+ (cTfh17) cells while CCC patients exhibited significantly increased frequencies of CD4+CD45RO+CXCR5+CCR7+ cells. Linear regression analysis revealed a positive trend of CD4+CD45RO+CXCR5+CXCR3+CCR6+ (cTfh1/17) cells and negative trends of cTfh1 and cTfh2 cells as disease was more severe. There was no correlation between the frequencies of cTfh cells and circulating CD19+IgD-IgG+ cells or serum levels of T. cruzi-specific IgG. These results demonstrate that the cTfh compartment of humans chronically infected with T. cruzi comprises expanded CCR6-expressing cells and reduced cTfh2 cells. The association of discrete phenotypic changes in cTfh subsets with different clinical forms suggests the potential contribution of T follicular helper cells to Chagas heart disease progression.
Collapse
Affiliation(s)
- Luz P Quebrada Palacio
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chabén", ANLIS/Malbran, Buenos Aires, Argentina
| | - Esteban R Fernández
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chabén", ANLIS/Malbran, Buenos Aires, Argentina
| | - Yolanda Hernández-Vásquez
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chabén", ANLIS/Malbran, Buenos Aires, Argentina
| | - Patricia B Petray
- Facultad de Medicina, IMPaM-Instituto de Investigaciones en Microbiología y Parasitología Médica (UBA-CONICET), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
| | - Miriam Postan
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chabén", ANLIS/Malbran, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
121
|
Parallels of Resistance between Angiogenesis and Lymphangiogenesis Inhibition in Cancer Therapy. Cells 2020; 9:cells9030762. [PMID: 32244922 PMCID: PMC7140636 DOI: 10.3390/cells9030762] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/13/2020] [Accepted: 03/18/2020] [Indexed: 12/24/2022] Open
Abstract
Metastasis is the primary cause of cancer-related mortality. Cancer cells primarily metastasize via blood and lymphatic vessels to colonize lymph nodes and distant organs, leading to worse prognosis. Thus, strategies to limit blood and lymphatic spread of cancer have been a focal point of cancer research for several decades. Resistance to FDA-approved anti-angiogenic therapies designed to limit blood vessel growth has emerged as a significant clinical challenge. However, there are no FDA-approved drugs that target tumor lymphangiogenesis, despite the consequences of metastasis through the lymphatic system. This review highlights several of the key resistance mechanisms to anti-angiogenic therapy and potential challenges facing anti-lymphangiogenic therapy. Blood and lymphatic vessels are more than just conduits for nutrient, fluid, and cancer cell transport. Recent studies have elucidated how these vasculatures often regulate immune responses. Vessels that are abnormal or compromised by tumor cells can lead to immunosuppression. Therapies designed to improve lymphatic vessel function while limiting metastasis may represent a viable approach to enhance immunotherapy and limit cancer progression.
Collapse
|
122
|
Integrated miRNA/mRNA Counter-Expression Analysis Highlights Oxidative Stress-Related Genes CCR7 and FOXO1 as Blood Markers of Coronary Arterial Disease. Int J Mol Sci 2020; 21:ijms21061943. [PMID: 32178422 PMCID: PMC7139611 DOI: 10.3390/ijms21061943] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 03/10/2020] [Indexed: 11/23/2022] Open
Abstract
Our interest in the mechanisms of atherosclerosis progression (ATHp) has led to the recent identification of 13 miRNAs and 1285 mRNAs whose expression was altered during ATHp. Here, we deepen the functional relationship among these 13 miRNAs and genes associated to oxidative stress, a crucial step in the onset and progression of vascular disease. We first compiled a list of genes associated to the response to oxidative stress (Oxstress genes) by performing a reverse Gene Ontology analysis (rGO, from the GO terms to the genes) with the GO terms GO0006979, GO1902882, GO1902883 and GO1902884, which included a total of 417 unique Oxstress genes. Next, we identified 108 putative targets of the 13 miRNAs among these unique Oxstress genes, which were validated by an integrated miRNA/mRNA counter-expression analysis with the 1285 mRNAs that yielded 14 genes, Map2k1, Mapk1, Mapk9, Dapk1, Atp2a2, Gata4, Fos, Egfr, Foxo1, Ccr7, Vkorc1l1, Rnf7, Kcnh3, and Mgat3. GO enrichment analysis and a protein–protein-interaction network analysis (PPI) identified most of the validated Oxstress transcripts as components of signaling pathways, highlighting a role for MAP signaling in ATHp. Lastly, expression of these Oxstress transcripts was measured in PBMCs from patients suffering severe coronary artery disease, a serious consequence of ATHp. This allowed the identification of FOXO1 and CCR7 as blood markers downregulated in CAD. These results are discussed in the context of the interaction of the Oxstress transcripts with the ATHp-associated miRNAs.
Collapse
|
123
|
Wei J, Mattapallil MJ, Horai R, Jittayasothorn Y, Modi AP, Sen HN, Gronert K, Caspi RR. A novel role for lipoxin A 4 in driving a lymph node-eye axis that controls autoimmunity to the neuroretina. eLife 2020; 9:e51102. [PMID: 32118582 PMCID: PMC7064344 DOI: 10.7554/elife.51102] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 02/29/2020] [Indexed: 12/14/2022] Open
Abstract
The eicosanoid lipoxin A4 (LXA4) has emerging roles in lymphocyte-driven diseases. We identified reduced LXA4 levels in posterior segment uveitis patients and investigated the role of LXA4 in the pathogenesis of experimental autoimmune uveitis (EAU). Immunization for EAU with a retinal self-antigen caused selective downregulation of LXA4 in lymph nodes draining the site of immunization, while at the same time amplifying LXA4 in the inflamed target tissue. T cell effector function, migration and glycolytic responses were amplified in LXA4-deficient mice, which correlated with more severe pathology, whereas LXA4 treatment attenuated disease. In vivo deletion or supplementation of LXA4 identified modulation of CC-chemokine receptor 7 (CCR7) and sphingosine 1- phosphate receptor-1 (S1PR1) expression and glucose metabolism in CD4+ T cells as potential mechanisms for LXA4 regulation of T cell effector function and trafficking. Our results demonstrate the intrinsic lymph node LXA4 pathway as a significant checkpoint in the development and severity of adaptive immunity.
Collapse
Affiliation(s)
- Jessica Wei
- Vision Science Program, University of California, BerkeleyBerkeleyUnited States
- Laboratory of Immunology, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Mary J Mattapallil
- Laboratory of Immunology, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Reiko Horai
- Laboratory of Immunology, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Yingyos Jittayasothorn
- Laboratory of Immunology, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Arnav P Modi
- School of Optometry, University of California, BerkeleyBerkeleyUnited States
| | - H Nida Sen
- Laboratory of Immunology, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Karsten Gronert
- Vision Science Program, University of California, BerkeleyBerkeleyUnited States
- School of Optometry, University of California, BerkeleyBerkeleyUnited States
- Infectious Disease and Immunity Program, University of California, BerkeleyBerkeleyUnited States
| | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, National Institutes of HealthBethesdaUnited States
| |
Collapse
|
124
|
Anatomical Uniqueness of the Mucosal Immune System (GALT, NALT, iBALT) for the Induction and Regulation of Mucosal Immunity and Tolerance. MUCOSAL VACCINES 2020. [PMCID: PMC7149644 DOI: 10.1016/b978-0-12-811924-2.00002-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
125
|
Løken OM, Bjørgen H, Hordvik I, Koppang EO. A teleost structural analogue to the avian bursa of Fabricius. J Anat 2019; 236:798-808. [PMID: 31877586 PMCID: PMC7163591 DOI: 10.1111/joa.13147] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2019] [Indexed: 01/26/2023] Open
Abstract
The bursa of Fabricius is a primary and secondary lymphoid organ considered exclusively present in birds, and studies of this structure have been vital to our current understanding of the adaptive immune system of vertebrates. In this study, we reveal substantial lymphoepithelial tissue in a previously undescribed bursa in Atlantic salmon (Salmo salar), situated caudal to the urogenital papilla of the cloaca and thus analogous to the anatomical placement of the bursa of Fabricius. We investigated three groups of Atlantic salmon at different maturational stages and characterized the structure by applying dissection, radiology, scanning electron microscopy and histological techniques, including immunohistochemistry and in situ hybridization. We found that the epithelial anlage of the salmon cloacal bursa developed into substantial lymphoepithelial tissue and subsequently regressed following sexual maturation. Such a dynamic development is also a key characteristic of the avian bursa. The presence of intraepithelial lymphocytes was concomitant with expression of the leukocyte-attracting chemokine CCL19, indicative of lymphoid organ functions. We did not observe recombination or gene conversion in salmon bursal lymphocytes at any developmental stage, indicating the absence of primary lymphoid organ functions in contrast to the bursa of Fabricius. However, the possibility of the bursa to trap both enteric and environmental antigens, combined with the presence of several antigen-presenting cells residing within the lymphoepithelium, suggest the structure has secondary lymphoid organ functions. We present the discovery of a lymphoid organ in Atlantic salmon with striking topographical similarities to that of the bursa of Fabricius in birds. In addition, the age-dependent dynamics of its lymphoepithelium suggest functions related to the maturation processes of lymphocytes.
Collapse
Affiliation(s)
- Oskar M Løken
- Section of Anatomy, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Håvard Bjørgen
- Section of Anatomy, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Ivar Hordvik
- Institute of Biology, University of Bergen, Bergen, Norway
| | - Erling O Koppang
- Section of Anatomy, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| |
Collapse
|
126
|
Wang T, Li W, Cheng H, Zhong L, Deng J, Ling S. The Important Role of the Chemokine Axis CCR7-CCL19 and CCR7-CCL21 in the Pathophysiology of the Immuno-inflammatory Response in Dry Eye Disease. Ocul Immunol Inflamm 2019; 29:266-277. [PMID: 31702421 DOI: 10.1080/09273948.2019.1674891] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Purpose: To explore whether CCR7-CCL19 and CCR7-CCL21 affect the pathophysiology of the dry eye disease (DED) immuno-inflammatory response using a murine model.Methods: The mRNA expression levels of CCR7, CCL19, CCL21 and VEGF-C within corneas in DED mice were detected by real-time PCR. Immunofluorescence and flow cytometric analyses were performed to mark dendritic cells (DCs) and detect correlations among CCR7, CCL19, CCL21 and lymphatic vessels.Results: CCR7, CCL19 and CCL21 expression was dramatically increased during the development of DED. In addition, CCR7, which is expressed in DCs, was located inside and around lymphatic vessels and colocalized with CCL19 or CCL21. Positive correlations were observed between CCR7 and CCL19 (P < .01, r = 0.862), CCL21 (P < .01, r = 0.759), and VEGF-C (P < .05, r = 0.607).Conclusions: Our study revealed that both the CCR7-CCL19 and CCR7-CCL21 chemokine axis are important for DC migration to lymphatic vessels, but CCL19 may have a greater effect on DED than CCL21.
Collapse
Affiliation(s)
- Ting Wang
- Department of Ophthalmology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Weihua Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, P.R. China
| | - Huanhuan Cheng
- Department of Ophthalmology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Lei Zhong
- Department of Ophthalmology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Juan Deng
- Department of Ophthalmology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Shiqi Ling
- Department of Ophthalmology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| |
Collapse
|
127
|
Martin-Rufino JD, Espinosa-Lara N, Osugui L, Sanchez-Guijo F. Targeting the Immune System With Mesenchymal Stromal Cell-Derived Extracellular Vesicles: What Is the Cargo's Mechanism of Action? Front Bioeng Biotechnol 2019; 7:308. [PMID: 31781552 PMCID: PMC6856662 DOI: 10.3389/fbioe.2019.00308] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/17/2019] [Indexed: 12/12/2022] Open
Abstract
The potent immunomodulatory activities displayed by mesenchymal stromal cells (MSCs) have motivated their application in hundreds of clinical trials to date. In some countries, they have subsequently been approved for the treatment of immune disorders such as Crohn's disease and graft-versus-host disease. Increasing evidence suggests that their main mechanism of action in vivo relies on paracrine signaling and extracellular vesicles. Mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) play a prominent role in intercellular communication by allowing the horizontal transfer of microRNAs, mRNAs, proteins, lipids and other bioactive molecules between MSCs and their targets. However, despite the considerable momentum gained by MSC-EV research, the precise mechanism by which MSC-EVs interact with the immune system is still debated. Available evidence is highly context-dependent and fragmentary, with a limited number of reports trying to link their efficacy to specific active components shuttled within them. In this concise review, currently available evidence on the molecular mechanisms underlying the effects of MSC-EV cargo on the immune system is analyzed. Studies that pinpoint specific MSC-EV-borne mediators of immunomodulation are highlighted, with a focus on the signaling events triggered by MSC-EVs in target immune cells. Reports that study the effects of preconditioning or “licensing” in MSC-EV-mediated immunomodulation are also presented. The need for further studies that dissect the mechanisms of MSC-EV cargo in the adaptive immune system is emphasized. Finally, the major challenges that need to be addressed to harness the full potential of these signaling vehicles are discussed, with the ultimate goal of effectively translating MSC-EV treatments into the clinic.
Collapse
Affiliation(s)
- Jorge Diego Martin-Rufino
- Unidad de Terapia Celular, Servicio de Hematología, IBSAL-Hospital Universitario de Salamanca, Salamanca, Spain.,Facultad de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Natalia Espinosa-Lara
- Unidad de Terapia Celular, Servicio de Hematología, IBSAL-Hospital Universitario de Salamanca, Salamanca, Spain
| | - Lika Osugui
- Unidad de Terapia Celular, Servicio de Hematología, IBSAL-Hospital Universitario de Salamanca, Salamanca, Spain
| | - Fermin Sanchez-Guijo
- Unidad de Terapia Celular, Servicio de Hematología, IBSAL-Hospital Universitario de Salamanca, Salamanca, Spain.,Facultad de Medicina, Universidad de Salamanca, Salamanca, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
| |
Collapse
|
128
|
Bjørgen H, Løken OM, Aas IB, Fjelldal PG, Hansen T, Austbø L, Koppang EO. Visualization of CCL19-like transcripts in the ILT, thymus and head kidney of Atlantic salmon (Salmo salar L.). FISH & SHELLFISH IMMUNOLOGY 2019; 93:763-765. [PMID: 31422180 DOI: 10.1016/j.fsi.2019.08.044] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/12/2019] [Accepted: 08/14/2019] [Indexed: 06/10/2023]
Affiliation(s)
- Håvard Bjørgen
- Section of Anatomy and Pathology, Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ullevålsveien 72, Oslo, Norway
| | - Oskar Mongstad Løken
- Section of Anatomy and Pathology, Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ullevålsveien 72, Oslo, Norway
| | - Ida Bergva Aas
- Section of Anatomy and Pathology, Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ullevålsveien 72, Oslo, Norway
| | | | - Tom Hansen
- Matre Research Station, Institute of Marine Research, 5498, Matre, Norway
| | - Lars Austbø
- Section of Anatomy and Pathology, Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ullevålsveien 72, Oslo, Norway
| | - Erling Olaf Koppang
- Section of Anatomy and Pathology, Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ullevålsveien 72, Oslo, Norway.
| |
Collapse
|
129
|
Yan Y, Chen R, Wang X, Hu K, Huang L, Lu M, Hu Q. CCL19 and CCR7 Expression, Signaling Pathways, and Adjuvant Functions in Viral Infection and Prevention. Front Cell Dev Biol 2019; 7:212. [PMID: 31632965 PMCID: PMC6781769 DOI: 10.3389/fcell.2019.00212] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/18/2019] [Indexed: 12/11/2022] Open
Abstract
Chemokine (C–C motif) ligand 19 (CCL19) is a critical regulator of the induction of T cell activation, immune tolerance, and inflammatory responses during continuous immune surveillance, homeostasis, and development. Migration of CC-chemokine receptor 7 (CCR7)-expressing cells to secondary lymphoid organs is a crucial step in the onset of adaptive immunity, which is initiated by a complex interaction between CCR7 and its cognate ligands. Recent advances in knowledge regarding the response of the CCL19-CCR7 axis to viral infections have elucidated the complex network of interplay among the invading virus, target cells and host immune responses. Viruses use various strategies to evade or delay the cytokine response, gaining additional time to replicate in the host. In this review, we summarize the impacts of CCL19 and CCR7 expression on the regulation of viral pathogenesis with an emphasis on the corresponding signaling pathways and adjuvant mechanisms. We present and discuss the expression, signaling adaptor proteins and effects of CCL19 and CCR7 as these molecules differentially impact different viral infections and viral life cycles in host homeostatic strategies. The underlying mechanisms discussed in this review may assist in the design of novel agents to modulate chemokine activity for viral prevention.
Collapse
Affiliation(s)
- Yan Yan
- Center of Clinical Laboratory, The Fifth People's Hospital of Wuxi, Affiliated Hospital of Jiangnan University, Wuxi, China.,The International Joint Research Laboratory for Infection and Immunity (China-Germany), Jiangnan University, Wuxi, China
| | - Renfang Chen
- The International Joint Research Laboratory for Infection and Immunity (China-Germany), Jiangnan University, Wuxi, China.,Hepatology Institute of Wuxi, The Fifth People's Hospital of Wuxi, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xu Wang
- Center of Clinical Laboratory, The Fifth People's Hospital of Wuxi, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Kai Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Lihua Huang
- The International Joint Research Laboratory for Infection and Immunity (China-Germany), Jiangnan University, Wuxi, China.,Hepatology Institute of Wuxi, The Fifth People's Hospital of Wuxi, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Mengji Lu
- The International Joint Research Laboratory for Infection and Immunity (China-Germany), Jiangnan University, Wuxi, China.,Institute of Virology, University Hospital of Essen, University Duisburg-Essen, Essen, Germany
| | - Qinxue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Institute for Infection and Immunity, St. George's, University of London, London, United Kingdom
| |
Collapse
|
130
|
Gonzalez Badillo FE, Zisi Tegou F, Abreu MM, Masina R, Sha D, Najjar M, Wright SH, Bayer AL, Korpos É, Pugliese A, Molano RD, Tomei AA. CCL21 Expression in β-Cells Induces Antigen-Expressing Stromal Cell Networks in the Pancreas and Prevents Autoimmune Diabetes in Mice. Diabetes 2019; 68:1990-2003. [PMID: 31371518 PMCID: PMC6754241 DOI: 10.2337/db19-0239] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/21/2019] [Indexed: 12/31/2022]
Abstract
Tumors induce tolerance toward their antigens by producing the chemokine CCL21, leading to the formation of tertiary lymphoid organs (TLOs). Ins2-CCL21 transgenic, nonobese diabetic (NOD) mice express CCL21 in pancreatic β-cells and do not develop autoimmune diabetes. We investigated by which mechanisms CCL21 expression prevented diabetes. Ins2-CCL21 mice develop TLOs by 4 weeks of age, consisting of naive CD4+ T cells compartmentalized within networks of CD45-gp38+CD31- fibroblastic reticular cell (FRC)-like cells. Importantly, 12-week-old Ins2-CCL21 TLOs contained FRC-like cells with higher contractility, regulatory, and anti-inflammatory properties and enhanced expression of β-cell autoantigens compared with nontransgenic NOD TLOs found in inflamed islets. Consistently, transgenic mice harbored fewer autoreactive T cells and a higher proportion of regulatory T cells in the islets. Using adoptive transfer and islet transplantation models, we demonstrate that TLO formation in Ins2-CCL21 transgenic islets is critical for the regulation of autoimmunity, and although the effect is systemic, the induction is mediated locally likely by lymphocyte trafficking through TLOs. Overall, our findings suggest that CCL21 promotes TLOs that differ from inflammatory TLOs found in type 1 diabetic islets in that they resemble lymph nodes, contain FRC-like cells expressing β-cell autoantigens, and are able to induce systemic and antigen-specific tolerance leading to diabetes prevention.
Collapse
Affiliation(s)
- Freddy E Gonzalez Badillo
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Biomedical Engineering, University of Miami, Miami, FL
| | - Flavia Zisi Tegou
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Biomedical Engineering, University of Miami, Miami, FL
| | - Maria M Abreu
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Riccardo Masina
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Divya Sha
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Mejdi Najjar
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Shane H Wright
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Allison L Bayer
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL
| | - Éva Korpos
- Institute of Physiological Chemistry and Pathobiochemistry and Cells in Motion, Cluster of Excellence, University of Muenster, Muenster, Germany
| | - Alberto Pugliese
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - R Damaris Molano
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Alice A Tomei
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Biomedical Engineering, University of Miami, Miami, FL
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
131
|
Zeng M, Liu J, Yang W, Zhang S, Liu F, Dong Z, Peng Y, Sun L, Xiao L. Multiple-microarray analysis for identification of hub genes involved in tubulointerstial injury in diabetic nephropathy. J Cell Physiol 2019; 234:16447-16462. [PMID: 30761531 DOI: 10.1002/jcp.28313] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 01/19/2019] [Accepted: 01/24/2019] [Indexed: 01/24/2023]
Abstract
Diabetic nephropathy (DN) is a primary cause of renal failure. However, studies providing renal gene expression profiles of diabetic tubulointerstitial injury are scarce and its molecular mechanisms still await clarification. To identify vital genes involved in the diabetic tubulointerstitial injury, three microarray data sets from gene expression omnibus (GEO) were downloaded. A total of 127 differentially expressed genes (DEGs) were identified by limma package. Gene set enrichment analysis (GSEA) plots showed that sister chromatid cohesion was the most significant enriched gene set positively correlated with the DN group while retinoid X receptor binding was the most significant enriched gene set positively correlated with the control group. Enriched Gene Ontology (GO) annotations and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of DEGs mostly included extracellular matrix organization, extracellular space, extracellular matrix structural constituent, and Staphylococcus aureus infection. Twenty hub genes from three significant modules were ascertained by Cytoscape. Correlation analysis and subgroup analysis between hub genes and clinical features of DN showed that ALB, ANXA1, APOH, C3, CCL19, COL1A2, COL3A1, COL4A1, COL6A3, CXCL6, DCN, EGF, HRG, KNG1, LUM, SERPINA3, SPARC, SRGN, and TIMP1 may involve in diabetic tubulointerstitial injury. ConnectivityMap analysis indicated the most significant three compounds are 5182598, thapsigargin and 5224221. In conclusion, this study may provide new insights into the molecular mechanisms underlying diabetic tubulointerstitial injury as well as potential targets for diagnosis and therapeutics of DN.
Collapse
Affiliation(s)
- Mengru Zeng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jialu Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenxia Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shumin Zhang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fuyou Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia
| | - Youming Peng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Xiao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
132
|
Ma T, Luan S, Tao R, Lu D, Guo L, Liu J, Shu J, Zhou X, Han Y, Jia Y, Li G, Zhang H, Han W, Han Y, Li H. Targeted Migration of Human Adipose-Derived Stem Cells to Secondary Lymphoid Organs Enhances Their Immunomodulatory Effect and Prolongs the Survival of Allografted Vascularized Composites. Stem Cells 2019; 37:1581-1594. [PMID: 31414513 DOI: 10.1002/stem.3078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022]
Abstract
The targeted delivery of therapeutic agents to secondary lymphoid organs (SLOs), which are the niches for immune initiation, provides an unprecedented opportunity for immune intolerance induction. The alloimmune rejection postvascularized composite allotransplantation (VCA) is mediated by T lymphocytes. Human adipose-derived stem cells (hASCs) possess the superiority of convenient availability and potent immunoregulatory property, but their therapeutic results in the VCA are unambiguous thus far. Chemokine receptor 7 (CCR7) can specifically guide immune cells migrating into SLOs. There, the genes of CCR7-GFP or GFP alone were introduced into hASCs by lentivirus. hASCs/CCR7 maintained the multidifferentiation and immunoregulatory abilities, but it gained the migration capacity elicited by secondary lymphoid organ chemokine (SCL) (CCR7 ligand) in vitro. Noteworthily, intravenously infused hASCs/CCR7 targetedly relocated in the T-cell aggression area in SLOs. In a rat VCA model, hASCs/GFP transfusion had a rare effect on the allografted vascularized composite. However, hASCs/CCR7 infusion potently prolonged the grafts' survival time. The ameliorated pathologic exhibition and the regulated inflammatory cytokines in the peripheral blood were also observed. The altered axis of Th1/Th2 and Tregs/Th17 in SLOs may underlie the downregulated rejection response. Moreover, the proteomic examination of splenic T lymphocytes also confirmed that hASCs/CCR7 decreased the proteins related to cytokinesis, lymphocyte proliferation, differentiation, and apoptotic process. In conclusion, our present study demonstrated that targeted migration of hASCs/CCR7 to SLOs highly intensifies their in vivo immunomodulatory effect in the VCA model for the first time. We believe this SLO-targeting strategy may improve the clinical therapeutic efficacy of hASC for allogeneic and autogenic immune disease. Stem Cells 2019;37:1581-1594.
Collapse
Affiliation(s)
- Tian Ma
- Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China.,Department of Plastic and Reconstructive Surgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - ShaoLiang Luan
- Department of Vascular Surgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Ran Tao
- Department of Plastic and Reconstructive Surgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Di Lu
- Department of Plastic and Reconstructive Surgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China.,Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Beijing, People's Republic of China
| | - LingLi Guo
- Department of Plastic and Reconstructive Surgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - JieJie Liu
- Department of Molecular Biology, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jun Shu
- Department of Plastic and Reconstructive Surgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - XiangBin Zhou
- Department of Stomatology, The Third Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - YuDi Han
- Department of Burn and Plastic Surgery, The Seventh Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - YiQing Jia
- Department of Emergency, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Guo Li
- Department of Plastic and Reconstructive Surgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Hui Zhang
- Department of Plastic Surgery, The Second Hospital of Shanxi Medical University, Shanxi, People's Republic of China
| | - WeiDong Han
- Department of Molecular Biology, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Yan Han
- Department of Plastic and Reconstructive Surgery, The First Medical Centre of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Hong Li
- Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China.,Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Beijing, People's Republic of China
| |
Collapse
|
133
|
López-Huertas MR, Morín M, Madrid-Elena N, Gutiérrez C, Jiménez-Tormo L, Santoyo J, Sanz-Rodríguez F, Moreno Pelayo MÁ, Bermejo LG, Moreno S. Selective miRNA Modulation Fails to Activate HIV Replication in In Vitro Latency Models. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:323-336. [PMID: 31288207 PMCID: PMC6614709 DOI: 10.1016/j.omtn.2019.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 06/09/2019] [Accepted: 06/10/2019] [Indexed: 02/08/2023]
Abstract
HIV remains incurable because of viral persistence in latent reservoirs that are inaccessible to antiretroviral therapy. A potential curative strategy is to reactivate viral gene expression in latently infected cells. However, no drug so far has proven to be successful in vivo in reducing the reservoir, and therefore new anti-latency compounds are needed. We explored the role of microRNAs (miRNAs) in latency maintenance and their modulation as a potential anti-latency strategy. Latency models based on treating resting CD4 T cells with chemokine (C-C motif) ligand 19 (CCL19) or interleukin-7 (IL7) before HIV infection and next-generation sequencing were used to identify the miRNAs involved in HIV latency. We detected four upregulated miRNAs (miRNA-98, miRNA-4516, miRNA-4488, and miRNA-7974). Individual or combined inhibition of these miRNAs was performed by transfection into cells latently infected with HIV. Viral replication, assessed 72 h after transfection, did not increase after miRNA modulation, despite miRNA inhibition and lack of toxicity. Furthermore, the combined modulation of five miRNAs previously associated with HIV latency was not effective in these models. Our results do not support the modulation of miRNAs as a useful strategy for the reversal of HIV latency. As shown with other drugs, the potential of miRNA modulation as an HIV reactivation strategy could be dependent on the latency model used.
Collapse
Affiliation(s)
- María Rosa López-Huertas
- Servicio de Enfermedades Infecciosas, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain.
| | - Matías Morín
- Servicio de Genética, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Hospital Universitario Ramón y Cajal, CIBERER, 28034 Madrid, Spain
| | - Nadia Madrid-Elena
- Servicio de Enfermedades Infecciosas, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Carolina Gutiérrez
- Servicio de Enfermedades Infecciosas, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Laura Jiménez-Tormo
- Servicio de Enfermedades Infecciosas, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Javier Santoyo
- Edinburgh Genomics, The Roslin Institute, University of Edinburgh, Scotland, UK
| | - Francisco Sanz-Rodríguez
- Fluorescence Imaging Group, Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Miguel Ángel Moreno Pelayo
- Servicio de Genética, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Hospital Universitario Ramón y Cajal, CIBERER, 28034 Madrid, Spain
| | - Laura García Bermejo
- Grupo de Biomarcadores y Dianas Terapéuticas, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain.
| | - Santiago Moreno
- Servicio de Enfermedades Infecciosas, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain; Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá de Henares, 28871 Alcalá de Henares, Spain
| |
Collapse
|
134
|
Jakobs BD, Spannagel L, Purvanov V, Uetz-von Allmen E, Matti C, Legler DF. Engineering of Nanobodies Recognizing the Human Chemokine Receptor CCR7. Int J Mol Sci 2019; 20:E2597. [PMID: 31137829 PMCID: PMC6566259 DOI: 10.3390/ijms20102597] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/20/2019] [Accepted: 05/24/2019] [Indexed: 01/12/2023] Open
Abstract
The chemokine receptor CCR7 plays a pivotal role in health and disease. In particular, CCR7 controls homing of antigen-bearing dendritic cells and T cells to lymph nodes, where adaptive immune responses are initiated. However, CCR7 also guides T cells to inflamed synovium and thereby contributes to rheumatoid arthritis and promotes cancer cell migration and metastasis formation. Nanobodies have recently emerged as versatile tools to study G-protein-coupled receptor functions and are being tested in diagnostics and therapeutics. In this study, we designed a strategy to engineer novel nanobodies recognizing human CCR7. We generated a nanobody library based on a solved crystal structure of the nanobody Nb80 recognizing the β2-adrenergic receptor (β2AR) and by specifically randomizing two segments within complementarity determining region 1 (CDR1) and CDR3 of Nb80 known to interact with β2AR. We fused the nanobody library to one half of split-YFP in order to identify individual nanobody clones interacting with CCR7 fused to the other half of split-YFP using bimolecular fluorescence complementation. We present three novel nanobodies, termed Nb1, Nb5, and Nb38, that recognize human CCR7 without interfering with G-protein-coupling and downstream signaling. Moreover, we were able to follow CCR7 trafficking upon CCL19 triggering using Nb1, Nb5, and Nb38.
Collapse
Affiliation(s)
- Barbara D Jakobs
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, CH-8280 Kreuzlingen, Switzerland.
- Graduate School for Cellular and Biomedical Sciences, University of Bern, CH-3012 Bern, Switzerland.
| | - Lisa Spannagel
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, CH-8280 Kreuzlingen, Switzerland.
| | - Vladimir Purvanov
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, CH-8280 Kreuzlingen, Switzerland.
| | - Edith Uetz-von Allmen
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, CH-8280 Kreuzlingen, Switzerland.
| | - Christoph Matti
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, CH-8280 Kreuzlingen, Switzerland.
| | - Daniel F Legler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, CH-8280 Kreuzlingen, Switzerland.
- Faculty of Biology, University of Konstanz, D-78464 Konstanz, Germany.
| |
Collapse
|
135
|
Hashizume-Takizawa T, Kobayashi R, Tsuzukibashi O, Saito M, Kurita-Ochiai T. CCR7-deficient mice exhibit a delayed antigen-specific mucosal IgA antibody response to an oral recombinant Salmonella strain. Pathog Dis 2019; 77:5480464. [DOI: 10.1093/femspd/ftz024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 04/25/2019] [Indexed: 12/11/2022] Open
Abstract
ABSTRACT
The migration of antigen (Ag)-loading dendritic cells (DCs) from Peyer's patches (PPs) to the draining mesenteric lymph nodes (MLNs) via chemokine receptor 7 (CCR7) is thought to be an important step in the initiation of acquired immunity. Our previous study showed that PPs were indispensable for Ag-specific secretory (S)IgA antibody (Ab) responses against oral recombinant Salmonella (rSalmonella). In this study, we attempted to show direct PP DC migration to MLNs by employing photoconvertible protein transgenic mice and investigated the role of the CCR7 signaling pathway in mucosal IgA induction. Our results demonstrated an actual flux of DCs from PPs to MLNs. The frequency of CCR7+ CD11c+ DCs in MLNs of PP-deficient mice was reduced, suggesting that some PP DCs migrated via CCR7. Immunization of CCR7−/− mice elicited significantly lower levels of Ag-specific SIgA Ab responses, which was associated with diminished formation of the germinal center in PPs. However, increased SIgA Ab production and dissemination of rSalmonella were observed at later time points. These results suggest that, although CCR7 was required for SIgA induction at normal velocity, the CCR7-mediated pathway is not essential for the induction of Ag-specific SIgA Ab responses to rSalmonella.
Collapse
Affiliation(s)
- Tomomi Hashizume-Takizawa
- Departments of Microbiology and Immunology, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakaecho-Nishi, Matsudo, Chiba 271-8587, Japan
| | - Ryoki Kobayashi
- Community Oral Health, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakaecho-Nishi, Matsudo, Chiba 271–8587, Japan
| | - Osamu Tsuzukibashi
- Division of Laboratory Medicine for Dentistry, Department of Oral Health Science, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakaecho-Nishi, Matsudo, Chiba 271–8587, Japan
| | - Masanori Saito
- Departments of Microbiology and Immunology, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakaecho-Nishi, Matsudo, Chiba 271-8587, Japan
| | - Tomoko Kurita-Ochiai
- Departments of Microbiology and Immunology, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakaecho-Nishi, Matsudo, Chiba 271-8587, Japan
| |
Collapse
|
136
|
Farnsworth RH, Karnezis T, Maciburko SJ, Mueller SN, Stacker SA. The Interplay Between Lymphatic Vessels and Chemokines. Front Immunol 2019; 10:518. [PMID: 31105685 PMCID: PMC6499173 DOI: 10.3389/fimmu.2019.00518] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/26/2019] [Indexed: 12/21/2022] Open
Abstract
Chemokines are a family of small protein cytokines that act as chemoattractants to migrating cells, in particular those of the immune system. They are categorized functionally as either homeostatic, constitutively produced by tissues for basal levels of cell migration, or inflammatory, where they are generated in association with a pathological inflammatory response. While the extravasation of leukocytes via blood vessels is a key step in cells entering the tissues, the lymphatic vessels also serve as a conduit for cells that are recruited and localized through chemoattractant gradients. Furthermore, the growth and remodeling of lymphatic vessels in pathologies is influenced by chemokines and their receptors expressed by lymphatic endothelial cells (LECs) in and around the pathological tissue. In this review we summarize the diverse role played by specific chemokines and their receptors in shaping the interaction of lymphatic vessels, immune cells, and other pathological cell types in physiology and disease.
Collapse
Affiliation(s)
- Rae H Farnsworth
- Tumor Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Tara Karnezis
- Lymphatic and Regenerative Medicine Laboratory, O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Simon J Maciburko
- Lymphatic and Regenerative Medicine Laboratory, O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia.,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Melbourne, VIC, Australia
| | - Steven A Stacker
- Tumor Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
137
|
Belluzzi E, Olivotto E, Toso G, Cigolotti A, Pozzuoli A, Biz C, Trisolino G, Ruggieri P, Grigolo B, Ramonda R, Favero M. Conditioned media from human osteoarthritic synovium induces inflammation in a synoviocyte cell line. Connect Tissue Res 2019; 60:136-145. [PMID: 29695173 DOI: 10.1080/03008207.2018.1470167] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
AIM Osteoarthritis (OA) is a whole joint pathology involving cartilage, synovial membrane, meniscus, subchondral bone, and infrapatellar fat pad (IFP). Synovitis has been widely documented in OA suggesting its important role in pathogenesis. The aim of this study was to investigate the role of different joint tissues in promoting synovitis. MATERIALS AND METHODS Conditioned media (CM) from cartilage, synovial membrane, meniscus, and IFP were generated from tissues of five patients undergoing total knee replacement and used to stimulate a human fibroblast-like synoviocytes cell line (K4IM). Cytokines, chemokines, and metalloproteases release was analyzed in all CM by Bio-Plex Assay and sulfated glycosaminoglycan (GAG) content by dimethylmethylene blue assay. Gene expression of several markers was evaluated by real-time PCR in K4IM cells stimulated with the CM obtained from joint tissues. RESULTS CM from all tissues produced high levels of IL-6, IL-8, and CCL2. CCL21, MMP-3, and -13 levels were detected in all CM except IFP. MMP-10 was present only in CM of cartilage and synovial tissues. IL-1β, IL-15, TNF-α, CCL5, and CCL19 were undetectable. However, only K4IM cells stimulated by the CM from OA synovium showed an increase of IL-6, CXCL-8, CCL21, MMP10, and IL-1β expression. CONCLUSION Our study showed that K4IM might be a suitable in vitro model for evaluating different cellular pathways in OA studies. Importantly, we demonstrated that in OA, all joint tissues might be involved in the progression of synovitis with a predominant role of synovial membrane itself compared to the other joint tissues.
Collapse
Affiliation(s)
- Elisa Belluzzi
- a Rheumatology Unit, Department of Medicine-DIMED , University Hospital of Padova , Padova , Italy.,b Musculoskeletal Pathology and Oncology Laboratory, Department of Orthopaedics and Orthopaedic Oncology , University of Padova , Padova , Italy
| | - Eleonora Olivotto
- c RAMSES Laboratory-RIT Department , IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| | - Giovanna Toso
- b Musculoskeletal Pathology and Oncology Laboratory, Department of Orthopaedics and Orthopaedic Oncology , University of Padova , Padova , Italy
| | - Augusto Cigolotti
- d Department of Orthopaedics and Orthopaedic Oncology , University of Padova , Padova , Italy
| | - Assunta Pozzuoli
- b Musculoskeletal Pathology and Oncology Laboratory, Department of Orthopaedics and Orthopaedic Oncology , University of Padova , Padova , Italy
| | - Carlo Biz
- d Department of Orthopaedics and Orthopaedic Oncology , University of Padova , Padova , Italy
| | - Giovanni Trisolino
- e Reconstructive Hip and Knee Joint Surgery , IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy.,f Pediatric orthopedic and Traumatology , IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| | - Pietro Ruggieri
- d Department of Orthopaedics and Orthopaedic Oncology , University of Padova , Padova , Italy
| | - Brunella Grigolo
- c RAMSES Laboratory-RIT Department , IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy.,g Laboratory of Immunorheumatology and Tissue Regeneration , IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| | - Roberta Ramonda
- a Rheumatology Unit, Department of Medicine-DIMED , University Hospital of Padova , Padova , Italy
| | - Marta Favero
- a Rheumatology Unit, Department of Medicine-DIMED , University Hospital of Padova , Padova , Italy.,c RAMSES Laboratory-RIT Department , IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| |
Collapse
|
138
|
Zu G, Luo B, Yang Y, Tan Y, Tang T, Zhang Y, Chen X, Sun D. Meta-analysis of the prognostic value of C-C chemokine receptor type 7 in patients with solid tumors. Cancer Manag Res 2019; 11:1881-1892. [PMID: 30881115 PMCID: PMC6396671 DOI: 10.2147/cmar.s190510] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Expression of C-C chemokine receptor type 7 (CCR7) is associated with the prognosis of several cancers. The aim of this study was to conduct the meta-analysis to determine the prognostic value of CCR7 expression in solid tumors. Materials and methods We searched for relevant literature in the PubMed, Embase, and Cochrane Library databases (last updated on January 15, 2018). The associations of CCR7 expression with overall survival (OS), disease-free survival (DFS), recurrence-free survival (RFS), progress-free survival (PFS), and disease-specific survival (DSS) were estimated. Results In total, 30 qualified studies including 3,413 patients were enrolled. The results revealed that higher expression of CCR7 predicted poorer OS (pooled HR =1.79; 95% CI =1.49–2.16; P<0.001) and PFS (pooled HR =2.18; 95% CI =1.49–3.18; P<0.001), but was not associated with DFS (pooled HR =1.69; 95% CI =0.79–3.61; P=0.175), RFS (pooled HR =1.29; 95% CI =0.48–3.44; P=0.618), or DSS (pooled HR =3.06; 95% CI =0.38–24.83; P<0.294). Conclusion From this meta-analysis, we concluded that high expression of CCR7 in tumor tissue is associated with poor survival in patients with solid tumors, and may be a prognostic biomarker for tumor progression.
Collapse
Affiliation(s)
- Guangchen Zu
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, People's Republic of China, ;
| | - Baoyang Luo
- Department of Hepatobiliary Surgery, Taizhou People's Hospital, Taizhou 225300, People's Republic of China
| | - Yong Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, People's Republic of China, ;
| | - Yuwei Tan
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, People's Republic of China, ;
| | - Tianyu Tang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, People's Republic of China, ;
| | - Yue Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, People's Republic of China, ;
| | - Xuemin Chen
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, People's Republic of China, ;
| | - Donglin Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, People's Republic of China, ;
| |
Collapse
|
139
|
Vezzani B, Shaw I, Lesme H, Yong L, Khan N, Tremolada C, Péault B. Higher Pericyte Content and Secretory Activity of Microfragmented Human Adipose Tissue Compared to Enzymatically Derived Stromal Vascular Fraction. Stem Cells Transl Med 2018; 7:876-886. [PMID: 30255987 PMCID: PMC6265639 DOI: 10.1002/sctm.18-0051] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 07/19/2018] [Indexed: 12/14/2022] Open
Abstract
Autologous adipose tissue is used for tissue repletion and/or regeneration as an intact lipoaspirate or as enzymatically derived stromal vascular fraction (SVF), which may be first cultured into mesenchymal stem cells (MSCs). Alternatively, transplant of autologous adipose tissue mechanically fragmented into submillimeter clusters has recently showed remarkable efficacy in diverse therapeutic indications. To document the biologic basis of the regenerative potential of microfragmented adipose tissue, we first analyzed the distribution of perivascular presumptive MSCs in adipose tissue processed with the Lipogems technology, observing a significant enrichment in pericytes, at the expense of adventitial cells, as compared to isogenic enzymatically processed lipoaspirates. The importance of MSCs as trophic and immunomodulatory cells, due to the secretion of specific factors, has been described. Therefore, we investigated protein secretion by cultured adipose tissue clusters or enzymatically derived SVF using secretome arrays. In culture, microfragmented adipose tissue releases many more growth factors and cytokines involved in tissue repair and regeneration, noticeably via angiogenesis, compared to isogenic SVF. Therefore, we suggest that the efficient tissue repair/regeneration observed after transplantation of microfragmented adipose tissue is due to the secretory ability of the intact perivascular niche. Stem Cells Translational Medicine 2018;7:876-886.
Collapse
Affiliation(s)
- Bianca Vezzani
- MRC Center for Regenerative MedicineUniversity of EdinburghEdinburghUnited Kingdom
| | - Isaac Shaw
- MRC Center for Regenerative MedicineUniversity of EdinburghEdinburghUnited Kingdom
| | - Hanna Lesme
- MRC Center for Regenerative MedicineUniversity of EdinburghEdinburghUnited Kingdom
| | - Li Yong
- MRC Center for Regenerative MedicineUniversity of EdinburghEdinburghUnited Kingdom
| | - Nusrat Khan
- MRC Center for Regenerative MedicineUniversity of EdinburghEdinburghUnited Kingdom
| | | | - Bruno Péault
- MRC Center for Regenerative MedicineUniversity of EdinburghEdinburghUnited Kingdom
- Orthopaedic Hospital Research Center and Broad Stem Cell Research CenterDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
140
|
Renart J, San Mauro D, Agorreta A, Rutherford K, Gemmell NJ, Quintanilla M. Evolutionary history of the podoplanin gene. GENE REPORTS 2018. [DOI: 10.1016/j.genrep.2018.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
141
|
Grajchen E, Hendriks JJA, Bogie JFJ. The physiology of foamy phagocytes in multiple sclerosis. Acta Neuropathol Commun 2018; 6:124. [PMID: 30454040 PMCID: PMC6240956 DOI: 10.1186/s40478-018-0628-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 11/02/2018] [Indexed: 12/15/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic disease of the central nervous system characterized by massive infiltration of immune cells, demyelination, and axonal loss. Active MS lesions mainly consist of macrophages and microglia containing abundant intracellular myelin remnants. Initial studies showed that these foamy phagocytes primarily promote MS disease progression by internalizing myelin debris, presenting brain-derived autoantigens, and adopting an inflammatory phenotype. However, more recent studies indicate that phagocytes can also adopt a beneficial phenotype upon myelin internalization. In this review, we summarize and discuss the current knowledge on the spatiotemporal physiology of foamy phagocytes in MS lesions, and elaborate on extrinsic and intrinsic factors regulating their behavior. In addition, we discuss and link the physiology of myelin-containing phagocytes to that of foamy macrophages in other disorders such atherosclerosis.
Collapse
Affiliation(s)
- Elien Grajchen
- Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium/School of Life Sciences, Transnationale Universiteit Limburg, Diepenbeek, Belgium
| | - Jerome J A Hendriks
- Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium/School of Life Sciences, Transnationale Universiteit Limburg, Diepenbeek, Belgium
| | - Jeroen F J Bogie
- Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium/School of Life Sciences, Transnationale Universiteit Limburg, Diepenbeek, Belgium.
| |
Collapse
|
142
|
Reis M, Mavin E, Nicholson L, Green K, Dickinson AM, Wang XN. Mesenchymal Stromal Cell-Derived Extracellular Vesicles Attenuate Dendritic Cell Maturation and Function. Front Immunol 2018; 9:2538. [PMID: 30473695 PMCID: PMC6237916 DOI: 10.3389/fimmu.2018.02538] [Citation(s) in RCA: 193] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 10/15/2018] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are potent regulators of immune responses largely through paracrine signaling. MSC secreted extracellular vesicles (MSC-EVs) are increasingly recognized as the key paracrine factors responsible for the biological and therapeutic function of MSCs. We report the first comprehensive study demonstrating the immunomodulatory effect of MSC-EVs on dendritic cell (DC) maturation and function. MSC-EVs were isolated from MSC conditioned media using differential ultracentrifugation. Human monocyte-derived DCs were generated in the absence or presence of MSC-EVs (20 ug/ml) then subjected to phenotypic and functional analysis in vitro. MSC-EV treatment impaired antigen uptake by immature DCs and halted DC maturation resulting in reduced expression of the maturation and activation markers CD83, CD38, and CD80, decreased secretion of pro-inflammatory cytokines IL-6 and IL-12p70 and increased production of anti-inflammatory cytokine TGF-β. MSC-EV treated DCs also demonstrated a diminished CCR 7 expression after LPS stimulation, coupled with a significantly reduced ability to migrate toward the CCR7-ligand CCL21, although they were still able to stimulate allogeneic T cell proliferation in vitro. Through microRNA profiling we have identified 49 microRNAs, which were significantly enriched in MSC-EVs compared to their parent MSCs. MicroRNAs with known effect on DC maturation and functions, including miR-21-5p, miR-142-3p, miR-223-3p, and miR-126-3p, were detected within the top 10 most enriched miRNAs in MSC-EVs, with MiR-21-5p as the third highest expressed miRNA in MSC-EVs. In silico analysis revealed that miR-21-5p targets the CCR7 gene for degradation. To verify these observations, DCs were transfected with miR-21-5p mimics and analyzed for their ability to migrate toward the CCR7-ligand CCL21 in vitro. MiR-21-5p mimic transfected DCs showed a clear trend of reduced CCR7 expression and a significantly decreased migratory ability toward the CCL21. Our findings suggest that MSC-EVs are able to recapitulate MSC mediated DC modulation and MSC-EV enclosed microRNAs may represent a novel mechanism through which MSCs modulate DC functions. As MSCs are currently used in clinical trials to treat numerous diseases associated with immune dysregulation, such as graft-versus-host disease and inflammatory bowel disease, our data provide novel evidence to inform potential future application of MSC-EVs as a cell-free therapeutic agent.
Collapse
Affiliation(s)
- Monica Reis
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Emily Mavin
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Lindsay Nicholson
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Kile Green
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Anne M Dickinson
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Xiao-Nong Wang
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
143
|
CCL19 suppresses angiogenesis through promoting miR-206 and inhibiting Met/ERK/Elk-1/HIF-1α/VEGF-A pathway in colorectal cancer. Cell Death Dis 2018; 9:974. [PMID: 30250188 PMCID: PMC6155262 DOI: 10.1038/s41419-018-1010-2] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/13/2022]
Abstract
The mechanisms underlying the role of chemokines in tumor angiogenesis is still not fully understood. In this study, we detected the influence of CCL19 on colorectal cancer (CRC) angiogenesis. The expression of CCL19 and CD31 in CRC tissues were detected by immunohistochemistry. Human CRC cell lines SW1116 and SW620 stably transfected with CCL19 lentivirus and CCL19 shRNA, and HUVEC stably transfected with CCR7 shRNA were used in our study. Our study showed that CCL19 was significantly low-expressed in CRC tissues and positively related to highly tumor microvessel density. In vitro, we observed that CCL19 high-expressed SW1116 supernatant was able to inhibit proliferation, migration, and sprouting responses of HUVEC, whereas CCL19 low-expressed SW620 supernatant can promote HUVEC angiogenesis. Additionally, we further demonstrated that these functions maybe achieved through promoting miR-206 thus inhibiting Met/ERK/Elk-1/HIF-1α/VEGF-A pathway in a CCR7-dependent manner. Mice angiogenesis model also confirmed that elevated expression of CCL19 inhibit the angiogenesis of CRC in vivo. In summary, our results supported that CCL19 can inhibit CRC angiogenesis through promoting miR-206 thus inhibiting Met/ERK/Elk-1/HIF-1α/VEGF-A pathway. This may be a novel therapeutic option for anti-vascular treatment in CRC.
Collapse
|
144
|
Drakes ML, Stiff PJ. Regulation of Ovarian Cancer Prognosis by Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2018; 10:E302. [PMID: 30200478 PMCID: PMC6162424 DOI: 10.3390/cancers10090302] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/20/2022] Open
Abstract
It is estimated that in the United States in 2018 there will be 22,240 new cases of ovarian cancer and 14,070 deaths due to this malignancy. The most common subgroup of this disease is high-grade serous ovarian cancer (HGSOC), which is known for its aggressiveness, high recurrence rate, metastasis to other sites, and the development of resistance to conventional therapy. It is important to understand the ovarian cancer tumor microenvironment (TME) from the viewpoint of the function of pre-existing immune cells, as immunocompetent cells are crucial to mounting robust antitumor responses to prevent visible tumor lesions, disease progression, or recurrence. Networks consisting of innate and adaptive immune cells, metabolic pathways, intracellular signaling molecules, and a vast array of soluble factors, shape the pathogenic nature of the TME and are useful prognostic indicators of responses to conventional therapy and immunotherapy, and subsequent survival rates. This review highlights key immune cells and soluble molecules in the TME of ovarian cancer, which are important in the development of effective antitumor immunity, as well as those that impair effector T cell activity. A more insightful knowledge of the HGSOC TME will reveal potential immune biomarkers to aid in the early detection of this disease, as well as biomarkers that may be targeted to advance the design of novel therapies that induce potent antitumor immunity and survival benefit.
Collapse
Affiliation(s)
- Maureen L Drakes
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| | - Patrick J Stiff
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| |
Collapse
|
145
|
Regulation of Ovarian Cancer Prognosis by Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2018. [PMID: 30200478 DOI: 10.3390/cancers10090302]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
It is estimated that in the United States in 2018 there will be 22,240 new cases of ovarian cancer and 14,070 deaths due to this malignancy. The most common subgroup of this disease is high-grade serous ovarian cancer (HGSOC), which is known for its aggressiveness, high recurrence rate, metastasis to other sites, and the development of resistance to conventional therapy. It is important to understand the ovarian cancer tumor microenvironment (TME) from the viewpoint of the function of pre-existing immune cells, as immunocompetent cells are crucial to mounting robust antitumor responses to prevent visible tumor lesions, disease progression, or recurrence. Networks consisting of innate and adaptive immune cells, metabolic pathways, intracellular signaling molecules, and a vast array of soluble factors, shape the pathogenic nature of the TME and are useful prognostic indicators of responses to conventional therapy and immunotherapy, and subsequent survival rates. This review highlights key immune cells and soluble molecules in the TME of ovarian cancer, which are important in the development of effective antitumor immunity, as well as those that impair effector T cell activity. A more insightful knowledge of the HGSOC TME will reveal potential immune biomarkers to aid in the early detection of this disease, as well as biomarkers that may be targeted to advance the design of novel therapies that induce potent antitumor immunity and survival benefit.
Collapse
|
146
|
Drakes ML, Stiff PJ. Regulation of Ovarian Cancer Prognosis by Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2018. [PMID: 30200478 DOI: 10.3390/cancers10090302] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
It is estimated that in the United States in 2018 there will be 22,240 new cases of ovarian cancer and 14,070 deaths due to this malignancy. The most common subgroup of this disease is high-grade serous ovarian cancer (HGSOC), which is known for its aggressiveness, high recurrence rate, metastasis to other sites, and the development of resistance to conventional therapy. It is important to understand the ovarian cancer tumor microenvironment (TME) from the viewpoint of the function of pre-existing immune cells, as immunocompetent cells are crucial to mounting robust antitumor responses to prevent visible tumor lesions, disease progression, or recurrence. Networks consisting of innate and adaptive immune cells, metabolic pathways, intracellular signaling molecules, and a vast array of soluble factors, shape the pathogenic nature of the TME and are useful prognostic indicators of responses to conventional therapy and immunotherapy, and subsequent survival rates. This review highlights key immune cells and soluble molecules in the TME of ovarian cancer, which are important in the development of effective antitumor immunity, as well as those that impair effector T cell activity. A more insightful knowledge of the HGSOC TME will reveal potential immune biomarkers to aid in the early detection of this disease, as well as biomarkers that may be targeted to advance the design of novel therapies that induce potent antitumor immunity and survival benefit.
Collapse
Affiliation(s)
- Maureen L Drakes
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| | - Patrick J Stiff
- Cardinal Bernardin Cancer Center, Department of Medicine, Loyola University Chicago, Building 112, 2160 South First Avenue, Maywood, IL 60153, USA.
| |
Collapse
|
147
|
St Clair EW, Baer AN, Wei C, Noaiseh G, Parke A, Coca A, Utset TO, Genovese MC, Wallace DJ, McNamara J, Boyle K, Keyes-Elstein L, Browning JL, Franchimont N, Smith K, Guthridge JM, Sanz I, James JA. Clinical Efficacy and Safety of Baminercept, a Lymphotoxin β Receptor Fusion Protein, in Primary Sjögren's Syndrome: Results From a Phase II Randomized, Double-Blind, Placebo-Controlled Trial. Arthritis Rheumatol 2018; 70:1470-1480. [PMID: 29604186 PMCID: PMC6115299 DOI: 10.1002/art.40513] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 03/22/2018] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To evaluate the clinical efficacy and safety of baminercept, a lymphotoxin β receptor IgG fusion protein (LTβR-Ig), for the treatment of primary Sjögren's syndrome (SS), and to explore the possible mechanisms of action of this treatment. METHODS In this multicenter trial, 52 patients with primary SS were randomized in a 2:1 ratio to receive subcutaneous injections of 100 mg of baminercept every week for 24 weeks or matching placebo. The primary end point was the change between screening and week 24 in the stimulated whole salivary flow (SWSF) rate. Secondary end points included the European League Against Rheumatism Sjögren's Syndrome Disease Activity Index (ESSDAI), as well as measurements of select chemokines and cytokines and enumeration of peripheral blood B and T cell subsets. RESULTS The change from baseline to week 24 in the SWSF rate was not significantly different between the baminercept and placebo treatment groups (baseline-adjusted mean change -0.01 versus 0.07 ml/minute; P = 0.332). The change in the ESSDAI during treatment was also not significantly different between the treatment groups (baseline-adjusted mean change -1.23 versus -0.15; P = 0.104). Although the incidence of adverse events was similar between the treatment groups, baminercept therapy was associated with a higher incidence of liver toxicity, including 2 serious adverse events. Baminercept also produced a significant decrease in plasma levels of CXCL13 and significant changes in the number of circulating B and T cells, consistent with its known inhibitory effects on LTβR signaling. CONCLUSION In this trial, treatment with baminercept failed to significantly improve glandular and extraglandular disease in patients with primary SS, despite evidence from mechanistic studies showing that it blocks LTβR signaling.
Collapse
Affiliation(s)
| | - Alan N Baer
- Johns Hopkins University, Baltimore, Maryland
| | - Chungwen Wei
- Ignacio Sanz, Emory University, Atlanta, Georgia
| | | | - Anne Parke
- Saint Francis Medical Group, Hartford, Connecticut
| | | | | | | | | | - James McNamara
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | | | | | | | | | | | | | - Ignacio Sanz
- Ignacio Sanz, Emory University, Atlanta, Georgia
| | | |
Collapse
|
148
|
Zhang JF, Li Y, Zhang AZ, He QQ, Du YC, Cao W. Expression and pathological significance of CC chemokine receptor 7 and its ligands in the airway of asthmatic rats exposed to cigarette smoke. J Thorac Dis 2018; 10:5459-5467. [PMID: 30416795 DOI: 10.21037/jtd.2018.08.124] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Cigarette smoking aggravates the symptoms of asthma, leading to the rapid decline of lung function. Dendritic cells (DCs) and lymphocytes are considered initiating and promoting factors for the airway inflammation reactions of asthma. In addition, activation of CC chemokine receptor 7 (CCR7) by chemokine (C-C motif) ligand (CCL) 19 and 21 promotes DCs and T cells migration to lymphoid tissues during inflammation. We aimed to examine how cigarette smoke affects the expression of CCR7 in the lungs of asthmatic rats and explore the signaling mechanism linking CCR7 expression to exacerbation of symptoms. Methods Forty Wistar rats were randomized to four groups: control, asthma, smoke exposure, and asthma with smoke exposure groups. A rat asthma model was established by intraperitoneal ovalbumin injection. CCR7 expression was examined with immunohistochemistry and western blotting. The number of airway DCs was determined by OX62 immunohistochemistry. Interferon (INF)-γ, interleukin (IL)-4, CCL19, and CCL21 expression levels in blood and bronchioalveolar lavage fluid (BALF) were determined by enzyme-linked immunosorbent assays (ELISAs). Results Tissue CCR7 expression, peripheral blood and BALF CCL19 and CCL21 concentrations, and the number of airway DCs were significantly higher in the asthma with smoke exposure group than the asthma group (P<0.01). In addition, INF-γ expression was decreased and IL-4 increased in the asthma and asthma with smoke exposure groups compared with the control group (P<0.01), and in the asthma with smoke exposure group compared with the asthma group (P<0.01). Expression of CCR7 correlated negatively with INF-γ expression in peripheral blood and BALF (P<0.01), and positively with the airway DCs and IL-4 expression in the peripheral blood and BALF (P<0.01). Conclusions Cigarette smoking may aggravate asthma symptoms by attenuating immunity, possibly through CCR7-mediated DCs aggregation in lung tissue.
Collapse
Affiliation(s)
- Jun-Feng Zhang
- Department of Health Statistics, Public Health of Shanxi Medical University, Taiyuan 030001, China.,Publishing house, Chinese Journal of Rheumatology, Taiyuan 030001, China
| | - Yi Li
- Department of Respiration Medicine, People's Hospital of Shanxi Province, Taiyuan 030001, China
| | - Ai-Zhen Zhang
- Department of Respiration Medicine, People's Hospital of Shanxi Province, Taiyuan 030001, China
| | - Qian-Qian He
- Department of Health Statistics, Public Health of Shanxi Medical University, Taiyuan 030001, China
| | - Yong-Cheng Du
- Department of Respiration Medicine, People's Hospital of Shanxi Province, Taiyuan 030001, China
| | - Wen Cao
- Department of Respiration Medicine, People's Hospital of Shanxi Province, Taiyuan 030001, China
| |
Collapse
|
149
|
Hoffmann-Vold AM, Hesselstrand R, Fretheim H, Ueland T, Andreassen AK, Brunborg C, Palchevskiy V, Midtvedt Ø, Garen T, Aukrust P, Belperio JA, Molberg Ø. CCL21 as a Potential Serum Biomarker for Pulmonary Arterial Hypertension in Systemic Sclerosis. Arthritis Rheumatol 2018; 70:1644-1653. [PMID: 29687634 DOI: 10.1002/art.40534] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 04/17/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Systemic sclerosis (SSc) is a major cause of pulmonary arterial hypertension (PAH). Murine models indicate key roles for chemokines CCL19 and CCL21 and their receptor CCR7 in lung inflammation leading to PAH. The objective of this study was to assess the chemokine CCL19-CCL21 axis in patients with SSc-related PAH. METHODS Serum samples obtained from 2 independent prospective SSc cohorts (n = 326), patients with idiopathic PAH (n = 12), and healthy control subjects (n = 100) were analyzed for CCL19/CCL21 levels, by enzyme-linked immunosorbent assay. The levels were defined as either high or low, using the mean + 2 SD value in controls as the cutoff value. Risk stratification at the time of PAH diagnosis and PAH-related events were performed. Descriptive and Cox regression analyses were conducted. RESULTS CCL21 levels were higher in patients with SSc compared with controls and were elevated prior to the diagnosis of PAH. PAH was more frequent in patients with high CCL21 levels (≥0.4 ng/ml) than in those with low CCL21 levels (33.3% versus 5.3% [P < 0.001]). In multivariate analyses, CCL21 was associated with PAH (hazard ratio [HR] 5.1, 95% CI 2.39-10.76 [P < 0.001]) and occurrence of PAH-related events (HR 4.7, 95% CI 2.12-10.46, P < 0.001). Risk stratification at the time of PAH diagnosis alone did not predict PAH-related events. However, when risk at diagnosis was combined with high or low CCL21 level, there was a significant predictive effect (HR 1.3, 95% CI 1.03-1.60 [P = 0.027]). A high CCL21 level was associated with decreased survival (P < 0.001). CONCLUSION CCL21 appears to be a promising marker for predicting the risk of SSc-related PAH and PAH progression. CCL21 may be part of a dysregulated immune pathway linked to the development of lung vascular damage in SSc.
Collapse
Affiliation(s)
| | | | - Håvard Fretheim
- Oslo University Hospital, Rikshospitalet, and University of Oslo, Oslo, Norway
| | - Thor Ueland
- Oslo University Hospital, Rikshospitalet, and University of Oslo, Oslo, Norway
| | | | | | | | | | - Torhild Garen
- Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Pål Aukrust
- Oslo University Hospital, Rikshospitalet, and University of Oslo, Oslo, Norway
| | - John A Belperio
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Øyvind Molberg
- Oslo University Hospital, Rikshospitalet, and University of Oslo, Oslo, Norway
| |
Collapse
|
150
|
Chung WS, Lin CL, Hsu CY. Women who had appendectomy have increased risk of systemic lupus erythematosus: a nationwide cohort study. Clin Rheumatol 2018; 37:3009-3016. [PMID: 29971583 DOI: 10.1007/s10067-018-4192-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 06/12/2018] [Accepted: 06/25/2018] [Indexed: 12/18/2022]
Abstract
The appendix is involved in immune function, and an appendectomy may alter the immune system. Studies evaluating the relationship between previous appendectomy and the risk of systemic lupus erythematosus (SLE) are lacking. This nationwide cohort study investigated the incidence and risk of SLE in patients who underwent appendectomy. Patients aged > 20 years who received appendectomy from 2000 to 2011 were identified from the National Health Insurance Research Database and assigned to the appendectomy cohort. Patients without appendectomy were randomly selected from the NHIRD and assigned to the control cohort; they were frequency matched to each study patient at a 4:1 ratio by sex, age, and index year. All patients were followed until SLE diagnosis, withdrawal from the National Health Insurance program, or the end of 2011. We used Cox models to estimate the hazard ratio (HR) and 95% confidence interval (CI) to compare the risk of SLE between the appendectomy and control cohorts. From 23.74 million people in the cohort, 80,582 patients undergoing appendectomy and 323,850 patients without appendectomy were followed for 723,438 and 2,931,737 person-years, respectively. The appendectomy cohort had a 2.04-fold higher risk of SLE than the control cohort (adjusted HR = 2.04, 95% CI = 1.52-2.76). Women aged ≤ 49 years who underwent appendectomy had a 2.27-fold higher risk of SLE than the corresponding controls (adjusted HR = 2.27, 95% CI = 1.62-3.19). Women aged ≤ 49 years who underwent appendectomy have a significantly higher risk of SLE.
Collapse
Affiliation(s)
- Wei-Sheng Chung
- Department of Internal Medicine, Taichung Hospital, Ministry of Health and Welfare, No. 199, Section 1, San-Min Road, Taichung, 40343, Taiwan. .,Department of Health Services Administration, China Medical University, Taichung, Taiwan. .,Department of Healthcare Administration, Central Taiwan University of Science and Technology, Taichung, Taiwan.
| | - Cheng-Li Lin
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan
| | - Chung-Y Hsu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|