1
|
Kim JH, Lee D, Hall K, Jo H, Bannantine JP, Davis WC, de Souza C. Major membrane protein of Mycobacterium avium subp. paratuberculosis activates immune and autophagic pathways in bovine monocyte-derived macrophages. Vet Immunol Immunopathol 2025; 282:110901. [PMID: 40015107 DOI: 10.1016/j.vetimm.2025.110901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/01/2025]
Abstract
Mycobacterium avium subspecies paratuberculosis (Map), the etiological agent of Johne's disease in ruminants, poses challenges to veterinary health and food safety. Despite an immune response that partially controls early infection, Map persists in macrophages through mechanisms not well understood. Here, we explored how the Map major membrane protein (MMP) modulates immune pathways in bovine monocyte-derived macrophages (MoMΦs). MMP is a key component of the bacterial cell membrane recognized in cattle with Johne's disease, making it a critical antigenic target for immune studies. Using high-resolution transcriptomics, we identified that MMP stimulation rapidly activates genes linked to pro-inflammatory cytokine signaling, antigen processing, and presentation via MHC I and II pathways. Gene Ontology and KEGG pathway enrichment analyses highlighted upregulation of TNF, IL-17, and NF-κB signaling cascades, suggesting an immune signaling that may foster cytotoxic T cell development. Phosphorylation assays confirmed that MMP triggers MAPK activation within minutes, implicating both p38 and JNK1/2 in early macrophage responses. Machine learning approaches revealed subtle yet significant MMP-specific gene signatures including ATG5 and ATG12, implicated in autophagosome assembly. These findings point to a dynamic interplay between antibacterial autophagy and immunostimulatory pathways elicited by MMP in bovine macrophages. Importantly, our results suggest the relevance of MMP as a potential vaccine target, as it not only elicits immune-activating signals but also engages host defenses critical to restricting Map survival. Overall, this work provides an ex vivo framework for delineating the molecular underpinnings of Map infection, offering new insights into macrophage-based immunity and informing development of novel therapeutic and prophylactic strategies against paratuberculosis. Our data open avenues for translational studies, illuminating the interplay between MMP, macrophages, and protective host immunity.
Collapse
Affiliation(s)
- Jong Hyuk Kim
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA; UF Health Cancer Center, University of Florida, Gainesville, FL, USA; Artificial Intelligence Academic Initiative (AI2) Center, University of Florida, Gainesville, FL, USA; Intelligent Critical Care Center (IC3), University of Florida, Gainesville, FL, USA
| | - Donghee Lee
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Kevin Hall
- Department of Comparative, Diagnostic & Population Medicine, College of Veterinary Medicine, Gainesville, University of Florida, FL, USA
| | - Hyunji Jo
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | | | - William C Davis
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Cleverson de Souza
- Artificial Intelligence Academic Initiative (AI2) Center, University of Florida, Gainesville, FL, USA; Department of Comparative, Diagnostic & Population Medicine, College of Veterinary Medicine, Gainesville, University of Florida, FL, USA.
| |
Collapse
|
2
|
Van Roy Z, Kak G, Fallet RW, Kielian T. Interferon-gamma receptor signaling regulates innate immunity during Staphylococcus aureus craniotomy infection. J Neuroinflammation 2025; 22:46. [PMID: 39987156 PMCID: PMC11847343 DOI: 10.1186/s12974-025-03376-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/13/2025] [Indexed: 02/24/2025] Open
Abstract
A craniotomy is a neurosurgical procedure performed to access the intracranial space. In 3-5% of cases, infections can develop, most caused by Staphylococcus aureus biofilm formation on the skull surface. Medical management of this infection is difficult, as biofilm properties confer immune and antimicrobial recalcitrance to the infection and necessitate additional surgical procedures. Furthermore, treatment failure rates can be appreciably high. These factors, compounded with rapidly expanding rates of antimicrobial resistance, highlight the need to develop alternative treatment strategies to target and reverse the immune dysfunction that occurs during biofilm infection. Our recent work has identified CD4+ Th1 and Th17 cells as potent regulators of innate immune cell activation during craniotomy infection. Here, we report the role of IFN-γ, versus other Th1- and Th17-derived cytokines, in programing the immune response to biofilm infection using both global and cell type-specific IFN-γR1-deficient (Ifngr1-/-) mice. Bacterial burdens were significantly higher in Ifngr1-/- relative to WT animals despite few changes in immune cell abundance. Single-cell transcriptomics identified candidate explanations for this phenotype as alterations in cell death pathways, innate immune cell activation, MHC-II expression, and T cell responses were significantly reduced in Ifngr1-/- mice. While caspase-1 activation in PMNs and macrophage/microglial MHC-II expression were regulated by IFN-γ signaling, no phenotypes were observed with either granulocyte- or macrophage/microglia Ifngr1-/- conditional knockout mice, suggestive of redundancy. Instead, a decreased Th1/Th17 ratio was identified in Ifngr1-/- animals that was corroborated by elevated IL-17 levels and correlated with dysfunctional T cell-innate immune communication. Further, Th17 cells were less effective than Th1 cells in promoting S. aureus bactericidal activity in microglia and macrophages. Collectively, this work identifies a key protective role for IFN-γ during craniotomy infection by enhancing macrophage and microglial antibacterial activity. Therefore, controlled programming of IFN-γ responses may represent a novel therapeutic strategy for chronic craniotomy infections.
Collapse
Affiliation(s)
- Zachary Van Roy
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska, 68198, USA
| | - Gunjan Kak
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska, 68198, USA
| | - Rachel W Fallet
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska, 68198, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska, 68198, USA.
| |
Collapse
|
3
|
Trojan A, Lone YC, Briceno I, Trojan J. Anti-Gene IGF-I Vaccines in Cancer Gene Therapy: A Review of a Case of Glioblastoma. Curr Med Chem 2024; 31:1983-2002. [PMID: 38031775 DOI: 10.2174/0109298673237968231106095141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 06/27/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023]
Abstract
OBJECTIVE Vaccines for the deadliest brain tumor - glioblastoma (GBM) - are generally based on targeting growth factors or their receptors, often using antibodies. The vaccines described in the review were prepared to suppress the principal cancer growth factor - IGF-I, using anti-gene approaches either of antisense (AS) or of triple helix (TH) type. Our objective was to increase the median survival of patients treated with AS and TH cell vaccines. METHODOLOGY The cells were transfected in vitro by both constructed IGF-I AS and IGF-I TH expression episomal vectors; part of these cells was co-cultured with plant phytochemicals, modulating IGF-I expression. Both AS and TH approaches completely suppressed IGF-I expression and induced MHC-1 / B7 immunogenicity related to the IGF-I receptor signal. RESULTS This immunogenicity proved to be stronger in IGF-I TH than in IGF-I AS-prepared cell vaccines, especially in TH / phytochemical cells. The AS and TH vaccines generated an important TCD8+ and TCD8+CD11b- immune response in treated GBM patients and increased the median survival of patients up to 17-18 months, particularly using TH vaccines; in some cases, 2- and 3-year survival was reported. These clinical results were compared with those obtained in therapies targeting other growth factors. CONCLUSION The anti-gene IGF-I vaccines continue to be applied in current GBM personalized medicine. Technical improvements in the preparation of AS and TH vaccines to increase MHC-1 and B7 immunogenicity have, in parallel, allowed to increase in the median survival of patients.
Collapse
Affiliation(s)
- Annabelle Trojan
- INSERM UMR 1197, Cancer Center & University of Paris / Saclay, PO Box: 94802 Villejuif, France
- Faculty of Medicine, University of Cartagena, PO Box: 130014 Cartagena de Indias, Colombia
| | - Yu-Chun Lone
- INSERM UMR 1197, Cancer Center & University of Paris / Saclay, PO Box: 94802 Villejuif, France
- CEDEA / ICGT - Center of Oncological Diseases Diagnosis, PO Box: 110231 Bogota, Colombia
| | - Ignacio Briceno
- Faculty of Medicine, University of La Sabana, PO Box: 250008 Chia, Colombia
| | - Jerzy Trojan
- INSERM UMR 1197, Cancer Center & University of Paris / Saclay, PO Box: 94802 Villejuif, France
- CEDEA / ICGT - Center of Oncological Diseases Diagnosis, PO Box: 110231 Bogota, Colombia
- National Academy of Medicine - ANM, PO Box: 75272 Paris, France
| |
Collapse
|
4
|
Tojjari A, Saeed A, Sadeghipour A, Kurzrock R, Cavalcante L. Overcoming Immune Checkpoint Therapy Resistance with SHP2 Inhibition in Cancer and Immune Cells: A Review of the Literature and Novel Combinatorial Approaches. Cancers (Basel) 2023; 15:5384. [PMID: 38001644 PMCID: PMC10670368 DOI: 10.3390/cancers15225384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 10/13/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
SHP2 (Src Homology 2 Domain-Containing Phosphatase 2) is a protein tyrosine phosphatase widely expressed in various cell types. SHP2 plays a crucial role in different cellular processes, such as cell proliferation, differentiation, and survival. Aberrant activation of SHP2 has been implicated in multiple human cancers and is considered a promising therapeutic target for treating these malignancies. The PTPN11 gene and functions encode SHP2 as a critical signal transduction regulator that interacts with key signaling molecules in both the RAS/ERK and PD-1/PD-L1 pathways; SHP2 is also implicated in T-cell signaling. SHP2 may be inhibited by molecules that cause allosteric (bind to sites other than the active site and attenuate activation) or orthosteric (bind to the active site and stop activation) inhibition or via potent SHP2 degraders. These inhibitors have anti-proliferative effects in cancer cells and suppress tumor growth in preclinical models. In addition, several SHP2 inhibitors are currently in clinical trials for cancer treatment. This review aims to provide an overview of the current research on SHP2 inhibitors, including their mechanism of action, structure-activity relationships, and clinical development, focusing on immune modulation effects and novel therapeutic strategies in the immune-oncology field.
Collapse
Affiliation(s)
- Alireza Tojjari
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Anwaar Saeed
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Arezoo Sadeghipour
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modarres University, Tehran P.O. Box 14115-175, Iran
| | - Razelle Kurzrock
- Department of Medicine, Genome Sciences and Precision Medicine Center, Medical College of Wisconsin Cancer Center, Milwaukee, WI 53226, USA
| | | |
Collapse
|
5
|
Khan I, Siraj M. An updated review on cell signaling pathways regulated by candidate miRNAs in coronary artery disease. Noncoding RNA Res 2023; 8:326-334. [PMID: 37077752 PMCID: PMC10106733 DOI: 10.1016/j.ncrna.2023.03.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/16/2023] [Accepted: 03/29/2023] [Indexed: 03/31/2023] Open
Abstract
MicroRNAs (miRNAs) are small endogenous non-coding RNA, size range from 17 to 25 nucleotides that regulate gene expression at the post-transcriptional level. More than 2000 different types of miRNAs have been identified in humans which regulate about 60% of gene expression, since the discovery of the first miRNA in 1993. MicroRNA performs many functions such as being involved in the regulation of various biological pathways for example cell migration, cell proliferation, cell differentiation, disease progression, and initiation. miRNAs also play an important role in the development of atherosclerosis lesions, cardiac fibroblast, cardiac hypertrophy, cancer, and neurological disorders. Abnormal activation of many cell signaling pathways has been observed in the development of coronary artery disease. Abnormal expression of these candidate miRNA genes leads to up or downregulation of specific genes, these specific genes play an important role in the regulation of cell signaling pathways involved in coronary artery disease. Many studies have found that miRNAs play a key role in the regulation of crucial signaling pathways that are involved in the pathophysiology of coronary artery disease. This review is designed to investigate the role of cell signaling pathways regulated by candidate miRNAs in Coronary artery disease.
Collapse
|
6
|
Farriol-Duran R, Vallejo-Vallés M, Amengual-Rigo P, Floor M, Guallar V. NetCleave: An Open-Source Algorithm for Predicting C-Terminal Antigen Processing for MHC-I and MHC-II. Methods Mol Biol 2023; 2673:211-226. [PMID: 37258917 DOI: 10.1007/978-1-0716-3239-0_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
T cell epitopes presented on the surface of mammalian cells are subjected to a complex network of antigen processing and presentation. Among them, C-terminal antigen processing constitutes one of the main bottlenecks for the generation of epitopes, as it defines the C-terminal end of the final epitope and delimits the peptidome that will be presented downstream. Previously (Amengual-Rigo and Guallar, Sci Rep 111(11):1-8, 2021), we demonstrated that NetCleave stands out as one of the best algorithms for the prediction of C-terminal processing, which in its turn can be crucial to design peptide-based vaccination strategies. In this chapter, we provide a pipeline to exploit the full capabilities of NetCleave, an open-source and retrainable algorithm for predicting the C-terminal antigen processing for the MHC-I and MHC-II pathways.
Collapse
Affiliation(s)
| | | | | | - Martin Floor
- Barcelona Supercomputing Center (BSC), Barcelona, Spain
| | - Víctor Guallar
- Barcelona Supercomputing Center (BSC), Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
7
|
Yu J, Wu X, Song J, Zhao Y, Li H, Luo M, Liu X. Loss of MHC-I antigen presentation correlated with immune checkpoint blockade tolerance in MAPK inhibitor-resistant melanoma. Front Pharmacol 2022; 13:928226. [PMID: 36091815 PMCID: PMC9459091 DOI: 10.3389/fphar.2022.928226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/27/2022] [Indexed: 11/20/2022] Open
Abstract
Immune checkpoint blockade and MAPK-targeted combined therapy is a promising regimen for advanced melanoma patients. However, the clinical benefit from this combo regimen remains limited, especially in patients who acquired resistance to MAPK-targeted therapy. Here, we systematically characterized the immune landscape during MAPK-targeted therapy in patients and mouse melanoma models. We observed that both the abundance of tumor-infiltrated T cells and the expression of immune-related genes were upregulated in the drug-responsive period, but downregulated in the resistance period, implying that acquired drug resistance dampens the antitumor immune response. Further transcriptomic dissection indicated that loss of MHC-I antigen presentation on tumor cells plays a critical role in the reduction of T cell infiltration during drug resistance. Survival analysis demonstrates that loss of antigen presentation and reduction of T-cell infiltration during acquired drug resistance are associated with poorer clinical response and prognosis of anti-PD-1 therapy in melanoma patients. In addition, we identified that alterations in the MAPK inhibitor resistance-related oncogenic signaling pathway closely correlated with deficiency of MHC-I antigen presentation, including activation of the PI3K-mTOR, MAPK, and Wnt pathways. In conclusion, our research illuminates that decreased infiltration of T cells is associated with acquired drug resistance during MAPK-targeted therapy, which may underlie the cross-resistance to immune checkpoint blockade.
Collapse
Affiliation(s)
- Jing Yu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Xi Wu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Jinen Song
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Yujie Zhao
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Huifang Li
- Research Core Facility, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Min Luo
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
- *Correspondence: Xiaowei Liu, ; Min Luo,
| | - Xiaowei Liu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
- *Correspondence: Xiaowei Liu, ; Min Luo,
| |
Collapse
|
8
|
Khan MA, Malik A, Alruwetei A, Alzohairy MA, Alhatlani BY, Al Rugaie O, Alhumaydhi FA, Khan A. Delivery of MERS antigen encapsulated in α-GalCer-bearing liposomes elicits stronger antigen-specific immune responses. J Drug Target 2022; 30:884-893. [PMID: 35418263 DOI: 10.1080/1061186x.2022.2066681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alpha-Galactosylceramide (α-GalCer) effectively activates the natural killer T (NKT) cells to secrete remarkable amounts of Th1 and Th2 cytokines and therefore, acts as a potential immunoadjuvant in vaccine formulation. In the present study, we prepared α-GalCer-bearing or α-GalCer-free liposomes and loaded them with Middle East Respiratory Syndrome Corona virus papain-like protease (α-GalCer-Lip-MERS-CoV PLpro or Lip- MERS-CoV PLpro). These formulations were injected in mice to investigate the antigen-specific humoral and cell-mediated immune responses. The immunization with α-GalCer-Lip-MERS-CoV PLpro or Lip- MERS-CoV PLpro did not induce any notable toxicity in immunized mice. The results demonstrated that mice immunized with α-GalCer-Lip-MERS-CoV PLpro showed greater antigen-specific antibody titer, switching of IgG isotyping to IgG2a subclass and higher lymphocyte proliferation. Moreover, the splenocytes from α-GalCer-Lip-MERS-CoV PLpro immunized mice secreted greater levels of IFN-γ, IL-4, IL-2 and IL-12. Interestingly, a booster dose induced stronger memory immune responses in mice previously immunized with α-GalCer-Lip-MERS-CoV PLpro. In summary, α-GalCer-Lip-MERS-CoV PLpro may prove to be a promising vaccine formulation to protect the individuals against MERS-CoV infection.
Collapse
Affiliation(s)
- Masood Alam Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Ajamaluddin Malik
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Abdulmohsen Alruwetei
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Mohammad A Alzohairy
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Bader Y Alhatlani
- Department of Applied Medical Sciences, Applied College in Unayzah, Qassim University, Unayzah, Saudi Arabia
| | - Osamah Al Rugaie
- Department of Basic Medical Sciences, College of Medicine and Medical Sciences, Qassim University, Unayzah, Saudi Arabia
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Arif Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
9
|
Abstract
Blood vessel endothelial cells (ECs) have long been known to modulate inflammation by regulating immune cell trafficking, activation status and function. However, whether the heterogeneous EC populations in various tissues and organs differ in their immunomodulatory capacity has received insufficient attention, certainly with regard to considering them for alternative immunotherapy. Recent single-cell studies have identified specific EC subtypes that express gene signatures indicative of phagocytosis or scavenging, antigen presentation and immune cell recruitment. Here we discuss emerging evidence suggesting a tissue-specific and vessel type-specific immunomodulatory role for distinct subtypes of ECs, here collectively referred to as 'immunomodulatory ECs' (IMECs). We propose that IMECs have more important functions in immunity than previously recognized, and suggest that these might be considered as targets for new immunotherapeutic approaches.
Collapse
|
10
|
Nazary Abrbekoh F, Salimi L, Saghati S, Amini H, Fathi Karkan S, Moharamzadeh K, Sokullu E, Rahbarghazi R. Application of microneedle patches for drug delivery; doorstep to novel therapies. J Tissue Eng 2022; 13:20417314221085390. [PMID: 35516591 PMCID: PMC9065468 DOI: 10.1177/20417314221085390] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/17/2022] [Indexed: 12/14/2022] Open
Abstract
In the past decade, microneedle-based drug delivery systems showed promising approaches to become suitable and alternative for hypodermic injections and can control agent delivery without side effects compared to conventional approaches. Despite these advantages, the procedure of microfabrication is facing some difficulties. For instance, drug loading method, stability of drugs, and retention time are subjects of debate. Besides, the application of novel refining fabrication methods, types of materials, and instruments are other issues that need further attention. Herein, we tried to summarize recent achievements in controllable drug delivery systems (microneedle patches) in vitro and in vivo settings. In addition, we discussed the influence of delivered drugs on the cellular mechanism and immunization molecular signaling pathways through the intradermal delivery route. Understanding the putative efficiency of microneedle patches in human medicine can help us develop and design sophisticated therapeutic modalities.
Collapse
Affiliation(s)
| | - Leila Salimi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Saghati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hassan Amini
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sonia Fathi Karkan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Keyvan Moharamzadeh
- Hamdan Bin Mohammed College of Dental Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Emel Sokullu
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
Cañas CA, Castaño-Valencia S, Castro-Herrera F. Pharmacological blockade of KV1.3 channel as a promising treatment in autoimmune diseases. J Transl Autoimmun 2022; 5:100146. [PMID: 35146402 PMCID: PMC8818563 DOI: 10.1016/j.jtauto.2022.100146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/20/2022] [Accepted: 01/22/2022] [Indexed: 11/27/2022] Open
Abstract
There are more than 100 autoimmune diseases (AD), which have a high prevalence that ranges between 5% and 8% of the general population. Type I diabetes mellitus, multiple sclerosis, systemic lupus erythematosus and rheumatoid arthritis remain the health problem of highest concern among people worldwide due to its high morbidity and mortality. The development of new treatment strategies has become a research hotspot. In recent years, the study of the ion channels presents in the cells of the immune system, regarding their functional role, the consequences of mutations in their genes and the different ways of blocking them are the subject of intense research. Pharmacological blockade of KV1.3 channel inhibits Ca2+ signaling, T cell proliferation, and pro-inflammatory interleukins production in human CD4+ effector memory T cells. These cells mediated most of the AD and their inhibition is a promising therapeutic target. In this review, we will highlight the biological function of KV1.3 channel in T cells, consequence of the pharmacological inhibition (through anemone and scorpion toxins, synthetic peptides, nanoparticles, or monoclonal antibodies) as well as the possible therapeutical application in AD.
Collapse
Affiliation(s)
- Carlos A. Cañas
- Universidad Icesi, CIRAT, Centro de Investigación en Reumatología, Autoinmunidad y Medicina Traslacional, Cali, Colombia
- Unit of Rheumatology, Fundación Valle del Lili, Cali, Colombia
- Corresponding author. Rheumatology Unit, Fundación Valle del Lili, Cra. 98 18-49, Cali, 760032, Colombia.
| | - Santiago Castaño-Valencia
- Department of Physiological Sciences, Department of Health Sciences, Universidad del Valle, Cali, Colombia
| | - Fernando Castro-Herrera
- Department of Physiological Sciences, Department of Health Sciences, Universidad del Valle, Cali, Colombia
| |
Collapse
|
12
|
Veletic I, Prijic S, Manshouri T, Nogueras-Gonzalez GM, Verstovsek S, Estrov Z. Altered T-cell subset repertoire affects treatment outcome of patients with myelofibrosis. Haematologica 2021; 106:2384-2396. [PMID: 32732359 PMCID: PMC8409049 DOI: 10.3324/haematol.2020.249441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Indexed: 12/13/2022] Open
Abstract
Phenotypic characterization of T cells in myelofibrosis is intriguing because of increased inflammation, markedly elevated pro-inflammatory cytokines, and altered distribution of T-cell subsets. Constitutive activation of Janus kinase-2 (JAK2) in the majority of patients with myelofibrosis contributes to the expression of the programmed cell death protein-1 (PD1) and T-cell exhaustion. We wondered whether T-cell activation affects treatment outcome of patients with myelofibrosis and sought to determine whether the JAK1/2 inhibitor ruxolitinib affects the activation of T-cell subsets. T cells from 47 myelofibrosis patients were analyzed and the percentages of either helper (CD4+) or cytotoxic (CD8+) naïve, central memory, effector memory, or effector T cells; and fractions of PD1-expressing cells in each subset were assessed. Higher numbers of T cells co-expressing CD4/PD1 and CD8/PD1 were found in myelofibrosis patients than in healthy controls (n=28), and the T cells were significantly skewed toward an effector phenotype in both CD4+ and CD8+ subsets, consistent with a shift from a quiescent to an activated state. Over the course of ruxolitinib treatment, the distribution of aberrant T-cell subsets significantly reversed towards resting cell phenotypes. CD4+ and CD8+ subsets at baseline correlated with monocyte and platelet counts, and their PD1+ fractions correlated with leukocyte counts and spleen size. Low numbers of PD1+/CD4+ and PD1+/CD8+ cells were associated with complete resolution of palpable splenomegaly and improved survival rate, suggesting that low levels of exhausted T cells confer a favorable response to ruxolitinib treatment.
Collapse
Affiliation(s)
- Ivo Veletic
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sanja Prijic
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Taghi Manshouri
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Srdan Verstovsek
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zeev Estrov
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
13
|
Khan MA, Khan A. Role of NKT Cells during Viral Infection and the Development of NKT Cell-Based Nanovaccines. Vaccines (Basel) 2021; 9:vaccines9090949. [PMID: 34579186 PMCID: PMC8473043 DOI: 10.3390/vaccines9090949] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/16/2021] [Accepted: 08/23/2021] [Indexed: 12/30/2022] Open
Abstract
Natural killer T (NKT) cells, a small population of T cells, are capable of influencing a wide range of the immune cells, including T cells, B cells, dendritic cells and macrophages. In the present review, the antiviral role of the NKT cells and the strategies of viruses to evade the functioning of NKT cell have been illustrated. The nanoparticle-based formulations have superior immunoadjuvant potential by facilitating the efficient antigen processing and presentation that favorably elicits the antigen-specific immune response. Finally, the immunoadjuvant potential of the NKT cell ligand was explored in the development of antiviral vaccines. The use of an NKT cell-activating nanoparticle-based vaccine delivery system was supported in order to avoid the NKT cell anergy. The results from the animal and preclinical studies demonstrated that nanoparticle-incorporated NKT cell ligands may have potential implications as an immunoadjuvant in the formulation of an effective antiviral vaccine that is capable of eliciting the antigen-specific activation of the cell-mediated and humoral immune responses.
Collapse
|
14
|
Role of interferon regulatory factor 7 in corneal endothelial cells after HSV-1 infection. Sci Rep 2021; 11:16487. [PMID: 34389779 PMCID: PMC8363731 DOI: 10.1038/s41598-021-95823-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/20/2021] [Indexed: 11/09/2022] Open
Abstract
Viral infections of the cornea including herpes simplex virus 1 (HSV-1) cause visual morbidity, and the corneal endothelial cell damage leads to significant visual impairment. Interferon regulatory factor 7 (IRF7) has been identified as a significant regulator in corneal endothelial cells after an HSV-1 infection. To examine the role played by IRF7, the DNA binding domain (DBD) of IRF7 of human corneal endothelial cells (HCEn) was disrupted. An RNAi inhibition of IRF7 and IRF7 DBD disruption (IRF7 ∆DBD) led to an impairment of IFN-β production. Impaired IFN-β production by IRF7 ∆DBD was regained by IRF7 DNA transfection. Transcriptional network analysis indicated that IRF7 plays a role in antigen presentation function of corneal endothelial cells. When the antigen presentation activity of HCEn cells were examined for priming of memory CD8 T cells, IRF7 disruption abolished the anti-viral cytotoxic T lymphocyte (CTL) response which was dependent on the major histocompatibility complex (MHC) class I. To further examine the roles played by IRF7 in CTL induction as acquired immunity, the contribution of IRF7 to MHC class I-mediated antigen presentation was assessed. Analysis of IRF7 ∆DBD cells indicated that IRF7 played an unrecognized role in MHC class I induction, and the viral infection induced-MHC class I induction was abolished by IRF7 disruption. Collectively, the IRF7 in corneal endothelial cells not only contributed to type I IFN response, but also to the mediation of viral infection-induced MHC class I upregulation and priming of CD8 arm of acquired immunity.
Collapse
|
15
|
Wang Z, Xu F, Hu J, Zhang H, Cui L, Lu W, He W, Wang X, Li M, Zhang H, Xiong W, Xie C, Liu Y, Zhou P, Liu J, Huang P, Qin XF, Xia X. Modulation of lactate-lysosome axis in dendritic cells by clotrimazole potentiates antitumor immunity. J Immunother Cancer 2021; 9:e002155. [PMID: 34016722 PMCID: PMC8141455 DOI: 10.1136/jitc-2020-002155] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Dendritic cells (DCs) play a critical role in antitumor immunity, but the therapeutic efficacy of DC-mediated cancer vaccine remains low, partly due to unsustainable DC function in tumor antigen presentation. Thus, identifying drugs that could enhance DC-based antitumor immunity and uncovering the underlying mechanism may provide new therapeutic options for cancer immunotherapy. METHODS In vitro antigen presentation assay was used for DC-modulating drug screening. The function of DC and T cells was measured by flow cytometry, ELISA, or qPCR. B16, MC38, CT26 tumor models and C57BL/6, Balb/c, nude, and Batf3-/- mice were used to analyze the in vivo therapy efficacy and impact on tumor immune microenvironment by clotrimazole treatment. RESULTS By screening a group of small molecule inhibitors and the US Food and Drug Administration (FDA)-approved drugs, we identified that clotrimazole, an antifungal drug, could promote DC-mediated antigen presentation and enhance T cell response. Mechanistically, clotrimazole acted on hexokinase 2 to regulate lactate metabolic production and enhanced the lysosome pathway and Chop expression in DCs subsequently induced DC maturation and T cell activation. Importantly, in vivo clotrimazole administration induced intratumor immune infiltration and inhibited tumor growth depending on both DCs and CD8+ T cells and potentiated the antitumor efficacy of anti-PD1 antibody. CONCLUSIONS Our findings showed that clotrimazole could trigger DC activation via the lactate-lysosome axis to promote antigen cross-presentation and could be used as a potential combination therapy approach to improving the therapeutic efficacy of anti-PD1 immunotherapy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Basic-Leucine Zipper Transcription Factors/genetics
- Basic-Leucine Zipper Transcription Factors/metabolism
- Cell Line, Tumor
- Clotrimazole/pharmacology
- Colonic Neoplasms/drug therapy
- Colonic Neoplasms/genetics
- Colonic Neoplasms/immunology
- Colonic Neoplasms/metabolism
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Female
- Hexokinase/metabolism
- Immune Checkpoint Inhibitors/pharmacology
- Immunomodulating Agents/pharmacology
- Lactic Acid/metabolism
- Lymphocyte Activation/drug effects
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lysosomes/drug effects
- Lysosomes/immunology
- Lysosomes/metabolism
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Nude
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Skin Neoplasms/drug therapy
- Skin Neoplasms/genetics
- Skin Neoplasms/immunology
- Skin Neoplasms/metabolism
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Transcription Factor CHOP/metabolism
- Tumor Burden
- Tumor Microenvironment
- Mice
Collapse
Affiliation(s)
- Zining Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Feifei Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jie Hu
- Department of Medical Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hongxia Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lei Cui
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenhua Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenzhuo He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of The VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaojuan Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mengyun Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Huanling Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenjing Xiong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chunyuan Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yongxiang Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Penghui Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jinyun Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Metabolic Innovation Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Peng Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Metabolic Innovation Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaofeng Frank Qin
- Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
| | - Xiaojun Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
16
|
McGill AR, Kahlil R, Dutta R, Green R, Howell M, Mohapatra S, Mohapatra SS. SARS-CoV-2 Immuno-Pathogenesis and Potential for Diverse Vaccines and Therapies: Opportunities and Challenges. Infect Dis Rep 2021; 13:102-125. [PMID: 33557330 PMCID: PMC7931091 DOI: 10.3390/idr13010013] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/24/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is a novel coronavirus that emerged from Wuhan, China in late 2019 causing coronavirus disease-19 (COVID-19). SARS-CoV-2 infection begins by attaching to angiotensin-converting enzyme 2 receptor (ACE2) via the spike glycoprotein, followed by cleavage by TMPRSS2, revealing the viral fusion domain. Other presumptive receptors for SARS-CoV-2 attachment include CD147, neuropilin-1 (NRP1), and Myeloid C-lectin like receptor (CLR), each of which might play a role in the systemic viral spread. The pathology of SARS-CoV-2 infection ranges from asymptomatic to severe acute respiratory distress syndrome, often displaying a cytokine storm syndrome, which can be life-threatening. Despite progress made, the detailed mechanisms underlying SARS-CoV-2 interaction with the host immune system remain unclear and are an area of very active research. The process's key players include viral non-structural proteins and open reading frame products, which have been implicated in immune antagonism. The dysregulation of the innate immune system results in reduced adaptive immune responses characterized by rapidly diminishing antibody titers. Several treatment options for COVID-19 are emerging, with immunotherapies, peptide therapies, and nucleic acid vaccines showing promise. This review discusses the advances in the immunopathology of SARS-CoV-2, vaccines and therapies under investigation to counter the effects of this virus, as well as viral variants.
Collapse
Affiliation(s)
- Andrew R. McGill
- Department of Veterans Affairs, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (A.R.M.); (R.K.); (R.D.); (R.G.); (M.H.)
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Roukiah Kahlil
- Department of Veterans Affairs, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (A.R.M.); (R.K.); (R.D.); (R.G.); (M.H.)
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Rinku Dutta
- Department of Veterans Affairs, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (A.R.M.); (R.K.); (R.D.); (R.G.); (M.H.)
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Ryan Green
- Department of Veterans Affairs, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (A.R.M.); (R.K.); (R.D.); (R.G.); (M.H.)
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Mark Howell
- Department of Veterans Affairs, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (A.R.M.); (R.K.); (R.D.); (R.G.); (M.H.)
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Subhra Mohapatra
- Department of Veterans Affairs, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (A.R.M.); (R.K.); (R.D.); (R.G.); (M.H.)
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Shyam S. Mohapatra
- Department of Veterans Affairs, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (A.R.M.); (R.K.); (R.D.); (R.G.); (M.H.)
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Pharmacy Graduate Programs, Taneja College, MDC30, 12908 USF Health Drive, Tampa, FL 33612, USA
| |
Collapse
|
17
|
Francian A, Widmer A, Olsson T, Ramirez M, Heald D, Rasic K, Adams L, Martinson H, Kullberg M. Delivery of toll-like receptor agonists by complement C3-targeted liposomes activates immune cells and reduces tumour growth. J Drug Target 2021; 29:754-760. [PMID: 33472457 DOI: 10.1080/1061186x.2021.1878364] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Activation of antigen presenting cells (APCs) is necessary for immune recognition and elimination of cancer. Our lab has developed a liposome nanoparticle that binds to complement C3 proteins present in serum. These C3-liposomes are specifically internalised by APCs and other myeloid cells, which express complement C3-binding receptors. Known immune stimulating compounds, toll-like receptor (TLR) agonists, were encapsulated within the C3-liposomes, including monophosphoryl lipid A (MPLA), R848, and CpG 1826, specific for TLR4, TLR7/8, and TLR9 respectively. When recognised by their respective TLRs within the myeloid cells, these compounds trigger signal cascades that ultimately lead to increased expression of inflammatory cytokines and activation markers (CD80, CD83, CD86 and CD40). RT-PCR analysis of murine bone marrow cells treated with C3-liposomes revealed a significant increase in gene expression of pro-inflammatory cytokines and factors (IL-1β, IL-6, IL-12, TNF-α, IRF7, and IP-10). Furthermore, treatment of 4T1 tumour-bearing mice with C3-liposomes containing TLR agonists resulted in reduced tumour growth, compared to PBS treated mice. Collectively, these results demonstrate that C3-liposome delivery of TLR agonists activates APCs and induces tumour-specific adaptive immune responses, leading to reduced tumour growth in a breast cancer model.
Collapse
Affiliation(s)
- Alexandra Francian
- WWAMI School of Medical Education, University of Alaska Anchorage, Anchorage, AK, USA
| | - Ashley Widmer
- WWAMI School of Medical Education, University of Alaska Anchorage, Anchorage, AK, USA
| | - Troy Olsson
- Department of Chemistry, University of Alaska Anchorage, Anchorage, AK, USA
| | - Marisabel Ramirez
- Department of Chemistry, University of Alaska Anchorage, Anchorage, AK, USA
| | - Darion Heald
- WWAMI School of Medical Education, University of Alaska Anchorage, Anchorage, AK, USA
| | - Keaton Rasic
- WWAMI School of Medical Education, University of Alaska Anchorage, Anchorage, AK, USA
| | - Luke Adams
- WWAMI School of Medical Education, University of Alaska Anchorage, Anchorage, AK, USA
| | - Holly Martinson
- WWAMI School of Medical Education, University of Alaska Anchorage, Anchorage, AK, USA
| | - Max Kullberg
- WWAMI School of Medical Education, University of Alaska Anchorage, Anchorage, AK, USA
| |
Collapse
|
18
|
Van Den Eeckhout B, Tavernier J, Gerlo S. Interleukin-1 as Innate Mediator of T Cell Immunity. Front Immunol 2021; 11:621931. [PMID: 33584721 PMCID: PMC7873566 DOI: 10.3389/fimmu.2020.621931] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/08/2020] [Indexed: 12/19/2022] Open
Abstract
The three-signal paradigm tries to capture how the innate immune system instructs adaptive immune responses in three well-defined actions: (1) presentation of antigenic peptides in the context of MHC molecules, which allows for a specific T cell response; (2) T cell co-stimulation, which breaks T cell tolerance; and (3) secretion of polarizing cytokines in the priming environment, thereby specializing T cell immunity. The three-signal model provides an empirical framework for innate instruction of adaptive immunity, but mainly discusses STAT-dependent cytokines in T cell activation and differentiation, while the multi-faceted roles of type I IFNs and IL-1 cytokine superfamily members are often neglected. IL-1α and IL-1β are pro-inflammatory cytokines, produced following damage to the host (release of DAMPs) or upon innate recognition of PAMPs. IL-1 activity on both DCs and T cells can further shape the adaptive immune response with variable outcomes. IL-1 signaling in DCs promotes their ability to induce T cell activation, but also direct action of IL-1 on both CD4+ and CD8+ T cells, either alone or in synergy with prototypical polarizing cytokines, influences T cell differentiation under different conditions. The activities of IL-1 form a direct bridge between innate and adaptive immunity and could therefore be clinically translatable in the context of prophylactic and therapeutic strategies to empower the formation of T cell immunity. Understanding the modalities of IL-1 activity during T cell activation thus could hold major implications for rational development of the next generation of vaccine adjuvants.
Collapse
Affiliation(s)
- Bram Van Den Eeckhout
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jan Tavernier
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Orionis Biosciences BV, Ghent, Belgium
| | - Sarah Gerlo
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
19
|
Khan MA. Therapeutic implications of statins beyond lipid lowering: In the perspective of their effects on the antigen presentation, T cells and NKT cells. Int J Health Sci (Qassim) 2021; 15:1-2. [PMID: 33708037 PMCID: PMC7934128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Masood Alam Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia,Address for correspondence: Masood Alam Khan, Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah - 51452, Saudi Arabia. Phone: +91-966507059437. E-mail:
| |
Collapse
|
20
|
Bacterial Lipoteichoic Acid Attenuates Toll-Like Receptor Dependent Dendritic Cells Activation and Inflammatory Response. Pathogens 2020; 9:pathogens9100825. [PMID: 33050033 PMCID: PMC7600882 DOI: 10.3390/pathogens9100825] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 12/16/2022] Open
Abstract
Toll-like receptor (TLR) signaling is an indispensable factor in immune cells activation. Many TLR ligands have been identified, and were characterized the immunological functions such as inflammatory cytokine production in immune cells. However, the anti-inflammatory response in TLR ligand-mediated manner is poorly understood. In this report, we show that bacterial lipoteichoic acid (LTA), which is a TLR2 ligand from gram-positive bacteria including Staphylococcus aureus (S. aureus), suppresses TLR-mediated inflammatory response in dendritic cells (DCs). The TLR ligand-induced Tumor Necrosis Factor-alpha (TNF-α) production was suppressed in the bone marrow derived dendritic cells (BMDCs) by co-treatment of LTA. The cellular activation, which was characterized as upregulations of CD80, CD86 and major histocompatibility complex II (MHC II) expression, was also suppressed in the TLR ligand stimulated BMDCs in the presence of LTA. While LTA itself didn’t induced both TNF-α production and upregulation of cell surface markers. The LTA mediated immunosuppressive function was abolished by TLR2 blocking in lipopolysaccharide (LPS)-stimulated BMDCs. Furthermore, LTA also showed the immunosuppressive function in the generation of IFN-γ+CD4+ T (Th1) cells by attenuation of antigen presenting activity in the BMDCs. In the imiquimod (IMQ)-induced acute skin inflammation, LTA suppressed the inflammation by downregulation of the activation in skin accumulated DCs. Thus, LTA is a TLR2 dependent immunological suppressor against inflammatory response induced by other TLR ligands in the DCs.
Collapse
|
21
|
Hernández RB, Carrascal M, Abian J, Michalke B, Farina M, Gonzalez YR, Iyirhiaro GO, Moteshareie H, Burnside D, Golshani A, Suñol C. Manganese-induced neurotoxicity in cerebellar granule neurons due to perturbation of cell network pathways with potential implications for neurodegenerative disorders. Metallomics 2020; 12:1656-1678. [PMID: 33206086 DOI: 10.1039/d0mt00085j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Manganese (Mn) is essential for living organisms, playing an important role in nervous system function. Nevertheless, chronic and/or acute exposure to this metal, especially during early life stages, can lead to neurotoxicity and dementia by unclear mechanisms. Thus, based on previous works of our group with yeast and zebrafish, we hypothesized that the mechanisms mediating manganese-induced neurotoxicity can be associated with the alteration of protein metabolism. These mechanisms may also depend on the chemical speciation of manganese. Therefore, the current study aimed at investigating the mechanisms mediating the toxic effects of manganese in primary cultures of cerebellar granule neurons (CGNs). By exposing cultured CGNs to different chemical species of manganese ([[2-[(dithiocarboxy)amino]ethyl]carbamodithioato]](2-)-kS,kS']manganese, named maneb (MB), and [[1,2-ethanediylbis[carbamodithioato]](2-)]manganese mixture with [[1,2-ethanediylbis[carbamodithioato]](2-)]zinc, named mancozeb (MZ), and manganese chloride (MnCl2)), and using the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, we observed that both MB and MZ induced similar cytotoxicity (LC50∼ 7-9 μM), which was higher than that of MnCl2 (LC50∼ 27 μM). Subsequently, we applied systems biology approaches, including metallomics, proteomics, gene expression and bioinformatics, and revealed that independent of chemical speciation, for non-cytotoxic concentrations (0.3-3 μM), Mn-induced neurotoxicity in CGNs is associated with metal dyshomeostasis and impaired protein metabolism. In this way, we verified that MB induced more post-translational alterations than MnCl2, which can be a plausible explanation for cytotoxic differences between both chemical species. The metabolism of proteins is one of the most energy consuming cellular processes and its impairment appears to be a key event of some cellular stress processes reported separately in other studies such as cell cycle arrest, energy impairment, cell signaling, excitotoxicity, immune response, potential protein accumulation and apoptosis. Interestingly, we verified that Mn-induced neurotoxicity shares pathways associated with the development of Alzheimer's disease, Amyotrophic Lateral Sclerosis, Huntington's disease, and Parkinson's disease. This has been observed in baker's yeast and zebrafish suggesting that the mode of action of Mn may be evolutionarily conserved.
Collapse
Affiliation(s)
- Raúl Bonne Hernández
- Laboratory of Bioinorganic and Environmental Toxicology - LABITA, Department of Exact and Earth Sciences, Federal University of São Paulo, Rua Prof. Artur Riedel, 275, CEP 09972-270, Diadema, SP, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
: The use of cytomegalovirus (CMV) as a vaccine vector to express antigens against multiple infectious diseases, including simian immunodeficiency virus, Ebola virus, plasmodium, and mycobacterium tuberculosis, in rhesus macaques has generated extraordinary levels of protective immunity against subsequent pathogenic challenge. Moreover, the mechanisms of immune protection have altered paradigms about viral vector-mediated immunity against ectopically expressed vaccine antigens. Further optimization of CMV-vectored vaccines, particularly as this approach moves to human clinical trials will be augmented by a more complete understanding of how CMV engenders mechanisms of immune protection. This review summarizes the particulars of the specific CMV vaccine vector that has been used to date (rhesus CMV strain 68-1) in relation to CMV natural history.
Collapse
|
23
|
Han P, Hanlon D, Arshad N, Lee JS, Tatsuno K, Robinson E, Filler R, Sobolev O, Cote C, Rivera-Molina F, Toomre D, Fahmy T, Edelson R. Platelet P-selectin initiates cross-presentation and dendritic cell differentiation in blood monocytes. SCIENCE ADVANCES 2020; 6:eaaz1580. [PMID: 32195350 PMCID: PMC7065880 DOI: 10.1126/sciadv.aaz1580] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/17/2019] [Indexed: 05/04/2023]
Abstract
Dendritic cells (DCs) are adept at cross-presentation and initiation of antigen-specific immunity. Clinically, however, DCs produced by in vitro differentiation of monocytes in the presence of exogenous cytokines have been met with limited success. We hypothesized that DCs produced in a physiological manner may be more effective and found that platelets activate a cross-presentation program in peripheral blood monocytes with rapid (18 hours) maturation into physiological DCs (phDCs). Differentiation of monocytes into phDCs was concomitant with the formation of an "adhesion synapse," a biophysical junction enriched with platelet P-selectin and monocyte P-selectin glycoprotein ligand 1, followed by intracellular calcium fluxing and nuclear localization of nuclear factor κB. phDCs were more efficient than cytokine-derived DCs in generating tumor-specific T cell immunity. Our findings demonstrate that platelets mediate a cytokine-independent, physiologic maturation of DC and suggest a novel strategy for DC-based immunotherapies.
Collapse
Affiliation(s)
- Patrick Han
- Department of Chemical and Environmental Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT 06511, USA
| | - Douglas Hanlon
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Najla Arshad
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Jung Seok Lee
- Department of Biomedical Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT 06511, USA
| | - Kazuki Tatsuno
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Eve Robinson
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Renata Filler
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Olga Sobolev
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Christine Cote
- Yale Flow Cytometry Facility, School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Felix Rivera-Molina
- Yale CINEMA Lab, School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Derek Toomre
- Yale CINEMA Lab, School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Tarek Fahmy
- Department of Chemical and Environmental Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT 06511, USA
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT 06511, USA
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT 06511, USA
- Department of Biomedical Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT 06511, USA
- Corresponding author. (T.F.); (R.E.)
| | - Richard Edelson
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT 06511, USA
- Corresponding author. (T.F.); (R.E.)
| |
Collapse
|
24
|
The epigenetic face of lupus: Focus on antigen-presenting cells. Int Immunopharmacol 2020; 81:106262. [PMID: 32045873 DOI: 10.1016/j.intimp.2020.106262] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 01/23/2020] [Accepted: 01/27/2020] [Indexed: 12/19/2022]
Abstract
In recent years, epigenetic mechanisms became widely known due to their ability to regulate and maintain physiological processes such as cell growth, development, differentiation and genomic stability. When dysregulated, epigenetic mechanisms, may introduce gene expression changes and disturbance in immune homeostasis leading to autoimmune diseases. Systemic lupus erythematosus (SLE), the most extensively studied autoimmune disorder, has already been correlated with epigenetic modifications, especially in T cells. Since these cell rely on antigen presentation, it may be assumed that erroneous activity of antigen-presenting cells (APCs), culminates in T cell abnormalities. In this review we summarize and discuss the epigenetic modifications in SLE affected APCs, with the focus on dendritic cells (DCs), B cells and monocytes. Unravelling this aspect of SLE pathogenesis, might result in identification of new disease biomarkers and putative therapeutic approaches.
Collapse
|
25
|
Jeon DB, Shin HG, Lee BW, Jeong SH, Kim JH, Ha JH, Park JY, Kwon HJ, Kim WJ, Ryu YB, Lee IC. Effect of heat-killed Enterococcus faecalis EF-2001 on ethanol-induced acute gastric injury in mice: Protective effect of EF-2001 on acute gastric ulcer. Hum Exp Toxicol 2020; 39:721-733. [PMID: 31957490 DOI: 10.1177/0960327119899987] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Enterococcus faecalis is a facultative anaerobic gram-positive commensal bacterium common in the gastrointestinal tract of animals and humans. This study aimed to investigate the protective effects of heat-killed E. faecalis EF-2001 (EF-2001) on acute gastric ulcer using a murine model of ethanol (EtOH)-induced acute gastric injury. EF-2001 (20, 40, and 80 mg/kg/day) was administered by oral gavage for 5 days before EtOH treatment (10 mL/kg body weight). EF-2001 effectively attenuated EtOH-induced gastric mucosal injury with reduced gastric mucosal ulcer and histological damage score. Pretreatment of EF-2001 markedly suppressed the phosphorylation of mitogen-activated protein kinases (MAPKs; ERK1/2, JNK, and p38MAPK). In addition, EF-2001 significantly inhibited phosphorylation of nuclear factor kappa B (NF-κB) and subsequently suppressed the upregulation of inducible nitric oxide synthase, cyclooxygenase-2, tumor necrosis factor alpha, interleukin 1 beta, and interleukin 6 in gastric tissues. Taken together, these results suggest that EF-2001 exerts a gastroprotective effect against acute gastric injury, and the underlying mechanism might be associated with the suppression of MAPKs and NF-κB signaling and consequent reduction of pro-inflammatory mediators or cytokines.
Collapse
Affiliation(s)
- D-B Jeon
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - H-G Shin
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - B-W Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - S-H Jeong
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - J-H Kim
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - J-H Ha
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - J-Y Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - H-J Kwon
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - W-J Kim
- Research and Development Center, Korea BeRM Co. Ltd, Seoul, Republic of Korea
| | - Y-B Ryu
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - I-C Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| |
Collapse
|
26
|
Metters G, Norville IH, Titball RW, Hemsley CM. From cell culture to cynomolgus macaque: infection models show lineage-specific virulence potential of Coxiella burnetii. J Med Microbiol 2019; 68:1419-1430. [PMID: 31424378 DOI: 10.1099/jmm.0.001064] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Coxiella burnetii is an obligate intracellular pathogen that causes the zoonotic disease Q fever in humans, which can occur in either an acute or a chronic form with serious complications. The bacterium has a wide host range, including unicellular organisms, invertebrates, birds and mammals, with livestock representing the most significant reservoir for human infections. Cell culture models have been used to decipher the intracellular lifestyle of C. burnetii, and several infection models, including invertebrates, rodents and non-human primates, are being used to investigate host-pathogen interactions and to identify bacterial virulence factors and vaccine candidates. However, none of the models replicate all aspects of human disease. Furthermore, it is becoming evident that C. burnetii isolates belonging to different lineages exhibit differences in their virulence in these models. Here, we compare the advantages and disadvantages of commonly used infection models and summarize currently available data for lineage-specific virulence.
Collapse
Affiliation(s)
- Georgina Metters
- College of Life and Environmental Sciences - Biosciences, University of Exeter, Exeter, UK
| | - Isobel H Norville
- Defence Science and Technology Laboratory, Porton Down, Salisbury, UK
| | - Richard W Titball
- College of Life and Environmental Sciences - Biosciences, University of Exeter, Exeter, UK
| | - Claudia M Hemsley
- College of Life and Environmental Sciences - Biosciences, University of Exeter, Exeter, UK
| |
Collapse
|
27
|
Banerjee R, Roy S, Samanta M, Das S. Molecular cloning, characterization and expression analysis of MHCI and chemokines CXCR3 and CXCR4 gene from freshwater carp, Catla catla. Microbiol Immunol 2019; 63:379-391. [PMID: 31310013 DOI: 10.1111/1348-0421.12728] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/24/2019] [Accepted: 07/08/2019] [Indexed: 12/01/2022]
Abstract
The immune system with large number of molecules protects the host against a plethora of continuously evolving microbes. Major histocompatibility complex (MHC) molecules serve as cardinal elements of the adaptive immune system responsible for the activation of the adaptive immunity in the host. The present study reports MHCI molecule in freshwater carp, Catla catla, and its differential expression in immunologically relevant tissues post-infection with Gram-negative and Gram-positive bacteria. The MHCI sequence of C. catla had 502 bp nucleotides encoding putative 146 amino acids. The phylogenetic analysis exhibited its evolutionary conservation within the Cyprinidae family and formed a different clade with the higher vertebrates. Simultaneously, CXCR3 and CXCR4 chemokines were cloned and characterized for their expression in infected tissues. Analysis of immunologically relevant tissues of the infected fish exhibited an increase of MHCI gene expression and the down-regulation of CXCR3 and CXCR4 chemokines, indicating a tricky interaction between the innate and adaptive immune system. It was found that intestine, skin and spleen played a crucial role in the contribution of the defense activity which instigated the self-immunity. These immune activities can provide useful information to understand the interaction of self and non-self- immune system in freshwater fish, Catla catla.
Collapse
Affiliation(s)
- Rajanya Banerjee
- Laboratory of Environmental Microbiology and Ecology (LEnME), Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Sudeshna Roy
- Laboratory of Environmental Microbiology and Ecology (LEnME), Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Mrinal Samanta
- Fish Health Management Division, ICAR-Central Institute of Freshwater Aquaculture, Kausalyaganga, Bhubaneswar, 751002, Odisha, India
| | - Surajit Das
- Laboratory of Environmental Microbiology and Ecology (LEnME), Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| |
Collapse
|
28
|
Araman C, 't Hart BA. Neurodegeneration meets immunology - A chemical biology perspective. Bioorg Med Chem 2019; 27:1911-1924. [PMID: 30910473 DOI: 10.1016/j.bmc.2019.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/14/2019] [Accepted: 03/19/2019] [Indexed: 11/16/2022]
Affiliation(s)
- C Araman
- Leiden Institute of Chemistry and the Institute for Chemical Immunology, Leiden University, Leiden, The Netherlands.
| | - B A 't Hart
- University of Groningen, Department of Biomedical Sciences of Cells and Systems, University Medical Centre, Groningen, The Netherlands; Department Anatomy and Neuroscience, Free University Medical Center (VUmc), Amsterdam, The Netherlands.
| |
Collapse
|
29
|
DeLong JH, O'Hara Hall A, Rausch M, Moodley D, Perry J, Park J, Phan AT, Beiting DP, Kedl RM, Hill JA, Hunter CA. IL-27 and TCR Stimulation Promote T Cell Expression of Multiple Inhibitory Receptors. Immunohorizons 2019; 3:13-25. [PMID: 31356173 PMCID: PMC6994206 DOI: 10.4049/immunohorizons.1800083] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 12/26/2018] [Indexed: 12/18/2022] Open
Abstract
Inhibitory receptors (IR) are a diverse group of cell surface molecules that modulate T cell activation, but there are gaps in our knowledge of the cell-extrinsic factors that regulate their expression. The present study found that in vivo overexpression of IL-27 in mice led to increased T cell expression of PD-L1, LAG-3, TIGIT, and TIM-3. In vitro, TCR stimulation alone promoted expression of multiple IRs, whereas IL-27 alone induced expression of PD-L1. However, the combination of intermediate TCR stimulation and IL-27 resulted in synergistic induction of LAG-3, CTLA-4, and TIGIT. In vivo, infection with Toxoplasma gondii resulted in parasite-specific effector T cells that expressed high levels of IR, and at local sites of infection where IL-27 production was highest, IL-27 was required for maximal effector cell expression of PD-L1, LAG-3, CTLA-4, and TIGIT. Together, these results affirm the critical role of TCR signals in the induction of IR expression but find that during infection, IL-27 promotes T cell expression of IR.
Collapse
Affiliation(s)
- Jonathan H DeLong
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Aisling O'Hara Hall
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Immunology Discovery Research, Janssen Research and Development, LLC, Spring House, PA 19477
| | | | | | - Joseph Perry
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Jeongho Park
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Anthony T Phan
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Daniel P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Ross M Kedl
- Department of Immunology and Microbiology, University of Colorado, Aurora, CO 80045
| | | | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104;
| |
Collapse
|
30
|
Canavan M, Walsh AM, Bhargava V, Wade SM, McGarry T, Marzaioli V, Moran B, Biniecka M, Convery H, Wade S, Orr C, Mullan R, Fletcher JM, Nagpal S, Veale DJ, Fearon U. Enriched Cd141+ DCs in the joint are transcriptionally distinct, activated, and contribute to joint pathogenesis. JCI Insight 2018; 3:95228. [PMID: 30518680 DOI: 10.1172/jci.insight.95228] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 10/29/2018] [Indexed: 12/13/2022] Open
Abstract
CD141+ DC are implicated in antiviral and antitumor immunity. However, mechanistic studies in autoimmune disease are limited. This is the first study to our knowledge examining CD141+ DC in autoimmune disease, specifically inflammatory arthritis (IA). We identified significant enrichment of CD141+ DC in the inflamed synovial joint, which were transcriptionally distinct from IA and healthy control (HC) blood CD141+ DC and significantly more activated, and they exhibited increased responsiveness to TLR3. Synovial CD141+ DC represent a bone fide CD141+ DC population that is distinct from CD1c+ DC. Synovial CD141+ DC induced higher levels of CD4+ and CD8+ T cell activation compared with their peripheral blood counterparts, as made evident by expression of IFN-γ, TNF-α, and granulocyte-macrophage CSF (GMCSF). Autologous synovial CD141+ DC cocultures also induce higher levels of these cytokines, further highlighting their contribution to synovial inflammation. Synovial CD141+ DC-T cell interactions had the ability to further activate synovial fibroblasts, inducing adhesive and invasive pathogenic mechanisms. Furthermore, we identify a mechanism in which synovial CD141+ DC are activated, via ligation of the hypoxia-inducible immune-amplification receptor TREM-1, which increased synovial CD141+ DC activation, migratory capacity, and proinflammatory cytokines. Thus, synovial CD141+ DC display unique mechanistic and transcriptomic signatures, which are distinguishable from blood CD141+ DC and can contribute to synovial joint inflammation.
Collapse
Affiliation(s)
- Mary Canavan
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Ireland
| | | | - Vipul Bhargava
- Discovery Sciences, Janssen Research & Development, Spring House, Pennsylvania, USA
| | - Sarah M Wade
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Ireland
| | - Trudy McGarry
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Ireland
| | - Viviana Marzaioli
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Ireland
| | - Barry Moran
- School of Biochemistry and Immunology, Trinity College Dublin, Ireland
| | - Monika Biniecka
- Centre for Arthritis & Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Ireland
| | - Hannah Convery
- Centre for Arthritis & Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Ireland
| | - Siobhan Wade
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Ireland
| | - Carl Orr
- Centre for Arthritis & Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Ireland
| | - Ronan Mullan
- Department of Rheumatology, Adelaide and Meath Hospital, Dublin, Ireland
| | - Jean M Fletcher
- Translational Immunology, Schools of Biochemistry and Immunology and Medicine, Trinity College Dublin, Ireland
| | | | - Douglas J Veale
- Centre for Arthritis & Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Ireland
| | - Ursula Fearon
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Ireland
| |
Collapse
|
31
|
Liu J, Gallo RM, Khan MA, Iyer AK, Kratzke IM, Brutkiewicz RR. JNK2 modulates the CD1d-dependent and -independent activation of iNKT cells. Eur J Immunol 2018; 49:255-265. [PMID: 30467836 DOI: 10.1002/eji.201847755] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 10/30/2018] [Accepted: 11/21/2018] [Indexed: 01/01/2023]
Abstract
Invariant natural killer T (iNKT) cells play critical roles in autoimmune, anti-tumor, and anti-microbial immune responses, and are activated by glycolipids presented by the MHC class I-like molecule, CD1d. How the activation of signaling pathways impacts antigen (Ag)-dependent iNKT cell activation is not well-known. In the current study, we found that the MAPK JNK2 not only negatively regulates CD1d-mediated Ag presentation in APCs, but also contributes to CD1d-independent iNKT cell activation. A deficiency in the JNK2 (but not JNK1) isoform enhanced Ag presentation by CD1d. Using a vaccinia virus (VV) infection model known to cause a loss in iNKT cells in a CD1d-independent, but IL-12-dependent manner, we found the virus-induced loss of iNKT cells in JNK2 KO mice was substantially lower than that observed in JNK1 KO or wild-type (WT) mice. Importantly, compared to WT mice, JNK2 KO mouse iNKT cells were found to express less surface IL-12 receptors. As with a VV infection, an IL-12 injection also resulted in a smaller decrease in JNK2 KO iNKT cells as compared to WT mice. Overall, our work strongly suggests JNK2 is a negative regulator of CD1d-mediated Ag presentation and contributes to IL-12-induced iNKT cell activation and loss during viral infections.
Collapse
Affiliation(s)
- Jianyun Liu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Richard M Gallo
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Masood A Khan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.,College of Applied Medical Sciences, Al-Qassim University, Buraidah, Saudi Arabia
| | - Abhirami K Iyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ian M Kratzke
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Randy R Brutkiewicz
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
32
|
Lv J, Yu Q, Lv J, Di C, Lin X, Su W, Wu M, Xia Z. Airway epithelial TSLP production of TLR2 drives type 2 immunity in allergic airway inflammation. Eur J Immunol 2018; 48:1838-1850. [PMID: 30184256 PMCID: PMC6282509 DOI: 10.1002/eji.201847663] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/29/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022]
Abstract
Epithelial cells (ECs)-derived cytokines are induced by different stimuli through pattern recognition receptors (PRRs) to mount a type-2-cell-mediated immune response; however, the underlying mechanisms are poorly characterized. Here, we demonstrated asthmatic features in both primary bronchial epithelial cells (pBECs) and mouse model using several allergens including ovalbumin (OVA), house dust mite (HDM), or Alternaria alternata. We found that toll-like receptor 2 (TLR2) was highly induced in ECs but not dendritic cells (DCs) by various allergens, leading to recruitment of circulating basophils into the lung via C-C chemokine ligand-2 (CCL2). TLR2 expression increased thymic stromal lymphopoietin (TSLP) production through the NF-κB and JNK signaling pathways to extend the survival of recruited basophils and resident DCs in the lung, predisposing a type-2-cell-mediated airway inflammation. Conversely, TLR2 deficiency impaired secretion of TSLP and CCL2, decreased infiltration of lung basophils, and increased resistance to Th2 response. Blocking TSLP also phenocopied these phenomena. Our findings reveal a pro-inflammatory role of airway ECs through a TLR2-dependent TSLP production, which may have implication for treating allergic asthma.
Collapse
Affiliation(s)
- Jiajia Lv
- Department of Pediatrics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianying Yu
- Department of Pediatrics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Lv
- Department of Pediatrics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Caixia Di
- Department of Pediatrics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoliang Lin
- Department of Pediatrics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Su
- Department of Pediatrics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Wu
- School of Medicine & Health Sciences, Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Zhenwei Xia
- Department of Pediatrics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Brutkiewicz RR, Yunes-Medina L, Liu J. Immune evasion of the CD1d/NKT cell axis. Curr Opin Immunol 2018; 52:87-92. [PMID: 29734045 DOI: 10.1016/j.coi.2018.04.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 04/19/2018] [Indexed: 01/03/2023]
Abstract
Many reviews on the CD1d/NKT cell axis focus on the ability of CD1d-restricted NKT cells to serve as effector cells in a variety of disorders, be they infectious diseases, cancer or autoimmunity. In contrast, here, we discuss the ways that viruses, bacteria and tumor cells can evade the CD1d/NKT cell axis. As a result, these disease states have a better chance to establish a foothold and potentially cause problems for the subsequent adaptive immune response, as the host tries to rid itself of infections or tumors.
Collapse
Affiliation(s)
- Randy R Brutkiewicz
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202-5181, United States.
| | - Laura Yunes-Medina
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202-5181, United States
| | - Jianyun Liu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202-5181, United States
| |
Collapse
|
34
|
Mo Z, Yu F, Han S, Yang S, Wu L, Li P, Jiao S. New peptide MY1340 revert the inhibition effect of VEGF on dendritic cells differentiation and maturation via blocking VEGF-NRP-1 axis and inhibit tumor growth in vivo. Int Immunopharmacol 2018; 60:132-140. [PMID: 29730556 DOI: 10.1016/j.intimp.2018.04.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 03/25/2018] [Accepted: 04/12/2018] [Indexed: 12/11/2022]
Abstract
The development and clinical application of immunostimulatory therapy provides us a new and exciting strategy in cancer treatment of which the agents act on crucial receptors. Given the fact that Neuropilin-1(NRP-1) is essential for vascular endothelial growth factor (VEGF) to inhibit LPS-dependent maturation of dendritic cells (DCs), it may present a potentially meaningful target in cancer immunotherapy. To explore this hypothesis, we synthesized a novel polypeptide called MY1340 consist of 32 amino acids with the aim of targeting VEGF-NRP-1 axis. Pull-down assay coupled with liquid chromatography-tandem mass spectrometry analysis (LC-MS/MS) was firstly conducted to identify NRP-1 as a potential MY1340 interacting protein, and the interaction between them was further confirmed by western blot. The competitive enzyme-linked immunosorbent assay (ELISA) results revealed that MY1340 was able to inhibit the binding between NRP-1 and VEGF with IC50 7.42 ng/ml, better than that of Tuftsin, although a natural ligand reportedly specific for the NRP-1 receptor. The presence of VEGF significantly reduced the expression of human leukocyte antigen-DR (HLA-DR), CD86 and CD11C on DCs, and this effect was reverted by MY1340-augment p65 NF-κB and ERK1/2 phosphorylation. We also present evidence that MY1340 is remarkably efficacious in the treatment of mice bearing subcutaneous liver cancer and induced DC maturation in the tumor environment in vivo. Taken together, these results indicate that MY1340 may represent a potential efficient immune therapeutic compound within disease that are rich in VEGF.
Collapse
Affiliation(s)
- Zheng Mo
- PLA General Hospital Cancer Center, PLA Postgraduate School of Medicine, Beijing, People's Republic of China
| | - Fei Yu
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Su Han
- DC Bio Lab, Beijing, People's Republic of China
| | | | - Liangliang Wu
- PLA General Hospital Cancer Center, PLA Postgraduate School of Medicine, Beijing, People's Republic of China
| | - Peng Li
- PLA General Hospital Cancer Center, PLA Postgraduate School of Medicine, Beijing, People's Republic of China
| | - Shunchang Jiao
- PLA General Hospital Cancer Center, PLA Postgraduate School of Medicine, Beijing, People's Republic of China.
| |
Collapse
|
35
|
Jain A, Pasare C. Innate Control of Adaptive Immunity: Beyond the Three-Signal Paradigm. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:3791-3800. [PMID: 28483987 PMCID: PMC5442885 DOI: 10.4049/jimmunol.1602000] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/02/2017] [Indexed: 12/14/2022]
Abstract
Activation of cells in the adaptive immune system is a highly orchestrated process dictated by multiples cues from the innate immune system. Although the fundamental principles of innate control of adaptive immunity are well established, it is not fully understood how innate cells integrate qualitative pathogenic information to generate tailored protective adaptive immune responses. In this review, we discuss complexities involved in the innate control of adaptive immunity that extend beyond TCR engagement, costimulation, and priming cytokine production but are critical for the generation of protective T cell immunity.
Collapse
Affiliation(s)
- Aakanksha Jain
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9093
| | - Chandrashekhar Pasare
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9093
| |
Collapse
|
36
|
Solbakken MH, Voje KL, Jakobsen KS, Jentoft S. Linking species habitat and past palaeoclimatic events to evolution of the teleost innate immune system. Proc Biol Sci 2017; 284:20162810. [PMID: 28446692 PMCID: PMC5413918 DOI: 10.1098/rspb.2016.2810] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/21/2017] [Indexed: 12/14/2022] Open
Abstract
Host-intrinsic factors as well as environmental changes are known to be strong evolutionary drivers defining the genetic foundation of immunity. Using a novel set of teleost genomes and a time-calibrated phylogeny, we here investigate the family of Toll-like receptor (TLR) genes and address the underlying evolutionary processes shaping the diversity of the first-line defence. Our findings reveal remarkable flexibility within the evolutionary design of teleost innate immunity characterized by prominent TLR gene losses and expansions. In the order of Gadiformes, expansions correlate with the loss of major histocompatibility complex class II (MHCII) and diversifying selection analyses support that this has fostered new immunological innovations in TLRs within this lineage. In teleosts overall, TLRs expansions correlate with species latitudinal distributions and maximum depth. By contrast, lineage-specific gene losses overlap with well-described changes in palaeoclimate (global ocean anoxia) and past Atlantic Ocean geography. In conclusion, we suggest that the evolvability of the teleost immune system has most likely played a prominent role in the survival and successful radiation of this lineage.
Collapse
Affiliation(s)
- Monica Hongrø Solbakken
- Centre for Ecological and Evolutionary Synthesis (CEES), Department of Biosciences, University of Oslo, PO Box 1066, Blindern, 0316 Oslo, Norway
| | - Kjetil Lysne Voje
- Centre for Ecological and Evolutionary Synthesis (CEES), Department of Biosciences, University of Oslo, PO Box 1066, Blindern, 0316 Oslo, Norway
| | - Kjetill Sigurd Jakobsen
- Centre for Ecological and Evolutionary Synthesis (CEES), Department of Biosciences, University of Oslo, PO Box 1066, Blindern, 0316 Oslo, Norway
| | - Sissel Jentoft
- Centre for Ecological and Evolutionary Synthesis (CEES), Department of Biosciences, University of Oslo, PO Box 1066, Blindern, 0316 Oslo, Norway
- Department of Natural Sciences, University of Agder, Kristiansand, Norway
| |
Collapse
|