1
|
de Oliveira Matos A, Henrique Dos Santos Dantas P, Rodrigues do Carmo Neto J, Contreras Colmenares MT, Felice AG, de Castro Soares S, Silva-Sales M, Sales-Campos H. Uncovering the role of TREM-1 in celiac disease: In silico insights into the recognition of gluten-derived peptides and inflammatory mechanisms. Comput Biol Med 2025; 189:109981. [PMID: 40056844 DOI: 10.1016/j.compbiomed.2025.109981] [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: 10/01/2024] [Revised: 01/31/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Celiac disease (CD) is a chronic enteropathy characterized by a permanent intolerance to gluten. While CD has been associated with heightened T cell responses and the involvement of distinct innate immunity components, the role of the triggering receptor expressed on myeloid cells (TREM) family in this disease remains unclear. Thus, as TREM-1 has already been implicated in other inflammatory bowel diseases, and given its role in the amplification of inflammation, we hypothesized that it might play a role in the pathophysiology of CD. METHODS AND RESULTS the STRING tool was used to predict protein-protein interaction networks between TREM-1 and CD signaling pathways. Then, molecular docking and molecular dynamics simulations were conducted to explore potential interactions between TREM-1 and different peptides derived from alpha-gliadin (25-mer, 33-mer and p31-43). Finally, we used transcriptomic data, available from public repositories, to assess TREM1 gene expression, and genes involved in its signaling pathway, in CD patients. Our results found an association between TREM-1 and CD markers, with STRING analysis, and the in silico simulations suggesting that the receptor might recognize the alpha-gliadin peptides, with the TREM-1/p31-43 interaction as the most likely interaction to occur biologically. Furthermore, TREM1 and its signaling pathway were increased in patients with active CD, while in those in clinical remission, the expression levels were similar to healthy controls. CONCLUSIONS collectively, our findings suggest that TREM-1 might recognize alpha-gliadin derived peptides, and TREM-1's activation may contribute to the intestinal inflammation observed in CD.
Collapse
Affiliation(s)
- Amanda de Oliveira Matos
- Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil.
| | | | | | | | - Andrei Giacchetto Felice
- Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil.
| | - Siomar de Castro Soares
- Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil.
| | - Marcelle Silva-Sales
- Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil.
| | | |
Collapse
|
2
|
Yeku OO, Barve M, Tan WW, Wang J, Patnaik A, LoRusso P, Richardson DL, Naqash AR, Lynam SK, Fu S, Gordon M, Hubbard J, Kummar S, Kyriakopoulos C, Dowlati A, Chamberlain M, Winer I. Myeloid targeting antibodies PY159 and PY314 for platinum-resistant ovarian cancer. J Immunother Cancer 2025; 13:e010959. [PMID: 40081941 PMCID: PMC11907075 DOI: 10.1136/jitc-2024-010959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/03/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Novel treatment options are required in patients with platinum-resistant ovarian cancer (PROC). Myeloid-derived suppressor cells promote a hostile tumor microenvironment and are associated with worse clinical outcomes in PROC. We evaluated the safety and preliminary efficacy of PY159, an agonist antibody to Triggering receptor expressed on myeloid cells-1 (TREM1) that reprograms immunosuppressive intratumoral myeloid cells, and PY314, an antagonist antibody to Triggering receptor expressed on myeloid cells-2 (TREM2) that depletes tumor-associated macrophages, as single agents and in combination with pembrolizumab in subjects with PROC. METHODS PY159 and PY314 were individually evaluated in patients with PROC. Patients were treated with monotherapy (PY159 3 mg/kg or PY314 10 mg/kg), based on the recommended dose for expansion derived from the phase 1a studies. At the time of first progression, patients could continue study drug and crossover to combination therapy with pembrolizumab (200 mg) every 3 weeks at the discretion of the investigator. Disease assessment by Response Evaluation Criteria in Solid Tumor version 1.1 was performed every 6 weeks. RESULTS 17 patients were enrolled in the PY159 study (median age 67, range 22-77; median prior therapies 6, range 2-18) and 16 patients in PY314 (median age 65.5, range 49-81; median prior therapies 4, range 2-10). 7 patients in PY159 and 8 patients in PY314 crossed over to combination therapy. Safety events included the following: treatment-related adverse events occurred in 15 patients (88.2%) in PY159 and 9 patients (56.3%) in PY314. Infusion-related reactions occurred in 6 patients (35.3%) in PY159 and 3 patients (18.8%) in PY314. Immune-related adverse events occurred in 13 patients (76.5%) in PY159 (arthralgias) and 1 patient (6.3%) in PY314 (diarrhea). Serious adverse events occurred in 6 patients (36.3%) in PY159 (1 related) and 12 patients (75%) in PY314 (all unrelated). The best radiographic response in PY159 was stable disease in 8/16 patients (50%; median 16 weeks, range 9-33), and in PY314, it was stable disease in 8/16 patients (50%; median 12 weeks, range 6-36). Median PFS was 2.76 months and 2.69 months in PY159 and PY314, respectively. There were no responses in the crossover arm. CONCLUSIONS Both PY159 and PY314 were well tolerated, with an acceptable safety profile, as both single agents and in combination with pembrolizumab. Both agents warrant further investigation in heavily pretreated PROC.
Collapse
Affiliation(s)
- Oladapo O Yeku
- Medicine/Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Minal Barve
- Mary Crowley Cancer Research, Dallas, Texas, USA
| | | | - Judy Wang
- Drug Development Unit, Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota, Florida, USA
| | - Amita Patnaik
- The START Center for Cancer Research, San Antonio, Texas, USA
| | | | - Debra L Richardson
- Division of Gynecologic Oncology, The University of Oklahoma Stephenson Cancer Center, Oklahoma City, Oklahoma, USA
- Sarah Cannon Research Institute, Oklahoma City, Oklahoma, USA
| | - Abdul Rafeh Naqash
- Medical Oncology/TSET Phase 1 Program, Stephenson Cancer Center/Sarah Cannon Research Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Sarah K Lynam
- University Hospitals Seidman Cancer Center and Case Western Reserve University, Cleveland, Ohio, USA
| | - Siqing Fu
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Joleen Hubbard
- Allina Health Cancer Institute, Minneapolis, Minnesota, USA
| | - Shivaani Kummar
- Oregon Health & Science University Knight Cancer Institute, Portland, Oregon, USA
| | | | - Afshin Dowlati
- University Hospitals Seidman Cancer Center and Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Ira Winer
- Wayne State University and Karmanos Cancer Center, Detroit, Michigan, USA
| |
Collapse
|
3
|
Sigalov AB. TREM-1 and TREM-2 as therapeutic targets: clinical challenges and perspectives. Front Immunol 2024; 15:1498993. [PMID: 39737196 PMCID: PMC11682994 DOI: 10.3389/fimmu.2024.1498993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/30/2024] [Indexed: 01/01/2025] Open
Abstract
TREM-1 and TREM-2 as Therapeutic Targets: Clinical Challenges and Perspectives.
Collapse
|
4
|
Ramatchandirin B, Balamurugan MA, Desiraju S, Chung Y, Wojczyk BS, MohanKumar K. Stored RBC transfusions leads to the systemic inflammatory response syndrome in anemic murine neonates. Inflamm Res 2024; 73:1859-1873. [PMID: 39235608 PMCID: PMC11540732 DOI: 10.1007/s00011-024-01936-y] [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: 05/06/2024] [Revised: 07/29/2024] [Accepted: 08/16/2024] [Indexed: 09/06/2024] Open
Abstract
OBJECTIVE RBC transfusions (RBCT) are life-saving treatment for premature and critically ill infants. However, the procedure has been associated with the development of systemic inflammatory response syndrome (SIRS) and potentially multiple organ dysfunction syndrome (MODS) in neonates. The present study aimed to investigate the mechanisms of RBCT-related SIRS in severely anemic murine neonates. METHODS C57BL/6 (WT), TLR4-/- and myeloid-specific triggered myeloid receptor-1 (trem1)-/- mouse pups were studied in 4 groups (n = 6 each): (1) naïve controls, (2) transfused control, (3) anemic (hematocrit 20-24%) and (4) anemic with RBC transfused using our established murine model of phlebotomy-induced anemia (PIA) and RBC transfusion. Plasma was measured for quantifying inflammatory cytokines (IFN-γ, IL-1β, TNF-α, IL-6, MIP-1α, MIP-1β, MIP2 and LIX) using a Luminex assay. In vitro studies included (i) sensitization by exposing the cells to a low level of lipopolysaccharide (LPS; 500 ng/ml) and (ii) trem1-siRNA transfection with/without plasma supernatant from stored RBC to assess the acute inflammatory response through trem1 by qRT-PCR and immunoblotting. RESULTS Anemic murine pups developed cytokine storm within 2 h of receiving stored RBCs, which increased until 6 h post-transfusion, as compared to non-anemic mice receiving stored RBCTs ("transfusion controls"), in a TLR4-independent fashion. Nonetheless, severely anemic pups had elevated circulating endotoxin levels, thereby sensitizing circulating monocytes to presynthesize proinflammatory cytokines (IFN-γ, IL-1β, TNF-α, IL-6, MIP-1α, MIP-1β, MIP2, LIX) and express trem1. Silencing trem1 expression in Raw264.7 cells mitigated both endotoxin-associated presynthesis of proinflammatory cytokines and the RBCT-induced release of inflammatory cytokines. Indeed, myeloid-specific trem1-/- murine pups had significantly reduced evidence of SIRS following RBCTs. CONCLUSION Severe anemia-associated low-grade inflammation sensitizes monocytes to enhance the synthesis of proinflammatory cytokines and trem1. In this setting, RBCTs further activate these monocytes, thereby inducing SIRS. Inhibiting trem1 in myeloid cells, including monocytes, alleviates the inflammatory response associated with the combined effects of anemia and RBCTs in murine neonates.
Collapse
Affiliation(s)
- Balamurugan Ramatchandirin
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Marie Amalie Balamurugan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Suneetha Desiraju
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Yerin Chung
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Boguslaw S Wojczyk
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Krishnan MohanKumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Child Health Research Institute, Omaha, NE, 68198, USA.
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
5
|
Vinolo E, Maillefer M, Jolly L, Colné N, Meiffren G, Carrasco K, Derive M. The potential of targeting TREM-1 in IBD. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 101:301-330. [PMID: 39521605 DOI: 10.1016/bs.apha.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Innate immune dysfunction is a hallmark of the pathogenesis of Inflammatory Bowel Disease, both in Crohn's disease and ulcerative colitis. Despite considerable efforts in research to better understand the pathophysiology of IBD and for the development of new therapeutic modalities for IBD patients, there is no therapy specifically targeting the dysregulations of the innate immune response available today in that field. TREM-1 is exclusively expressed by innate immune cells and is an immune amplifier. Its engagement following the primary activation of Pattern Recognition Receptors, including Toll-Like Receptors, triggers the development of a dysregulated and sustained innate immune response, promoting the perpetuation of the inflammatory response in the mucosa of IBD patients, microscopic mucosal tissue alterations, impaired autophagy, impaired epithelial barrier integrity and function, ulcerations, and mucosal damages. In patients, TREM-1 activation is associated with the active status of the disease as well as with severity. Blocking TREM-1 in experimental colitis attenuates the dysregulated innate immune response leading to improved clinical signs. Anti-TREM-1 approaches have the potential of controlling the pathogenic dysregulation of the immune response in IBD by targeting an upstream amplification loop of the activation of innate immunity.
Collapse
|
6
|
Warner S, Teague HL, Ramos-Benitez MJ, Panicker S, Allen K, Gairhe S, Moyer T, Parachalil Gopalan B, Douagi I, Shet A, Kanthi Y, Suffredini AF, Chertow DS, Strich JR. R406 reduces lipopolysaccharide-induced neutrophil activation. Cell Immunol 2024; 403-404:104860. [PMID: 39084187 PMCID: PMC11387147 DOI: 10.1016/j.cellimm.2024.104860] [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: 05/15/2024] [Revised: 07/05/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
Modulating SYK has been demonstrated to have impacts on pathogenic neutrophil responses in COVID-19. During sepsis, neutrophils are vital in early bacterial clearance but also contribute to the dysregulated immune response and organ injury when hyperactivated. Here, we evaluated the impact of R406, the active metabolite of fostamatinib, on neutrophils stimulated by LPS. We demonstrate that R406 was able to effectively inhibit NETosis, degranulation, ROS generation, neutrophil adhesion, and the formation of CD16low neutrophils that have been linked to detrimental outcomes in severe sepsis. Further, the neutrophils remain metabolically active, capable of releasing cytokines, perform phagocytosis, and migrate in response to IL-8. Taken together, this data provides evidence of the potential efficacy of utilizing fostamatinib in bacterial sepsis.
Collapse
Affiliation(s)
- Seth Warner
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Heather L Teague
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Marcos J Ramos-Benitez
- Basic Science Department, Microbiology Division, School of Medicine, Ponce Health Sciences University, Ponce, PR, USA
| | - Sumith Panicker
- Laboratory of Vascular Thrombosis and Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kiana Allen
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Salina Gairhe
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Tom Moyer
- Flow Cytometry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bindu Parachalil Gopalan
- Laboratory of Sickle Thrombosis and Vascular Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Iyadh Douagi
- Flow Cytometry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; NIH Center for Human Immunology, Inflammation, and Autoimmunity, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Arun Shet
- Laboratory of Sickle Thrombosis and Vascular Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yogendra Kanthi
- Laboratory of Vascular Thrombosis and Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anthony F Suffredini
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Daniel S Chertow
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA; Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey R Strich
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
7
|
Shimizu J, Murao A, Lee Y, Aziz M, Wang P. Extracellular CIRP promotes Kupffer cell inflammatory polarization in sepsis. Front Immunol 2024; 15:1411930. [PMID: 38881891 PMCID: PMC11177612 DOI: 10.3389/fimmu.2024.1411930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/16/2024] [Indexed: 06/18/2024] Open
Abstract
Introduction Sepsis is a life-threatening inflammatory condition caused by dysregulated host responses to infection. Extracellular cold-inducible RNA-binding protein (eCIRP) is a recently discovered damage-associated molecular pattern that causes inflammation and organ injury in sepsis. Kupffer cells can be activated and polarized to the inflammatory M1 phenotype, contributing to tissue damage by producing proinflammatory mediators. We hypothesized that eCIRP promotes Kupffer cell M1 polarization in sepsis. Methods We stimulated Kupffer cells isolated from wild-type (WT) and TLR4-/- mice with recombinant mouse (rm) CIRP (i.e., eCIRP) and assessed supernatant IL-6 and TNFα levels by ELISA. The mRNA expression of iNOS and CD206 for M1 and M2 markers, respectively, was assessed by qPCR. We induced sepsis in WT and CIRP-/- mice by cecal ligation and puncture (CLP) and assessed iNOS and CD206 expression in Kupffer cells by flow cytometry. Results eCIRP dose- and time-dependently increased IL-6 and TNFα release from WT Kupffer cells. In TLR4-/- Kupffer cells, their increase after eCIRP stimulation was prevented. eCIRP significantly increased iNOS gene expression, while it did not alter CD206 expression in WT Kupffer cells. In TLR4-/- Kupffer cells, however, iNOS expression was significantly decreased compared with WT Kupffer cells after eCIRP stimulation. iNOS expression in Kupffer cells was significantly increased at 20 h after CLP in WT mice. In contrast, Kupffer cell iNOS expression in CIRP-/- mice was significantly decreased compared with WT mice after CLP. CD206 expression in Kupffer cells was not different across all groups. Kupffer cell M1/M2 ratio was significantly increased in WT septic mice, while it was significantly decreased in CIRP-/- mice compared to WT mice after CLP. Conclusion Our data have clearly shown that eCIRP induces Kupffer cell M1 polarization via TLR4 pathway in sepsis, resulting in overproduction of inflammatory cytokines. eCIRP could be a promising therapeutic target to attenuate inflammation by preventing Kupffer cell M1 polarization in sepsis.
Collapse
Affiliation(s)
- Junji Shimizu
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Atsushi Murao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Yongchan Lee
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| |
Collapse
|
8
|
Kaczanowska S, Murty T, Alimadadi A, Contreras CF, Duault C, Subrahmanyam PB, Reynolds W, Gutierrez NA, Baskar R, Wu CJ, Michor F, Altreuter J, Liu Y, Jhaveri A, Duong V, Anbunathan H, Ong C, Zhang H, Moravec R, Yu J, Biswas R, Van Nostrand S, Lindsay J, Pichavant M, Sotillo E, Bernstein D, Carbonell A, Derdak J, Klicka-Skeels J, Segal JE, Dombi E, Harmon SA, Turkbey B, Sahaf B, Bendall S, Maecker H, Highfill SL, Stroncek D, Glod J, Merchant M, Hedrick CC, Mackall CL, Ramakrishna S, Kaplan RN. Immune determinants of CAR-T cell expansion in solid tumor patients receiving GD2 CAR-T cell therapy. Cancer Cell 2024; 42:35-51.e8. [PMID: 38134936 PMCID: PMC10947809 DOI: 10.1016/j.ccell.2023.11.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 09/18/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023]
Abstract
Chimeric antigen receptor T cells (CAR-Ts) have remarkable efficacy in liquid tumors, but limited responses in solid tumors. We conducted a Phase I trial (NCT02107963) of GD2 CAR-Ts (GD2-CAR.OX40.28.z.iC9), demonstrating feasibility and safety of administration in children and young adults with osteosarcoma and neuroblastoma. Since CAR-T efficacy requires adequate CAR-T expansion, patients were grouped into good or poor expanders across dose levels. Patient samples were evaluated by multi-dimensional proteomic, transcriptomic, and epigenetic analyses. T cell assessments identified naive T cells in pre-treatment apheresis associated with good expansion, and exhausted T cells in CAR-T products with poor expansion. Myeloid cell assessment identified CXCR3+ monocytes in pre-treatment apheresis associated with good expansion. Longitudinal analysis of post-treatment samples identified increased CXCR3- classical monocytes in all groups as CAR-T numbers waned. Together, our data uncover mediators of CAR-T biology and correlates of expansion that could be utilized to advance immunotherapies for solid tumor patients.
Collapse
Affiliation(s)
- Sabina Kaczanowska
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tara Murty
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Ahmad Alimadadi
- La Jolla Institute for Immunology, La Jolla, CA, USA; Immunology Center of Georgia, Augusta University, Augusta, GA, USA; Georgia Cancer Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Cristina F Contreras
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Department of Oncology, University of Oxford, Oxford, UK
| | - Caroline Duault
- Stanford Human Immune Monitoring Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Priyanka B Subrahmanyam
- Stanford Human Immune Monitoring Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Warren Reynolds
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Reema Baskar
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Catherine J Wu
- Broad Institute, Cambridge, MA, USA; Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | - Yang Liu
- Broad Institute, Cambridge, MA, USA
| | | | - Vandon Duong
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Hima Anbunathan
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Claire Ong
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hua Zhang
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Radim Moravec
- Cancer Therapy Evaluation Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Joyce Yu
- Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | | | - Mina Pichavant
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA
| | - Elena Sotillo
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Donna Bernstein
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amanda Carbonell
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Joanne Derdak
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jacquelyn Klicka-Skeels
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Julia E Segal
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eva Dombi
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephanie A Harmon
- Artificial Intelligence Resource, Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Baris Turkbey
- Artificial Intelligence Resource, Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bita Sahaf
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Sean Bendall
- Georgia Cancer Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Holden Maecker
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA
| | - Steven L Highfill
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, MD, USA
| | - David Stroncek
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, MD, USA
| | - John Glod
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Melinda Merchant
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Catherine C Hedrick
- La Jolla Institute for Immunology, La Jolla, CA, USA; Immunology Center of Georgia, Augusta University, Augusta, GA, USA; Georgia Cancer Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Crystal L Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Sneha Ramakrishna
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
| | - Rosandra N Kaplan
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
9
|
Tong C, Wu Y, Zhuang Z, Wang Z, Yu Y. Combining proteomic markers to construct a logistic regression model for polycystic ovary syndrome. Front Endocrinol (Lausanne) 2023; 14:1227252. [PMID: 37854181 PMCID: PMC10579946 DOI: 10.3389/fendo.2023.1227252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/11/2023] [Indexed: 10/20/2023] Open
Abstract
Introduction Proteomics technology has been used in various fields in recent years for the Q6 exploration of novel markers and the study of disease pathogenesis, and has become one of the most important tools for researchers to explore unknown areas. However, there are fewer studies related to the construction of clinical models using proteomics markers. Methods In our previous study we used DIA proteomics to screen for proteins that were significant in 31 PCOS patients compared to women of normal reproductive age. In this study, we used logistic regression among these protein markers to screen out variables with diagnostic value and constructed logistic regression models. Results We constructed a logistic model using these protein markers, where HIST1H4A (OR=1.037) was an independent risk factor for polycystic ovary syndrome and TREML1 (OR=0.976) were protective factors for the disease. The logistic regression model equation is: Logit (PCOS) =0.036*[HIST1H4A]-0.024*[TREML1]-16.368. The ROC curve analyzing the diagnostic value of the model has an AUC value of 0.977 and a Youden index of0.903, which gives a cutoff value of 0.518 at this point. The model has a sensitivity of 93.5% and a specificity of 96.8%. Calibration curves show fair consistency of the model. Discussion Our study is the first to use proteomic results with clinical biochemical data to construct a logistic regression model, and the model is consistent. However, our study still needs a more complete sample to confirm our findings.
Collapse
Affiliation(s)
- Cheng Tong
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yue Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Zhenchao Zhuang
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Zhejiong Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Ying Yu
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
10
|
Li M, Wang M, Wen Y, Zhang H, Zhao G, Gao Q. Signaling pathways in macrophages: molecular mechanisms and therapeutic targets. MedComm (Beijing) 2023; 4:e349. [PMID: 37706196 PMCID: PMC10495745 DOI: 10.1002/mco2.349] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 09/15/2023] Open
Abstract
Macrophages play diverse roles in development, homeostasis, and immunity. Accordingly, the dysfunction of macrophages is involved in the occurrence and progression of various diseases, such as coronavirus disease 2019 and atherosclerosis. The protective or pathogenic effect that macrophages exert in different conditions largely depends on their functional plasticity, which is regulated via signal transduction such as Janus kinase-signal transducer and activator of transcription, Wnt and Notch pathways, stimulated by environmental cues. Over the past few decades, the molecular mechanisms of signaling pathways in macrophages have been gradually elucidated, providing more alternative therapeutic targets for diseases treatment. Here, we provide an overview of the basic physiology of macrophages and expound the regulatory pathways within them. We also address the crucial role macrophages play in the pathogenesis of diseases, including autoimmune, neurodegenerative, metabolic, infectious diseases, and cancer, with a focus on advances in macrophage-targeted strategies exploring modulation of components and regulators of signaling pathways. Last, we discuss the challenges and possible solutions of macrophage-targeted therapy in clinical applications. We hope that this comprehensive review will provide directions for further research on therapeutic strategies targeting macrophage signaling pathways, which are promising to improve the efficacy of disease treatment.
Collapse
Affiliation(s)
- Ming Li
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Mengjie Wang
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuanjia Wen
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hongfei Zhang
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Guang‐Nian Zhao
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Qinglei Gao
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
11
|
Campbell GR, Rawat P, To RK, Spector SA. HIV-1 Tat Upregulates TREM1 Expression in Human Microglia. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:429-442. [PMID: 37326481 PMCID: PMC10352590 DOI: 10.4049/jimmunol.2300152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/30/2023] [Indexed: 06/17/2023]
Abstract
Because microglia are a reservoir for HIV and are resistant to the cytopathic effects of HIV infection, they are a roadblock for any HIV cure strategy. We have previously identified that triggering receptor expressed on myeloid cells 1 (TREM1) plays a key role in human macrophage resistance to HIV-mediated cytopathogenesis. In this article, we show that HIV-infected human microglia express increased levels of TREM1 and are resistant to HIV-induced apoptosis. Moreover, upon genetic inhibition of TREM1, HIV-infected microglia undergo cell death in the absence of increased viral or proinflammatory cytokine expression or the targeting of uninfected cells. We also show that the expression of TREM1 is mediated by HIV Tat through a TLR4, TICAM1, PG-endoperoxide synthase 2, PGE synthase, and PGE2-dependent manner. These findings highlight the potential of TREM1 as a therapeutic target to eradicate HIV-infected microglia without inducing a proinflammatory response.
Collapse
Affiliation(s)
- Grant R. Campbell
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD
| | - Pratima Rawat
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA
| | - Rachel K. To
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA
| | - Stephen A. Spector
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA
- Rady Children’s Hospital, San Diego, CA
| |
Collapse
|
12
|
Vandestienne M, Braik R, Lavillegrand JR, Hariri G, Demailly Z, Ben Hamouda N, Tamion F, Clavier T, Ait-Oufella H. Soluble TREM-1 plasma levels are associated with acute kidney injury, acute atrial fibrillation and prolonged ICU stay after cardiac surgery- a proof-concept study. Front Cardiovasc Med 2023; 10:1098914. [PMID: 37522081 PMCID: PMC10373879 DOI: 10.3389/fcvm.2023.1098914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Background Cardiopulmonary bypass (CPB) during cardiac surgery leads to deleterious systemic inflammation. We hypothesized that TREM-1, a myeloid receptor shed after activation, drives systemic inflammation during CPB. Methods Prospective observational bi-centric study. Blood analysis (flow cytometry and ELISA) before and at H2 and H24 after CPB. Inclusion of adult patients who underwent elective cardiac surgery with CPB. Results TREM-1 expression on neutrophils decreased between H0 and H2 while soluble (s)TREM-1 plasma levels increased. sTREM-1 levels increased at H2 and at H24 (p < 0.001). IL-6, IL-8, G-CSF and TNF-α, but not IL-1β, significantly increased at H2 compared to H0 (p < 0.001), but dropped at H24. Principal component analysis showed a close relationship between sTREM-1 and IL-8. Three patterns of patients were identified: Profile 1 with high baseline sTREM-1 levels and high increase and profile 2/3 with low/moderate baseline sTREM-1 levels and no/moderate increase overtime. Profile 1 patients developed more severe organ failure after CPB, with higher norepinephrine dose, higher SOFA score and more frequently acute kidney injury at both H24 and H48. Acute atrial fibrillation was also more frequent in profile 1 patients at H24 (80% vs. 19.4%, p = 0.001). After adjustment on age and duration of CPB, H0, H2 and H24 sTREM-1 levels remained associated with prolonged ICU and hospital length of stay. Conclusions Baseline sTREM-1 levels as well as early kinetics after cardiac surgery identified patients at high risk of post-operative complications and prolonged length of stay.
Collapse
Affiliation(s)
- Marie Vandestienne
- Centre de Recherche Cardiovasculaire de Paris (PARCC), Université de Paris, Inserm U970, Paris, France
| | - Rayan Braik
- Centre de Recherche Cardiovasculaire de Paris (PARCC), Université de Paris, Inserm U970, Paris, France
| | - Jean-Rémi Lavillegrand
- Centre de Recherche Cardiovasculaire de Paris (PARCC), Université de Paris, Inserm U970, Paris, France
| | - Geoffroy Hariri
- Service de Réanimation Chirurgicale, Assistance Publique – Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpétrière, Institut du Coeur, Paris, France
| | - Zoe Demailly
- Université de Normandie, UNIROUEN, Inserm U1096, FHU REMOD-VHF, Rouen, France
- Service D’anesthésie-Réanimation Chirurgicale, CHU De Rouen, Rouen, France
| | - Nadine Ben Hamouda
- Service D’Immunologie, Hôpital Européen Georges Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Fabienne Tamion
- Université de Normandie, UNIROUEN, Inserm U1096, FHU REMOD-VHF, Rouen, France
- Service de Médecine Intensive-Réanimation, CHU De Rouen, Rouen, France
| | - Thomas Clavier
- Université de Normandie, UNIROUEN, Inserm U1096, FHU REMOD-VHF, Rouen, France
- Service D’anesthésie-Réanimation Chirurgicale, CHU De Rouen, Rouen, France
| | - Hafid Ait-Oufella
- Centre de Recherche Cardiovasculaire de Paris (PARCC), Université de Paris, Inserm U970, Paris, France
- Service de Médecine Intensive-Réanimation, Assistance Publique – Hôpitaux de Paris, Sorbonne Université, Paris, France
- Hôpital Saint-Antoine, Sorbonne Université, Paris, France
| |
Collapse
|
13
|
Abstract
Alzheimer's disease (AD) is a debilitating age-related neurodegenerative condition. Unbiased genetic studies have implicated a central role for microglia, the resident innate immune cells of the central nervous system, in AD pathogenesis. On-going efforts are clarifying the biology underlying these associations and the microglial pathways that are dysfunctional in AD. Several genetic risk factors converge to decrease the function of activating microglial receptors and increase the function of inhibitory receptors, resulting in a seemingly dampened microglial phenotype in AD. Moreover, many of these microglial proteins that are genetically associated with AD appear to interact and share pathways or regulatory mechanisms, presenting several points of convergence that may be strategic targets for therapeutic intervention. Here, we review some of these studies and their implications for microglial participation in AD pathogenesis.
Collapse
|
14
|
Campbell GR, Spector SA. Current strategies to induce selective killing of HIV-1-infected cells. J Leukoc Biol 2022; 112:1273-1284. [PMID: 35707952 PMCID: PMC9613504 DOI: 10.1002/jlb.4mr0422-636r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/24/2022] [Indexed: 01/02/2023] Open
Abstract
Although combination antiretroviral therapy (ART) has led to significant HIV-1 suppression and improvement in immune function, persistent viral reservoirs remain that are refractory to intensified ART. ART poses many challenges such as adherence to drug regimens, the emergence of resistant virus, and cumulative toxicity resulting from long-term therapy. Moreover, latent HIV-1 reservoir cells can be stochastically activated to produce viral particles despite effective ART and contribute to the rapid viral rebound that typically occurs within 2 weeks of ART interruption; thus, lifelong ART is required for continued viral suppression. Several strategies have been proposed to address the HIV-1 reservoir such as reactivation of HIV-1 transcription using latency reactivating agents with a combination of ART, host immune clearance and HIV-1-cytotoxicity to purge the infected cells-a "shock and kill" strategy. However, these approaches do not take into account the multiple transcriptional and translational blocks that contribute to HIV-1 latency or the complex heterogeneity of the HIV-1 reservoir, and clinical trials have thus far failed to produce the desired results. Here, we describe alternative strategies being pursued that are designed to kill selectively HIV-1-infected cells while sparing uninfected cells in the absence of enhanced humoral or adaptive immune responses.
Collapse
Affiliation(s)
- Grant R. Campbell
- Department of PediatricsDivision of Infectious DiseasesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Stephen A. Spector
- Department of PediatricsDivision of Infectious DiseasesUniversity of California San DiegoLa JollaCaliforniaUSA,Division of Infectious DiseasesRady Children's HospitalSan DiegoCaliforniaUSA
| |
Collapse
|
15
|
Panagopoulos A, Samant S, Bakhos JJ, Liu M, Khan B, Makadia J, Muhammad F, Kievit FM, Agrawal DK, Chatzizisis YS. Triggering receptor expressed on myeloid cells-1 (TREM-1) inhibition in atherosclerosis. Pharmacol Ther 2022; 238:108182. [DOI: 10.1016/j.pharmthera.2022.108182] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 03/14/2022] [Accepted: 03/30/2022] [Indexed: 11/29/2022]
|
16
|
Sîrbe C, Rednic S, Grama A, Pop TL. An Update on the Effects of Vitamin D on the Immune System and Autoimmune Diseases. Int J Mol Sci 2022; 23:9784. [PMID: 36077185 PMCID: PMC9456003 DOI: 10.3390/ijms23179784] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 12/16/2022] Open
Abstract
Vitamin D intervenes in calcium and phosphate metabolism and bone homeostasis. Experimental studies have shown that 1,25-dihydroxyvitamin D (calcitriol) generates immunologic activities on the innate and adaptive immune system and endothelial membrane stability. Low levels of serum 25-hydroxyvitamin D (25(OH)D) are associated with an increased risk of developing immune-related diseases such as psoriasis, type 1 diabetes, multiple sclerosis, and autoimmune diseases. Various clinical trials describe the efficacy of supplementation of vitamin D and its metabolites for treating these diseases that result in variable outcomes. Different disease outcomes are observed in treatment with vitamin D as high inter-individual difference is present with complex gene expression in human peripheral blood mononuclear cells. However, it is still not fully known what level of serum 25(OH)D is needed. The current recommendation is to increase vitamin D intake and have enough sunlight exposure to have serum 25(OH)D at a level of 30 ng/mL (75 nmol/L) and better at 40-60 ng/mL (100-150 nmol/L) to obtain the optimal health benefits of vitamin D.
Collapse
Affiliation(s)
- Claudia Sîrbe
- 2nd Pediatric Discipline, Department of Mother and Child, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- 2nd Pediatric Clinic, Emergency Clinical Hospital for Children, 400177 Cluj-Napoca, Romania
| | - Simona Rednic
- Rheumatology Department, Emergency County Hospital Cluj, 400347 Cluj-Napoca, Romania
- Rheumatology Discipline, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Alina Grama
- 2nd Pediatric Discipline, Department of Mother and Child, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- 2nd Pediatric Clinic, Emergency Clinical Hospital for Children, 400177 Cluj-Napoca, Romania
| | - Tudor Lucian Pop
- 2nd Pediatric Discipline, Department of Mother and Child, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- 2nd Pediatric Clinic, Emergency Clinical Hospital for Children, 400177 Cluj-Napoca, Romania
| |
Collapse
|
17
|
Sigalov AB. Inhibition of TREM-2 Markedly Suppresses Joint Inflammation and Damage in Experimental Arthritis. Int J Mol Sci 2022; 23:ijms23168857. [PMID: 36012120 PMCID: PMC9408405 DOI: 10.3390/ijms23168857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/04/2022] [Accepted: 08/07/2022] [Indexed: 11/16/2022] Open
Abstract
The triggering receptors expressed on myeloid cells (TREMs) are a family of activating immune receptors that regulate the inflammatory response. TREM-1, which is expressed on monocytes and/or macrophages and neutrophils, functions as an inflammation amplifier and plays a role in the pathogenesis of rheumatoid arthritis (RA). Unlike TREM-1, the role in RA of TREM-2, which is expressed on macrophages, immature monocyte-derived dendritic cells, osteoclasts, and microglia, remains unclear and controversial. TREM-2 ligands are still unknown, adding further uncertainty to our understanding of TREM-2 function. Previously, we demonstrated that TREM-1 blockade, using a ligand-independent TREM-1 inhibitory peptide sequence GF9 rationally designed by our signaling chain homooligomerization (SCHOOL) model of cell signaling, ameliorates collagen-induced arthritis (CIA) severity in mice. Here, we designed a TREM-2 inhibitory peptide sequence IA9 and tested it in the therapeutic CIA model, either as a free 9-mer peptide IA9, or as a part of a 31-mer peptide IA31 incorporated into lipopeptide complexes (IA31-LPC), for targeted delivery. We demonstrated that administration of IA9, but not a control peptide, after induction of arthritis diminished release of proinflammatory cytokines and dramatically suppressed joint inflammation and damage, suggesting that targeting TREM-2 may be a promising approach for the treatment of RA.
Collapse
|
18
|
Siskind S, Brenner M, Wang P. TREM-1 Modulation Strategies for Sepsis. Front Immunol 2022; 13:907387. [PMID: 35784361 PMCID: PMC9240770 DOI: 10.3389/fimmu.2022.907387] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/17/2022] [Indexed: 12/28/2022] Open
Abstract
The triggering receptor expressed on myeloid cells-1 (TREM-1) is a pattern recognition receptor, which can be upregulated in inflammatory diseases as an amplifier of immune responses. Once activated, TREM-1 induces the production and release of pro-inflammatory cytokines and chemokines, in addition to increasing its own expression and circulating levels of the cleaved soluble extracellular portion of TREM-1 (sTREM-1). This amplification of the inflammatory response by TREM-1 has now been considered as a critical contributor to the dysregulated immune responses in sepsis. Studies have shown that in septic patients there is an elevated expression of TREM-1 on immune cells and increased circulating levels of sTREM-1, associated with increased mortality. As a result, a considerable effort has been made towards identifying endogenous ligands of TREM-1 and developing TREM-1 inhibitory peptides to attenuate the exacerbated inflammatory response in sepsis. TREM-1 modulation has proven a promising strategy for the development of therapeutic agents to treat sepsis. Therefore, this review encompasses the ligands investigated as activators of TREM-1 thus far and highlights the development and efficacy of novel inhibitors for the treatment of sepsis and septic shock.
Collapse
Affiliation(s)
- Sara Siskind
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
| | - Max Brenner
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
- *Correspondence: Ping Wang, ; Max Brenner,
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
- *Correspondence: Ping Wang, ; Max Brenner,
| |
Collapse
|
19
|
Perez-Sanchez C, Barbera Betancourt A, Lyons PA, Zhang Z, Suo C, Lee JC, McKinney EF, Modis LK, Ellson C, Smith KG. miR-374a-5p regulates inflammatory genes and monocyte function in patients with inflammatory bowel disease. J Exp Med 2022; 219:e20211366. [PMID: 35363256 PMCID: PMC8980842 DOI: 10.1084/jem.20211366] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/23/2021] [Accepted: 02/17/2022] [Indexed: 02/02/2023] Open
Abstract
MicroRNAs are critical regulators of gene expression controlling cellular processes including inflammation. We explored their role in the pathogenesis of inflammatory bowel disease (IBD) and identified reduced expression of miR-374a-5p in IBD monocytes that correlated with a module of up-regulated genes related to the inflammatory response. Key proinflammatory module genes, including for example TNFα, IL1A, IL6, and OSM, were inversely correlated with miR-374a-5p and were validated in vitro. In colonic biopsies, miR-374a-5p was again reduced in expression and inversely correlated with the same inflammatory module, and its levels predicted subsequent response to anti-TNF therapy. Increased miR-374a-5p expression was shown to control macrophage-driven inflammation by suppressing proinflammatory mediators and to reduce the capacity of monocytes to migrate and activate T cells. Our findings suggest that miR-374a-5p reduction is a central driver of inflammation in IBD, and its therapeutic supplementation could reduce monocyte-driven inflammation in IBD or other immune-mediated diseases.
Collapse
Affiliation(s)
- Carlos Perez-Sanchez
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Rheumatology Service, Reina Sofia University Hospital, Maimonides Biomedical Research Institute of Córdoba, University of Cordoba, Cordoba, Spain
| | - Ariana Barbera Betancourt
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Paul A. Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Zinan Zhang
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology and Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Chenqu Suo
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Department of Paediatrics, Cambridge University Hospitals, Cambridge, UK
| | - James C. Lee
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Eoin F. McKinney
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | | | | | - Kenneth G.C. Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
20
|
Cao L, Yang K. Paeoniflorin Attenuated TREM-1-Mediated Inflammation in THP-1 Cells. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:7051643. [PMID: 35480155 PMCID: PMC9038380 DOI: 10.1155/2022/7051643] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/04/2022] [Accepted: 03/13/2022] [Indexed: 12/04/2022]
Abstract
Sepsis is caused by bacterial infections or viral infections. Clinically, there exist confirmed or highly suspected infection foci. Mortality caused by septic shock remains in a high rate even though antibiotic treatment works effectively. In this study, we treat THP-1 cells with 1 ug/mL LPS (lipopolysaccharide) and add paeoniflorin or LR-12 inhibitor. TREM-1 (triggering receptor expressed on myeloid cells-1), IL-6, IL-1β, and TNF-α (tumour necrosis factor alpha (a)-cachectin) were detected by ELISA and qRT-PCR, and western blotting is performed to detect related proteins in the NF-κB signaling pathway. As a result, paeoniflorin can significantly reduce the production of LPS-stimulated TREM-1 as well as inflammatory factors and attenuate the phosphorylation of NF-κB signaling pathway-related factors, such as p65 and IκBα. At the same time, the combined effect of paeoniflorin and LR-12 is more significant. The results of this study solidly prove that paeoniflorin plays a role in inhibiting TREM-1-mediated inflammation and the NF-κB pathway could be a potential mechanism of action.
Collapse
Affiliation(s)
- Li Cao
- Department of Critical Medicine, Shenzhen Baoan Shiyan People's Hospital, Shenzhen 5515108, China
| | - Kerong Yang
- Department of Suzhou Medical College, Soochow University, Suzhou 215123, China
| |
Collapse
|
21
|
Wang TY, Feng JY, Shu CC, Lee SSJ, Chen CY, Wei YF, Lin CB, Huang WC, Su WJ, Lin SM. Plasma Concentrations of sTREM-1 as Markers for Systemic Adverse Reactions in Subjects Treated With Weekly Rifapentine and Isoniazid for Latent Tuberculosis Infection. Front Microbiol 2022; 13:821066. [PMID: 35308376 PMCID: PMC8927064 DOI: 10.3389/fmicb.2022.821066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Background A regimen of once-weekly rifapentine plus isoniazid for 3 months (3HP) is an effective treatment for subjects with latent tuberculosis infection; however, no reliable biomarker exists for predicting systemic adverse reactions (SARs) to 3HP treatment. Methods This prospective, multi-center study evaluated the plasma concentrations of soluble triggering receptors expressed on myeloid cells (sTREM)-1 and sTREM-2 in subjects undergoing 3HP treatment and examined the associations between these biomarkers and SARs. Results This study enrolled 80 consecutive subjects receiving 3HP treatment, 25 of whom had SARs and 55 of whom did not. Subjects with SARs presented higher concentrations of sTREM-1 at baseline than those without SARs (240.1 ± 19.1 vs. 176.7 ± 9.4 pg/mL, P = 0.001). The area under the receiver operating characteristic curves revealed that day 1 plasma levels of sTREM-1 (0.708, 95% CI, 0.584–0.833, P = 0.003) and sTREM-2 (0.343, 95% CI, 0.227–0.459, P = 0.025) as well as the sTREM-1/sTREM-2 ratio (0.748, 95% CI, 0.638–0.858, P = 0.001) had modest discriminative power pertaining to the development of SARs. An sTREM-1 level exceeding the cut-off value (>187.4 pg/mL) (hazard ratio [HR], 6.15; 95% CI 1.67–22.70, P = 0.006) and a sTREM-2 below the cut-off value (<237.2 pg/mL) (HR, 4.46; 95% CI 1.41–14.1, P = 0.011) were independent predictors of SARs after controlling for other variables. Conclusions Plasma sTREM-1 and sTREM-2 levels are useful biomarkers for predicting SARs during 3HP treatment. Clinical trial government NCT04655794
Collapse
Affiliation(s)
- Tsai-Yu Wang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital Linkou Main Branch, Taoyuan, Taiwan.,School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jia-Yih Feng
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Chin-Chung Shu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Susan Shin-Jung Lee
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan.,Division of Infectious Diseases, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Chung-Yu Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Yu-Feng Wei
- Division of Chest Medicine, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Chih-Bin Lin
- Division of Chest Medicine, Department of Internal Medicine, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Wei-Chang Huang
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan.,Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Wei-Juin Su
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shu-Min Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital Linkou Main Branch, Taoyuan, Taiwan.,School of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
22
|
Zhang X, Moore CM, Harmacek LD, Domenico J, Rangaraj VR, Ideozu JE, Knapp JR, Woods KJ, Jump S, Jia S, Prokop JW, Bowler R, Hessner MJ, Gelfand EW, Levy H. CFTR-mediated monocyte/macrophage dysfunction revealed by cystic fibrosis proband-parent comparisons. JCI Insight 2022; 7:152186. [PMID: 35315363 PMCID: PMC8986072 DOI: 10.1172/jci.insight.152186] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 02/02/2022] [Indexed: 12/23/2022] Open
Abstract
Cystic fibrosis (CF) is an inherited disorder caused by biallelic mutations of the CF transmembrane conductance regulator (CFTR) gene. Converging evidence suggests that CF carriers with only 1 defective CFTR copy are at increased risk for CF-related conditions and pulmonary infections, but the molecular mechanisms underpinning this effect remain unknown. We performed transcriptomic profiling of peripheral blood mononuclear cells (PBMCs) of CF child-parent trios (proband, father, and mother) and healthy control (HC) PBMCs or THP-1 cells incubated with the plasma of these participants. Transcriptomic analyses revealed suppression of cytokine-enriched immune-related genes (IL-1β, CXCL8, CREM), implicating lipopolysaccharide tolerance in innate immune cells (monocytes) of CF probands and their parents. These data suggest that a homozygous as well as a heterozygous CFTR mutation can modulate the immune/inflammatory system. This conclusion is further supported by the finding of lower numbers of circulating monocytes in CF probands and their parents, compared with HCs, and the abundance of mononuclear phagocyte subsets, which correlated with Pseudomonas aeruginosa infection, lung disease severity, and CF progression in the probands. This study provides insight into demonstrated CFTR-related innate immune dysfunction in individuals with CF and carriers of a CFTR mutation that may serve as a target for personalized therapy.
Collapse
Affiliation(s)
- Xi Zhang
- Data Science program, Weinberg College of Arts and Sciences, Northwestern University, Evanston, Illinois, USA.,Division of Pediatric Pulmonary Medicine, Department of Pediatrics, and
| | - Camille M Moore
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA
| | - Laura D Harmacek
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA
| | - Joanne Domenico
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, and
| | - Vittobai Rashika Rangaraj
- Division of Pulmonary & Sleep Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Justin E Ideozu
- Genomic Medicine, Genomics Research Center, AbbVie, North Chicago, Illinois, USA
| | - Jennifer R Knapp
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA
| | - Katherine J Woods
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Stephanie Jump
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Shuang Jia
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Max McGee Center for Juvenile Diabetes, Children's Research Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jeremy W Prokop
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Russell Bowler
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Martin J Hessner
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Max McGee Center for Juvenile Diabetes, Children's Research Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin, USA
| | - Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA.,Division of Immunology, Microbiology and Pediatrics, Department of Pediatrics, University of Colorado School of Medicine, Denver, Colorado, USA
| | - Hara Levy
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, and
| |
Collapse
|
23
|
Borjas T, Jacob A, Yen H, Patel V, Coppa G, Aziz M, Wang P. Inhibition of the Interaction of TREM-1 and eCIRP Attenuates Inflammation and Improves Survival in Hepatic Ischemia/Reperfusion. Shock 2022; 57:246-255. [PMID: 34864782 PMCID: PMC8758526 DOI: 10.1097/shk.0000000000001894] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Triggering receptor expressed on myeloid cells-1 (TREM-1) has important implications in sepsis and inflammation and is a novel receptor for extracellular cold-inducible RNA-binding protein (eCIRP). We hypothesize that the inhibition of TREM-1 via its interaction with eCIRP by novel peptide inhibitor M3 or knockout gene will attenuate the inflammation and injury associated with severe hepatic ischemia/reperfusion (I/R). METHODS Wild-type (WT) C57BL/6 and TREM-1-/- mice underwent 60 min of 70% hepatic ischemia, with 24 h of reperfusion. Additionally, WT mice underwent hepatic I/R and were treated with M3 (10 mg/kg body weight) or vehicle (normal saline) at the start of reperfusion. Blood and ischemic liver tissues were collected, and analysis was performed using enzymatic assays, enzyme-linked immunosorbent assay, reverse-transcription quantitative polymerase chain reaction, and pathohistology techniques. For survival surgery, mice additionally underwent resection of non-ischemic lobes of the liver and survival was monitored for 10 days. RESULTS There was an increase in serum levels of tissue markers including aspartate aminotransferase, alanine aminotransferase, and lactate dehydrogenase as well as cytokine levels (IL-6) and histological scoring of hematoxylin and eosin sections in WT I/R mice. These markers decreased substantially in TREM-1-/- mice. Additionally, neutrophil infiltration markers and markers of local inflammation (myeloperoxidase, macrophage inflammatory protein-2, cyclooxygenase-2) were attenuated in TREM-1-/- mice. Similarly, we show a significant decrease in injury and inflammation markers with M3 treatment. Additionally, we demonstrate decreased apoptosis with TREM-1 inhibition. Finally, M3 treatment improved the survival rate from 42% to 75% after hepatic I/R. CONCLUSION TREM-1 is an important eCIRP receptor in the inflammatory response of hepatic I/R, and deficiency of TREM-1 via knockout gene or peptide inhibition attenuated liver injury and inflammation, and improved survival. Inhibition of the TREM-1 and eCIRP interaction in hepatic I/R may have important therapeutic potential.
Collapse
Affiliation(s)
- Timothy Borjas
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY
| | - Asha Jacob
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY
| | - HaoTing Yen
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY
| | - Vihas Patel
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
| | - Gene Coppa
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
| | - Monowar Aziz
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY
| | - Ping Wang
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY
| |
Collapse
|
24
|
Vandestienne M, Joffre J, Lemarié J, Ait-Oufella H. [Role of TREM-1 in cardiovascular diseases]. Med Sci (Paris) 2022; 38:32-37. [PMID: 35060884 DOI: 10.1051/medsci/2021242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The innate immune system plays a crucial role in cardiovascular disease initiation, progression and complications. TREM-1, a receptor mainly expressed by myeloid cells, orchestrates inflammatory responses and amplifies cytokine and chemokine production as well as oxidative burst. Recent experimental studies have demonstrated that TREM-1 blockade is protective, limiting atherosclerosis and abdominal aortic aneurysm development, as well as adverse tissue remodeling after cardiac or cerebral ischemic injuries. Plasma soluble TREM-1 level is a promising biomarker in patients with cardiovascular diseases for risk stratification, paving the way for personalized immune-modulatory approaches.
Collapse
Affiliation(s)
- Marie Vandestienne
- Université de Paris, Inserm U970, PARCC (Paris Cardiovascular Research Center), Paris, France
| | - Jérémie Joffre
- Service de Médecine intensive-Réanimation, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France
| | - Jérémie Lemarié
- Université de Paris, Inserm U970, PARCC (Paris Cardiovascular Research Center), Paris, France - Service de Médecine intensive-Réanimation, CHU de Nantes, Nantes, France
| | - Hafid Ait-Oufella
- Université de Paris, Inserm U970, PARCC (Paris Cardiovascular Research Center), Paris, France - Service de Médecine intensive-Réanimation, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France
| |
Collapse
|
25
|
Pullikuth AK, Routh ED, Zimmerman KD, Chifman J, Chou JW, Soike MH, Jin G, Su J, Song Q, Black MA, Print C, Bedognetti D, Howard-McNatt M, O’Neill SS, Thomas A, Langefeld CD, Sigalov AB, Lu Y, Miller LD. Bulk and Single-Cell Profiling of Breast Tumors Identifies TREM-1 as a Dominant Immune Suppressive Marker Associated With Poor Outcomes. Front Oncol 2021; 11:734959. [PMID: 34956864 PMCID: PMC8692779 DOI: 10.3389/fonc.2021.734959] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
BackgroundTriggering receptor expressed on myeloid cells (TREM)-1 is a key mediator of innate immunity previously associated with the severity of inflammatory disorders, and more recently, the inferior survival of lung and liver cancer patients. Here, we investigated the prognostic impact and immunological correlates of TREM1 expression in breast tumors.MethodsBreast tumor microarray and RNAseq expression profiles (n=4,364 tumors) were analyzed for associations between gene expression, tumor immune subtypes, distant metastasis-free survival (DMFS) and clinical response to neoadjuvant chemotherapy (NAC). Single-cell (sc)RNAseq was performed using the 10X Genomics platform. Statistical associations were assessed by logistic regression, Cox regression, Kaplan-Meier analysis, Spearman correlation, Student’s t-test and Chi-square test.ResultsIn pre-treatment biopsies, TREM1 and known TREM-1 inducible cytokines (IL1B, IL8) were discovered by a statistical ranking procedure as top genes for which high expression was associated with reduced response to NAC, but only in the context of immunologically “hot” tumors otherwise associated with a high NAC response rate. In surgical specimens, TREM1 expression varied among tumor molecular subtypes, with highest expression in the more aggressive subtypes (Basal-like, HER2-E). High TREM1 significantly and reproducibly associated with inferior distant metastasis-free survival (DMFS), independent of conventional prognostic markers. Notably, the association between high TREM1 and inferior DMFS was most prominent in the subset of immunogenic tumors that exhibited the immunologically hot phenotype and otherwise associated with superior DMFS. Further observations from bulk and single-cell RNAseq analyses indicated that TREM1 expression was significantly enriched in polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) and M2-like macrophages, and correlated with downstream transcriptional targets of TREM-1 (IL8, IL-1B, IL6, MCP-1, SPP1, IL1RN, INHBA) which have been previously associated with pro-tumorigenic and immunosuppressive functions.ConclusionsTogether, these findings indicate that increased TREM1 expression is prognostic of inferior breast cancer outcomes and may contribute to myeloid-mediated breast cancer progression and immune suppression.
Collapse
Affiliation(s)
- Ashok K. Pullikuth
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Eric D. Routh
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kip D. Zimmerman
- Center for Precision Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Julia Chifman
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Mathematics and Statistics, American University, Washington, DC, United States
| | - Jeff W. Chou
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston Salem, NC, United States
- The Comprehensive Cancer Center of Wake Forest University, Winston Salem, NC, United States
| | - Michael H. Soike
- Department of Radiation Oncology, University of Alabama-Birmingham, Birmingham, AL, United States
| | - Guangxu Jin
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, United States
- The Comprehensive Cancer Center of Wake Forest University, Winston Salem, NC, United States
| | - Jing Su
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Qianqian Song
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, United States
- Center for Cancer Genomics and Precision Oncology, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Michael A. Black
- Department of Biochemistry, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Cristin Print
- Department of Molecular Medicine and Pathology and Maurice Wilkins Institute, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Davide Bedognetti
- Cancer Program, Sidra Medicine, Doha, Qatar & Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
| | - Marissa Howard-McNatt
- Surgical Oncology Service, Department of Surgery, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Stacey S. O’Neill
- The Comprehensive Cancer Center of Wake Forest University, Winston Salem, NC, United States
- Department of Pathology, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Alexandra Thomas
- The Comprehensive Cancer Center of Wake Forest University, Winston Salem, NC, United States
- Section of Hematology and Oncology, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston Salem, NC, United States
| | - Carl D. Langefeld
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston Salem, NC, United States
- The Comprehensive Cancer Center of Wake Forest University, Winston Salem, NC, United States
| | | | - Yong Lu
- The Comprehensive Cancer Center of Wake Forest University, Winston Salem, NC, United States
- Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Lance D. Miller
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, United States
- The Comprehensive Cancer Center of Wake Forest University, Winston Salem, NC, United States
- *Correspondence: Lance D. Miller,
| |
Collapse
|
26
|
Häger SC, Dias C, Sønder SL, Olsen AV, da Piedade I, Heitmann ASB, Papaleo E, Nylandsted J. Short-term transcriptomic response to plasma membrane injury. Sci Rep 2021; 11:19141. [PMID: 34580330 PMCID: PMC8476590 DOI: 10.1038/s41598-021-98420-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022] Open
Abstract
Plasma membrane repair mechanisms are activated within seconds post-injury to promote rapid membrane resealing in eukaryotic cells and prevent cell death. However, less is known about the regeneration phase that follows and how cells respond to injury in the short-term. Here, we provide a genome-wide study into the mRNA expression profile of MCF-7 breast cancer cells exposed to injury by digitonin, a mild non-ionic detergent that permeabilizes the plasma membrane. We focused on the early transcriptional signature and found a time-dependent increase in the number of differentially expressed (> twofold, P < 0.05) genes (34, 114 and 236 genes at 20-, 40- and 60-min post-injury, respectively). Pathway analysis highlighted a robust and gradual three-part transcriptional response: (1) prompt activation of immediate-early response genes, (2) activation of specific MAPK cascades and (3) induction of inflammatory and immune pathways. Therefore, plasma membrane injury triggers a rapid and strong stress and immunogenic response. Our meta-analysis suggests that this is a conserved transcriptome response to plasma membrane injury across different cell and injury types. Taken together, our study shows that injury has profound effects on the transcriptome of wounded cells in the regeneration phase (subsequent to membrane resealing), which is likely to influence cellular status and has been previously overlooked.
Collapse
Affiliation(s)
- Swantje Christin Häger
- Membrane Integrity, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Catarina Dias
- Membrane Integrity, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Stine Lauritzen Sønder
- Membrane Integrity, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - André Vidas Olsen
- Computational Biology Laboratory, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Isabelle da Piedade
- Computational Biology Laboratory, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Anne Sofie Busk Heitmann
- Membrane Integrity, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Elena Papaleo
- Computational Biology Laboratory, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
- Translational Disease Systems Biology, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Jesper Nylandsted
- Membrane Integrity, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark.
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3C, 2200, Copenhagen N, Denmark.
| |
Collapse
|
27
|
Vandestienne M, Zhang Y, Santos-Zas I, Al-Rifai R, Joffre J, Giraud A, Laurans L, Esposito B, Pinet F, Bruneval P, Raffort J, Lareyre F, Vilar J, Boufenzer A, Guyonnet L, Guerin C, Clauser E, Silvestre JS, Lang S, Soulat-Dufour L, Tedgui A, Mallat Z, Taleb S, Boissonnas A, Derive M, Chinetti G, Ait-Oufella H. TREM-1 orchestrates angiotensin II-induced monocyte trafficking and promotes experimental abdominal aortic aneurysm. J Clin Invest 2021; 131:142468. [PMID: 33258804 DOI: 10.1172/jci142468] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 11/20/2020] [Indexed: 01/16/2023] Open
Abstract
The triggering receptor expressed on myeloid cells 1 (TREM-1) drives inflammatory responses in several cardiovascular diseases but its role in abdominal aortic aneurysm (AAA) remains unknown. Our objective was to explore the role of TREM-1 in a mouse model of angiotensin II-induced (AngII-induced) AAA. TREM-1 expression was detected in mouse aortic aneurysm and colocalized with macrophages. Trem1 gene deletion (Apoe-/-Trem1-/-), as well as TREM-1 pharmacological blockade with LR-12 peptide, limited both AAA development and severity. Trem1 gene deletion attenuated the inflammatory response in the aorta, with a reduction of Il1b, Tnfa, Mmp2, and Mmp9 mRNA expression, and led to a decreased macrophage content due to a reduction of Ly6Chi classical monocyte trafficking. Conversely, antibody-mediated TREM-1 stimulation exacerbated Ly6Chi monocyte aorta infiltration after AngII infusion through CD62L upregulation and promoted proinflammatory signature in the aorta, resulting in worsening AAA severity. AngII infusion stimulated TREM-1 expression and activation on Ly6Chi monocytes through AngII receptor type I (AT1R). In human AAA, TREM-1 was detected and TREM1 mRNA expression correlated with SELL mRNA expression. Finally, circulating levels of sTREM-1 were increased in patients with AAA when compared with patients without AAA. In conclusion, TREM-1 is involved in AAA pathophysiology and may represent a promising therapeutic target in humans.
Collapse
Affiliation(s)
- Marie Vandestienne
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France
| | - Yujiao Zhang
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France
| | - Icia Santos-Zas
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France
| | - Rida Al-Rifai
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France
| | - Jeremie Joffre
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France
| | - Andreas Giraud
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France
| | - Ludivine Laurans
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France
| | - Bruno Esposito
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France
| | | | - Patrick Bruneval
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France.,Department of Anatomopathology, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Juliette Raffort
- Université Côte d'Azur, Centre Hospitalo-Universitaire (CHU), INSERM, C3M, Nice, France
| | - Fabien Lareyre
- Université Côte d'Azur, Centre Hospitalo-Universitaire (CHU), INSERM, C3M, Nice, France
| | - Jose Vilar
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France
| | | | - Lea Guyonnet
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, F-75006, Paris, France.,Institut Curie, Cytometry Platform F-75006, Paris, France.,Department of Infection and Immunity, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Coralie Guerin
- Université de Paris, Innovative Therapies in Haemostasis, INSERM, F-75006, Paris, France.,Institut Curie, Cytometry Platform F-75006, Paris, France.,Department of Infection and Immunity, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Eric Clauser
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France
| | | | - Sylvie Lang
- Cardiology Department, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France
| | - Laurie Soulat-Dufour
- Cardiology Department, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France
| | - Alain Tedgui
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France
| | - Ziad Mallat
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France.,Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Soraya Taleb
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France
| | - Alexandre Boissonnas
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses Cimi-Paris, Paris, France
| | | | - Giulia Chinetti
- Université Côte d'Azur, Centre Hospitalo-Universitaire (CHU), INSERM, C3M, Nice, France
| | - Hafid Ait-Oufella
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France.,Medical Intensive Care Unit, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France
| |
Collapse
|
28
|
Soluble Triggering Receptor on Myeloid Cell-1 and its Predictive Factors in Patients With End-Stage Kidney Disease on Hemodialysis. ASAIO J 2021; 68:605-609. [PMID: 34352818 DOI: 10.1097/mat.0000000000001541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Triggering receptor expressed on myeloid cells (TREM)-1 is a potent and early amplifier of the inflammatory response expressed on neutrophils and monocytes/macrophages. TREM-1, and its soluble form (sTREM-1), are increased in sepsis and other noninfectious inflammatory conditions. However, virtually no data are available in kidney disease. To determine serum sTREM-1 and its associated variables in patients on hemodialysis (HD), cross-sectional study including 264 HD patients and 148 controls. sTREM-1 was measured by quantitative sandwich enzyme immunoassay; soluble tumor necrosis factor receptor-1 (sTNF-R1), interleukin-6 (IL-6), and C-reactive protein (CRP) were also measured. All inflammation markers were significantly higher in HD patients than controls. Median (IQR) sTREM-1 was 1,006 (613-1,650) pg/mL but undetectable in controls. Considering only HD patients, sTREM-1 was positively correlated with IL-6 (r = 0.19, p = 0.008), and its levels were significantly higher in patients with arteriovenous fistula than in those with temporary catheter (1,226 vs. 743 pg/mL), in patients with 3 HD sessions/week than in those with 2 sessions/week (1,150 vs. 646 pg/mL), and in patients with >1 year on HD than in those with ≤1 year (1,100 vs. 948 pg/mL), whereas they were not different regarding age or presence of infection. Serum sTREM-1, sTNF-R1, IL-6, and CRP were higher in HD patients compared to controls. In HD patients, sTREM-1 displayed higher levels in individuals with arteriovenous fistula, 3 sessions/week and longer vintage, but not in those with infection or older age; in multivariate analysis, only the first two variables significantly predicted higher sTREM-1 levels.
Collapse
|
29
|
Activation of TREM-1 induces endoplasmic reticulum stress through IRE-1α/XBP-1s pathway in murine macrophages. Mol Immunol 2021; 135:294-303. [PMID: 33957479 DOI: 10.1016/j.molimm.2021.04.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 04/16/2021] [Accepted: 04/26/2021] [Indexed: 12/14/2022]
Abstract
Increasing evidence suggests that endoplasmic reticulum (ER) stress activates several pro-inflammatory signaling pathways in many diseases, including acute lung injury (ALI). We have reported that blocking triggering receptor expressed on myeloid cells 1 (TREM-1) protects against ALI by suppressing pulmonary inflammation in mice with ALI induced by lipopolysaccharides (LPS). However, the molecular mechanism underlying the TREM-1-induced pro-inflammatory microenvironment in macrophages remains unclearly. Herein, we aimed to determine whether TREM-1 regulates the inflammatory responses induced by LPS associated with ER stress activation. We found that the activation of TREM-1 by a monoclonal agonist antibody (anti-TREM-1) increased the mRNA and protein levels of IL-1β, TNF-α, and IL-6 in primary macrophages. Treatment of the anti-TREM-1 antibody increased the expression of ER stress markers (ATF6, PERK, IRE-1α, and XBP-1s) in primary macrophages. While pretreatment with 4-PBA, an inhibitor of ER stress, significantly inhibited the expression of ER stress markers and pro-inflammatory cytokines and reduced LDH release. Furthermore, inhibiting the activity of the IRE-1α/XBP-1s pathway by STF-083010 significantly mitigated the increased levels of IL-1β, TNF-α, and IL-6 in macrophages treated by the anti-TREM-1 antibody. XBP-1 silencing attenuated pro-inflammatory microenvironment evoked by activation of TREM-1. Besides, we found that blockade of TREM-1 with LR12 ameliorated ER stress induced by LPS in vitro and in vivo. In conclusion, we conclude that TREM-1 activation induces ER stress through the IRE-1α/XBP-1s pathway in macrophages, contributing to the pro-inflammatory microenvironment.
Collapse
|
30
|
Hossian AKMN, Zahra FT, Poudel S, Abshire CF, Polk P, Garai J, Zabaleta J, Mikelis CM, Mattheolabakis G. Advanced bioinformatic analysis and pathway prediction of NSCLC cells upon cisplatin resistance. Sci Rep 2021; 11:6520. [PMID: 33753779 PMCID: PMC7985311 DOI: 10.1038/s41598-021-85930-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 03/05/2021] [Indexed: 11/08/2022] Open
Abstract
This study aims to identify pathway involvement in the development of cisplatin (cis-diamminedichloroplatinum (II); CDDP) resistance in A549 lung cancer (LC) cells by utilizing advanced bioinformatics software. We developed CDDP-resistant A549 (A549/DDP) cells through prolonged incubation with the drug and performed RNA-seq on RNA extracts to determine differential mRNA and miRNA expression between A549/DDP and A549 cells. We analyzed the gene dysregulation with Ingenuity Pathway Analysis (IPA; QIAGEN) software. In contrast to prior research, which relied on the clustering of dysregulated genes to pathways as an indication of pathway activity, we utilized the IPA software for the dynamic evaluation of pathway activity depending on the gene dysregulation levels. We predicted 15 pathways significantly contributing to the chemoresistance, with several of them to have not been previously reported or analyzed in detail. Among them, the PKR signaling, cholesterol biosynthesis, and TEC signaling pathways are included, as well as genes, such as PIK3R3, miR-34c-5p, and MDM2, among others. We also provide a preliminary analysis of SNPs and indels, present exclusively in A549/DDP cells. This study's results provide novel potential mechanisms and molecular targets that can be explored in future studies and assist in improving the understanding of the chemoresistance phenotype.
Collapse
Affiliation(s)
- A K M Nawshad Hossian
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Fatema Tuz Zahra
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Sagun Poudel
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Camille F Abshire
- Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Paula Polk
- Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Jone Garai
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Jovanny Zabaleta
- Department of Pediatrics and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA.
| |
Collapse
|
31
|
Singh H, Rai V, Nooti SK, Agrawal DK. Novel ligands and modulators of triggering receptor expressed on myeloid cells receptor family: 2015-2020 updates. Expert Opin Ther Pat 2021; 31:549-561. [PMID: 33507843 DOI: 10.1080/13543776.2021.1883587] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Triggering receptors expressed on myeloid cells (TREMs) are inflammatory amplifiers with defined pathophysiological role in various infectious diseases, acute and chronic aseptic inflammations, and a variety of cancers, depicting TREMs as prominent therapeutic targets.Areas covered: Herein, updates from 2015 to 2020 are discussed to divulge the TREM ligands, as well as their peptide blockers, claimed to modulate their expression. The article also presents different strategies employed during the last five years to block interactions between TREMs and their ligands to treat various disease conditions by modulating their expression and activity.Expert opinion: There has been significant progress in the discovery of novel ligands and modulators of TREMs in the last five years that mainly revolved around the function of TREM molecules. A few peptides showed encouraging results to modulate the expression and activity of TREMs in preclinical studies, and these peptides are currently under clinical investigation. Based on the findings so far in several careful studies, we expect novel therapeutics in the near future which could have the ability to treat various disease conditions associated with TREM expression.
Collapse
Affiliation(s)
- Harbinder Singh
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences , Pomona, California, USA
| | - Vikrant Rai
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences , Pomona, California, USA
| | - Sunil K Nooti
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences , Pomona, California, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences , Pomona, California, USA
| |
Collapse
|
32
|
Bernal-Martínez L, Gonçalves SM, de Andres B, Cunha C, Gonzalez Jimenez I, Lagrou K, Mellado E, Gaspar ML, Maertens JA, Carvalho A, Alcazar-Fuoli L. TREM1 regulates antifungal immune responses in invasive pulmonary aspergillosis. Virulence 2021; 12:570-583. [PMID: 33525982 PMCID: PMC7872058 DOI: 10.1080/21505594.2021.1879471] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pattern recognition receptors (PRRs) are responsible for Aspergillus fumigatus recognition by innate immunity and its subsequent immune signaling. The triggering receptor expressed on myeloid cells 1 (TREM1) is a recently characterized pro-inflammatory receptor constitutively expressed on the surface of neutrophils and macrophages. A soluble form (sTREM1) of this protein that can be detected in human body fluids has been identified. Here we investigated the role of TREM1 during invasive pulmonary aspergillosis (IPA). IPA patients displayed significantly higher levels of sTREM1 in bronchoalveolar lavages when compared to control patients. Functional analysis in TREM1 showed that the levels of sTREM1 and TREM1 pathway-related cytokines were influenced by single nucleotide polymorphisms in TREM1. In addition, we confirmed a role of TREM1 on antifungal host defense against A. fumigatus in a murine model of IPA. TREM1 deficiency increased susceptibility to infection in the immunosuppressed murine host. Deletion of TREM1 showed delayed innate and adaptive immune responses and impaired pro-inflammatory cytokine responses. The absence of TREM1 in primary macrophages attenuated the TLR signaling by altering the expression of both receptor and effector proteins that are critical to the response against A. fumigatus. In this study, and for the first time, we demonstrate the key role for the TREM1 receptor pathway during IPA.
Collapse
Affiliation(s)
- L Bernal-Martínez
- Mycology Reference Laboratory, National Centre for Microbiology, Instituto De Salud Carlos III , Madrid, Spain.,Spanish Network for the Research in Infectious Diseases (REIPI), Instituto de Salud Carlos III , Madrid, Spain
| | - S M Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães , Portugal
| | - B de Andres
- Department of Immunology, National Centre for Microbiology, Instituto De Salud Carlos III , Madrid, Spain
| | - C Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães , Portugal
| | - I Gonzalez Jimenez
- Mycology Reference Laboratory, National Centre for Microbiology, Instituto De Salud Carlos III , Madrid, Spain
| | - K Lagrou
- Department of Microbiology, Immunology, and Transplantation, Laboratory of Clinical Bacteriology and Mycology , KU Leuven, Leuven, Belgium.,Department of Laboratory Medicine and National Reference Center for Medical Mycology, University Hospitals Leuven , Leuven, Belgium
| | - E Mellado
- Mycology Reference Laboratory, National Centre for Microbiology, Instituto De Salud Carlos III , Madrid, Spain.,Spanish Network for the Research in Infectious Diseases (REIPI), Instituto de Salud Carlos III , Madrid, Spain
| | - M L Gaspar
- Department of Immunology, National Centre for Microbiology, Instituto De Salud Carlos III , Madrid, Spain
| | - J A Maertens
- Department of Microbiology, Immunology, and Transplantation, Laboratory of Clinical Bacteriology and Mycology , KU Leuven, Leuven, Belgium.,Department of Haematology, University Hospitals Leuven , Leuven, Belgium
| | - A Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães , Portugal
| | - L Alcazar-Fuoli
- Mycology Reference Laboratory, National Centre for Microbiology, Instituto De Salud Carlos III , Madrid, Spain.,Spanish Network for the Research in Infectious Diseases (REIPI), Instituto de Salud Carlos III , Madrid, Spain
| |
Collapse
|
33
|
Matos ADO, Dantas PHDS, Silva-Sales M, Sales-Campos H. TREM-1 isoforms in bacterial infections: to immune modulation and beyond. Crit Rev Microbiol 2021; 47:290-306. [PMID: 33522328 DOI: 10.1080/1040841x.2021.1878106] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The triggering receptor expressed on myeloid cells 1 (TREM-1) is an innate immunity receptor associated with the amplification of inflammation in sterile and non-sterile inflammatory disorders. Since its first description, the two isoforms of the receptor, membrane and soluble (mTREM-1 and sTREM-1, respectively) have been largely explored in the immunopathogenesis of several bacterial diseases and sepsis. The role of the receptor in these scenarios seems to be at least partly dependent on the source/type of bacteria, host and context. As uncontrolled inflammation is a result of several bacterial infections, the inhibition of the receptor has been considered as a promising approach to treat such conditions. Further, sTREM-1 has been explored as a biomarker for diagnosis and/or prognosis of several bacterial diseases. Therefore, this review aims to provide an updated insight into how the receptor influences and is influenced by bacterial infections, highlighting the advances regarding the use/manipulation of TREM-1 isoforms in biomedical research and clinical practice.
Collapse
Affiliation(s)
| | | | - Marcelle Silva-Sales
- Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil
| | | |
Collapse
|
34
|
Taylor SA, Chen SY, Gadhvi G, Feng L, Gromer KD, Abdala-Valencia H, Nam K, Dominguez ST, Montgomery AB, Reyfman PA, Ostilla L, Wechsler JB, Cuda CM, Green RM, Perlman H, Winter DR. Transcriptional profiling of pediatric cholestatic livers identifies three distinct macrophage populations. PLoS One 2021; 16:e0244743. [PMID: 33411796 PMCID: PMC7790256 DOI: 10.1371/journal.pone.0244743] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
Background & aims Limited understanding of the role for specific macrophage subsets in the pathogenesis of cholestatic liver injury is a barrier to advancing medical therapy. Macrophages have previously been implicated in both the mal-adaptive and protective responses in obstructive cholestasis. Recently two macrophage subsets were identified in non-diseased human liver; however, no studies to date fully define the heterogeneous macrophage subsets during the pathogenesis of cholestasis. Here, we aim to further characterize the transcriptional profile of macrophages in pediatric cholestatic liver disease. Methods We isolated live hepatic immune cells from patients with biliary atresia (BA), Alagille syndrome (ALGS), and non-cholestatic pediatric liver by fluorescence activated cell sorting. Through single-cell RNA sequencing analysis and immunofluorescence, we characterized cholestatic macrophages. We next compared the transcriptional profile of pediatric cholestatic and non-cholestatic macrophage populations to previously published data on normal adult hepatic macrophages. Results We identified 3 distinct macrophage populations across cholestatic liver samples and annotated them as lipid-associated macrophages, monocyte-like macrophages, and adaptive macrophages based on their transcriptional profile. Immunofluorescence of liver tissue using markers for each subset confirmed their presence across BA (n = 6) and ALGS (n = 6) patients. Cholestatic macrophages demonstrated reduced expression of immune regulatory genes as compared to normal hepatic macrophages and were distinct from macrophage populations defined in either healthy adult or pediatric non-cholestatic liver. Conclusions We are the first to perform single-cell RNA sequencing on human pediatric cholestatic liver and identified three macrophage subsets with distinct transcriptional signatures from healthy liver macrophages. Further analyses will identify similarities and differences in these macrophage sub-populations across etiologies of cholestatic liver disease. Taken together, these findings may allow for future development of targeted therapeutic strategies to reprogram macrophages to an immune regulatory phenotype and reduce cholestatic liver injury.
Collapse
Affiliation(s)
- Sarah A. Taylor
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Ann and Robert H Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| | - Shang-Yang Chen
- Division of Rheumatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Gaurav Gadhvi
- Division of Rheumatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Liang Feng
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Ann and Robert H Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
| | - Kyle D. Gromer
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Ann and Robert H Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
| | - Hiam Abdala-Valencia
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Kiwon Nam
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Salina T. Dominguez
- Division of Rheumatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Anna B. Montgomery
- Division of Rheumatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Paul A. Reyfman
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Lorena Ostilla
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Ann and Robert H Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
| | - Joshua B. Wechsler
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Ann and Robert H Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
| | - Carla M. Cuda
- Division of Rheumatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Richard M. Green
- Division of Gastroenterology and Hepatology, Northwestern University, Chicago, Illinois, United States of America
| | - Harris Perlman
- Division of Rheumatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Deborah R. Winter
- Division of Rheumatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| |
Collapse
|
35
|
Cytokines TNFα, IFNγ and IL-2 Are Responsible for Signal Transmission from the Innate Immunity Protein Tag7 (PGLYRP1) to Cytotoxic Effector Lymphocytes. Cells 2020; 9:cells9122602. [PMID: 33291689 PMCID: PMC7761954 DOI: 10.3390/cells9122602] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/26/2020] [Accepted: 12/02/2020] [Indexed: 12/24/2022] Open
Abstract
Studies on the mechanisms of activation of cytotoxic lymphocyte subpopulations are an important research direction in modern immunology. This study provides a detailed analysis of the effect of Tag7 (PGRP-S, PGLYRP1) on the development of lymphocyte subpopulations cytotoxic against MHC-negative tumor cells in a pool of peripheral blood mononuclear cells (PBMCs). The results show that Tag7 can bind to the TREM-1 receptor on the surfaces of monocytes, thereby triggering the expression of mRNA TNFα and IFNγ. The appearance of these cytokines in conditioned medium leads to IL-2 cytokine secretion by CD3+CD4+ lymphocytes. In turn, IL-2 facilitates unspecific activation of three cytotoxic cell subpopulations in the PBMC pool: NK (CD16+CD56+), CD3+CD4+ and CD3+CD8+. These subpopulations appear after a certain period of incubation with Tag7 and show toxicity against tumor cells.
Collapse
|
36
|
Denning NL, Aziz M, Diao L, Prince JM, Wang P. Targeting the eCIRP/TREM-1 interaction with a small molecule inhibitor improves cardiac dysfunction in neonatal sepsis. Mol Med 2020; 26:121. [PMID: 33276725 PMCID: PMC7716442 DOI: 10.1186/s10020-020-00243-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Background Neonatal sepsis and the associated myocardial dysfunction remain a leading cause of infant mortality. Extracellular cold-inducible RNA-binding protein (eCIRP) acts as a ligand of triggering receptor expressed on myeloid cells-1 (TREM-1). M3 is a small CIRP-derived peptide that inhibits the eCIRP/TREM-1 interaction. We hypothesize that the eCIRP/TREM-1 interaction in cardiomyocytes contributes to sepsis-induced cardiac dysfunction in neonatal sepsis, while M3 is cardioprotective. Methods Serum was collected from neonates in the Neonatal Intensive Care Unit (NICU). 5–7-day old C57BL/6 mouse pups were used in this study. Primary murine neonatal cardiomyocytes were stimulated with recombinant murine (rm) CIRP with M3. TREM-1 mRNA and supernatant cytokine levels were assayed. Mitochondrial oxidative stress, ROS, and membrane potential were assayed. Neonatal mice were injected with rmCIRP and speckle-tracking echocardiography was conducted to measure cardiac strain. Sepsis was induced by i.p. cecal slurry. Mouse pups were treated with M3 or vehicle. After 16 h, echocardiography was performed followed by euthanasia for tissue analysis. A 7-day survival study was conducted. Results Serum eCIRP levels were elevated in septic human neonates. rmCIRP stimulation of cardiomyocytes increased TREM-1 gene expression. Stimulation of cardiomyocytes with rmCIRP upregulated TNF-α and IL-6 in the supernatants, while this upregulation was inhibited by M3. Stimulation of cardiomyocytes with rmCIRP resulted in a reduction in mitochondrial membrane potential (MMP) while M3 treatment returned MMP to near baseline. rmCIRP caused mitochondrial calcium overload; this was inhibited by M3. rmCIRP injection impaired longitudinal and radial cardiac strain. Sepsis resulted in cardiac dysfunction with a reduction in cardiac output and left ventricular end diastolic diameter. Both were improved by M3 treatment. Treatment with M3 attenuated serum, cardiac, and pulmonary levels of pro-inflammatory cytokines compared to vehicle-treated septic neonates. M3 dramatically increased sepsis survival. Conclusions Inhibition of eCIRP/TREM-1 interaction with M3 is cardioprotective, decreases inflammation, and improves survival in neonatal sepsis. Trial registration Retrospectively registered.
Collapse
Affiliation(s)
- Naomi-Liza Denning
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA.,Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, Hempstead, NY, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA
| | - Li Diao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Jose M Prince
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA.,Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, Hempstead, NY, USA.,Division of Pediatric Surgery, Cohen Children's Medical Center At Hofstra/Northwell, New Hyde Park, NY, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA. .,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA. .,Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
37
|
An extracellular cold-inducible RNA-binding protein-derived small peptide targeting triggering receptor expressed on myeloid cells-1 attenuates hemorrhagic shock. J Trauma Acute Care Surg 2020; 88:809-815. [PMID: 32453257 DOI: 10.1097/ta.0000000000002664] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Extracellular cold-inducible RNA-binding protein (eCIRP) is a damage-associated molecular pattern, which is released into the circulation after hemorrhagic shock (HS). Recently, we discovered that triggering receptor expressed on myeloid cells-1 (TREM-1) serves as a new receptor of eCIRP to exaggerate inflammation. Here, we hypothesize that by inhibiting the interaction between eCIRP and TREM-1 with the use of a novel short peptide derived from human eCIRP known as M3, we can inhibit the inflammatory response and acute lung injury in HS. METHODS Hemorrhagic shock was induced using C57BL/6 mice by cannulating both femoral arteries. One femoral artery was used for removal of blood while the other was used for continuous monitoring of mean arterial blood pressure. The mean arterial pressure of 25 mm Hg to 30 mm Hg was maintained for 90 minutes, followed by a resuscitation phase of 30 minutes with 1 mL of normal saline. The treatment group was given 10 mg/kg of M3 during the resuscitation phase. Four hours after resuscitation, serum and lungs were collected and analyzed for various injury and inflammatory markers by using colorimetry, real-time polymerase chain reaction, and enzyme-linked immunosorbent assay. RESULTS There was an increase in the serum levels of tissue injury markers (alanine aminotransferase, aspartate aminotransferase, and lactate dehydrogenase) as well as cytokines (TNF-α and IL-6) when comparing the vehicle group versus the sham group. This increase was significantly inhibited in the M3-treated group. The mRNA expression of proinflammatory cytokines TNF-α, IL-6, and IL-1β and the chemokines MIP-2 and KC in lungs was significantly increased in the vehicle-treated HS mice, while their expression was significantly decreased in M3-treated HS mice. Finally, M3 treatment significantly decreased the lung injury score compared with vehicle-treated HS mice. CONCLUSION The novel eCIRP-derived TREM-1 antagonist (M3) can be a potential therapeutic adjunct in the management of hemorrhagic shock.
Collapse
|
38
|
Feng JY, Su WJ, Chuang FY, Pan SW, Yeh YC, Lin YY, Chen NJ. TREM-1 enhances Mycobacterium tuberculosis-induced inflammatory responses in macrophages. Microbes Infect 2020; 23:104765. [PMID: 33049389 DOI: 10.1016/j.micinf.2020.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/09/2020] [Accepted: 10/05/2020] [Indexed: 01/06/2023]
Abstract
Triggering receptor expressed on myeloid cells 1 (TREM-1) extensively interacts with toll-like receptors and amplifies pro-inflammatory responses. The effect of TREM-1 on Mycobacterium tuberculosis (MTB)-related immune responses remains to be elucidated. We isolated bone marrow-derived macrophages (BMDMs) from wild-type mice and Trem-1 KO mice and treated them with MTB whole cell lysate and EsxA (ESAT-6). Cytokine production and mRNA expression, including Trem-1, following stimulation were evaluated. Intratracheal instillation of heat-killed MTB (HKMTB) in mice was performed and the presence of TREM-1-positive macrophages was investigated by immunohistochemistry analysis. In our study, BMDMs isolated from wild-type mice produced more pro-inflammatory cytokines and demonstrated higher inflammatory gene expression levels compared with those isolated from Trem-1 KO mice when stimulated with MTB whole cell lysate. EsxA had a synergistic effect with MTB whole cell lysate on the induction of pro-inflammatory responses. The gene expression of Trem-1 was upregulated when treated with MTB-related proteins. TREM-1-positive macrophages were identified in the lung tissues from patients with active TB and from wild-type mice treated with intratracheal instillation of HKMTB. In conclusion, in mouse macrophages, TREM-1 could enhance pro-inflammatory immune responses when stimulated with MTB-related proteins. The gene expression of Trem-1 could also be induced by MTB-related stimulation.
Collapse
Affiliation(s)
- Jia-Yih Feng
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wei-Juin Su
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Fan-Yi Chuang
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Sheng-Wei Pan
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Chen Yeh
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yung-Yang Lin
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Division of Cerebrovascular Diseases, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Nien-Jung Chen
- Institute of Microbiology and Immunology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
39
|
Cioni B, Zaalberg A, van Beijnum JR, Melis MHM, van Burgsteden J, Muraro MJ, Hooijberg E, Peters D, Hofland I, Lubeck Y, de Jong J, Sanders J, Vivié J, van der Poel HG, de Boer JP, Griffioen AW, Zwart W, Bergman AM. Androgen receptor signalling in macrophages promotes TREM-1-mediated prostate cancer cell line migration and invasion. Nat Commun 2020; 11:4498. [PMID: 32908142 PMCID: PMC7481219 DOI: 10.1038/s41467-020-18313-y] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 06/29/2020] [Indexed: 12/14/2022] Open
Abstract
The androgen receptor (AR) is the master regulator of prostate cancer (PCa) development, and inhibition of AR signalling is the most effective PCa treatment. AR is expressed in PCa cells and also in the PCa-associated stroma, including infiltrating macrophages. Macrophages have a decisive function in PCa initiation and progression, but the role of AR in macrophages remains largely unexplored. Here, we show that AR signalling in the macrophage-like THP-1 cell line supports PCa cell line migration and invasion in culture via increased Triggering Receptor Expressed on Myeloid cells-1 (TREM-1) signalling and expression of its downstream cytokines. Moreover, AR signalling in THP-1 and monocyte-derived macrophages upregulates IL-10 and markers of tissue residency. In conclusion, our data suggest that AR signalling in macrophages may support PCa invasiveness, and blocking this process may constitute one mechanism of anti-androgen therapy. Anti-androgen therapy inhibits prostate cancer (PC) progression, and is thought to act directly on cancer cells. Here the authors show that androgen receptor is expressed on normal and PC-associated macrophages, and its stimulation alters macrophage secretome to promote migration of cultured PC cell lines.
Collapse
Affiliation(s)
- Bianca Cioni
- Divisions of Oncogenomics, The Netherlands Cancer Institute (NKI), Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Anniek Zaalberg
- Divisions of Oncogenomics, The Netherlands Cancer Institute (NKI), Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Judy R van Beijnum
- Angiogenesis laboratory, Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Monique H M Melis
- Molecular Genetics, NKI, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | | | - Mauro J Muraro
- Hubrecht Institute - KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584CT, Utrecht, The Netherlands
| | - Erik Hooijberg
- Division of Pathology, NKI, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Dennis Peters
- Core Facility Molecular Pathology, NKI, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Ingrid Hofland
- Core Facility Molecular Pathology, NKI, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Yoni Lubeck
- Division of Pathology, NKI, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Jeroen de Jong
- Division of Pathology, NKI, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Joyce Sanders
- Division of Pathology, NKI, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Judith Vivié
- Hubrecht Institute - KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584CT, Utrecht, The Netherlands
| | - Henk G van der Poel
- Urology and Medical Oncology, NKI, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Jan Paul de Boer
- Urology and Medical Oncology, NKI, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Arjan W Griffioen
- Angiogenesis laboratory, Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Divisions of Oncogenomics, The Netherlands Cancer Institute (NKI), Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands. .,Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600MB, Eindhoven, The Netherlands. .,, Oncode Institute, The Netherlands.
| | - Andries M Bergman
- Divisions of Oncogenomics, The Netherlands Cancer Institute (NKI), Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands. .,Urology and Medical Oncology, NKI, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands.
| |
Collapse
|
40
|
Gangwar RS, Vinayachandran V, Rengasamy P, Chan R, Park B, Diamond-Zaluski R, Cara EA, Cha A, Das L, Asase C, Maiseyeu A, Deiuliis J, Zhong J, Mitzner W, Biswal S, Rajagopalan S. Differential contribution of bone marrow-derived infiltrating monocytes and resident macrophages to persistent lung inflammation in chronic air pollution exposure. Sci Rep 2020; 10:14348. [PMID: 32873817 PMCID: PMC7462977 DOI: 10.1038/s41598-020-71144-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 07/28/2020] [Indexed: 11/30/2022] Open
Abstract
Chronic exposure to particulate matter < 2.5µ (PM2.5) has been linked to cardiopulmonary disease. Tissue-resident (TR) alveolar macrophages (AΦ) are long-lived, self-renew and critical to the health impact of inhalational insults. There is an inadequate understanding of the impact of PM2.5 exposure on the nature/time course of transcriptional responses, self-renewal of AΦ, and the contribution from bone marrow (BM) to this population. Accordingly, we exposed chimeric (CD45.2/CD45.1) mice to concentrated PM2.5 or filtered air (FA) to evaluate the impact on these end-points. PM2.5 exposure for 4-weeks induced an influx of BM-derived monocytes into the lungs with no contribution to the overall TR-AΦ pool. Chronic (32-weeks) PM2.5 exposure on the other hand while associated with increased recruitment of BM-derived monocytes and their incorporation into the AΦ population, resulted in enhanced apoptosis and decreased proliferation of TR-AΦ. RNA-seq analysis of isolated TR-AΦ and BM-AΦ from 4- and 32-weeks exposed mice revealed a unique time-dependent pattern of differentially expressed genes. PM2.5 exposure resulted in altered histological changes in the lungs, a reduced alveolar fraction which corresponded to protracted lung inflammation. Our findings suggest a time-dependent entrainment of BM-derived monocytes into the AΦ population of PM2.5 exposed mice, that together with enhanced apoptosis of TR-AΦ and reorganization of transcriptional responses, could collectively contribute to the perpetuation of chronic inflammation.
Collapse
Affiliation(s)
- Roopesh Singh Gangwar
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Vinesh Vinayachandran
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Palanivel Rengasamy
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ricky Chan
- Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Bongsoo Park
- Department of Environmental Health and Engineering, Johns Hopkins University School of Public Health, Baltimore, MD, 21205, USA
| | - Rachel Diamond-Zaluski
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Elaine Ann Cara
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Anthony Cha
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Lopa Das
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Courteney Asase
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Andrei Maiseyeu
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jeffrey Deiuliis
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jixin Zhong
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Wayne Mitzner
- Department of Environmental Health and Engineering, Johns Hopkins University School of Public Health, Baltimore, MD, 21205, USA
| | - Shyam Biswal
- Department of Environmental Health and Engineering, Johns Hopkins University School of Public Health, Baltimore, MD, 21205, USA
| | - Sanjay Rajagopalan
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Division of Cardiovascular Medicine, University Hospitals, Harrington Heart and Vascular Institute (HHVI), Cleveland, OH, USA.
- Herman K. Hellerstein MD, Professor of Cardiovascular Research Department of Internal Medicine and Radiology, Case Cardiovascular Research Institute, Case Western Reserve University, 11100 Euclid Ave., Mailstop: 5038, Cleveland, OH, 44106, USA.
| |
Collapse
|
41
|
Herrera-Uribe J, Liu H, Byrne KA, Bond ZF, Loving CL, Tuggle CK. Changes in H3K27ac at Gene Regulatory Regions in Porcine Alveolar Macrophages Following LPS or PolyIC Exposure. Front Genet 2020; 11:817. [PMID: 32973863 PMCID: PMC7468443 DOI: 10.3389/fgene.2020.00817] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 07/08/2020] [Indexed: 12/17/2022] Open
Abstract
Changes in chromatin structure, especially in histone modifications (HMs), linked with chromatin accessibility for transcription machinery, are considered to play significant roles in transcriptional regulation. Alveolar macrophages (AM) are important immune cells for protection against pulmonary pathogens, and must readily respond to bacteria and viruses that enter the airways. Mechanism(s) controlling AM innate response to different pathogen-associated molecular patterns (PAMPs) are not well defined in pigs. By combining RNA sequencing (RNA-seq) with chromatin immunoprecipitation and sequencing (ChIP-seq) for four histone marks (H3K4me3, H3K4me1, H3K27ac and H3K27me3), we established a chromatin state map for AM stimulated with two different PAMPs, lipopolysaccharide (LPS) and Poly(I:C), and investigated the potential effect of identified histone modifications on transcription factor binding motif (TFBM) prediction and RNA abundance changes in these AM. The integrative analysis suggests that the differential gene expression between non-stimulated and stimulated AM is significantly associated with changes in the H3K27ac level at active regulatory regions. Although global changes in chromatin states were minor after stimulation, we detected chromatin state changes for differentially expressed genes involved in the TLR4, TLR3 and RIG-I signaling pathways. We found that regions marked by H3K27ac genome-wide were enriched for TFBMs of TF that are involved in the inflammatory response. We further documented that TF whose expression was induced by these stimuli had TFBMs enriched within H3K27ac-marked regions whose chromatin state changed by these same stimuli. Given that the dramatic transcriptomic changes and minor chromatin state changes occurred in response to both stimuli, we conclude that regulatory elements (i.e. active promoters) that contain transcription factor binding motifs were already active/poised in AM for immediate inflammatory response to PAMPs. In summary, our data provides the first chromatin state map of porcine AM in response to bacterial and viral PAMPs, contributing to the Functional Annotation of Animal Genomes (FAANG) project, and demonstrates the role of HMs, especially H3K27ac, in regulating transcription in AM in response to LPS and Poly(I:C).
Collapse
Affiliation(s)
- Juber Herrera-Uribe
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Haibo Liu
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Kristen A Byrne
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, USDA-Agriculture Research Service, Ames, IA, United States
| | - Zahra F Bond
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, USDA-Agriculture Research Service, Ames, IA, United States
| | - Crystal L Loving
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, USDA-Agriculture Research Service, Ames, IA, United States
| | | |
Collapse
|
42
|
TREM-1 and TREM-2 Expression on Blood Monocytes Could Help Predict Survival in High-Grade Glioma Patients. Mediators Inflamm 2020; 2020:1798147. [PMID: 32684831 PMCID: PMC7350089 DOI: 10.1155/2020/1798147] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/27/2020] [Accepted: 05/25/2020] [Indexed: 12/16/2022] Open
Abstract
Objective In recent years, the role of the modern inflammatory markers TREM-1 (triggering receptors expressed on myeloid cells) and HMGB1 (high mobility group box 1 protein) in tumorigenesis has begun to be studied. Their role in gliomas is not clear. The aim of our study was to find the role of inflammation in gliomas. Patients and Methods. In 63 adult patients with gliomas and 31 healthy controls, the expressions of TREM-1 and TREM-2 on CD14+ blood cells (method: flow cytometry) and the levels of soluble sTREM-1, HMGB1, IL-6, and IL-10 (Elisa tests) were analyzed. Results Cox proportional hazard analysis showed that a TREM-1/TREM-2 ratio was associated with reduced overall survival (HR = 1.001, P = 0.023). Patients with a TREM-1/TREM-2 ratio above 125 survived significantly shorter than patients with a TREM-1/TREM-2 ratio below 125. The percentage of CD14+ TREM-1+ cells was strongly associated with a plasma IL-6/IL-10 ratio (positively) and with IL-10 (negatively). Conversely, we found a higher percentage of CD14+ TREM-2+ monocytes in better surviving patients; these cells could downregulate the exaggerated inflammation and potentiate the phagocytosis in the tumor. The serum levels of HMGB1 negatively correlated with the percentage of CD14+ TREM-1+ cells and with the TREM-1/TREM-2 ratio. The positive correlation between the serum levels of a late proinflammatory cytokine HMGB1 with the percentage of TREM2+ CD14+ monocytes can be explained as an effort for suppression of systemic inflammation by anti-inflammatory acting CD14+ TREM-2+ cells. Conclusion We showed that the TREM-1/TREM-2 ratio (expression on the surface of blood monocytes) could help predict prognosis in patients with gliomas, especially in high-grade gliomas, and that systemic inflammation has an impact on the patient's overall survival. This is the first study that showed that TREM expression on monocytes in peripheral blood could help predict prognosis in patients with gliomas.
Collapse
|
43
|
Ma Q, Honarpisheh M, Li C, Sellmayr M, Lindenmeyer M, Böhland C, Romagnani P, Anders HJ, Steiger S. Soluble Uric Acid Is an Intrinsic Negative Regulator of Monocyte Activation in Monosodium Urate Crystal-Induced Tissue Inflammation. THE JOURNAL OF IMMUNOLOGY 2020; 205:789-800. [PMID: 32561569 DOI: 10.4049/jimmunol.2000319] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022]
Abstract
Although monosodium urate (MSU) crystals are known to trigger inflammation, published data on soluble uric acid (sUA) in this context are discrepant. We hypothesized that diverse sUA preparation methods account for this discrepancy and that an animal model with clinically relevant levels of asymptomatic hyperuricemia and gouty arthritis can ultimately clarify this issue. To test this, we cultured human monocytes with different sUA preparation solutions and found that solubilizing uric acid (UA) by prewarming created erroneous results because of UA microcrystal contaminants triggering IL-1β release. Solubilizing UA with NaOH avoided this artifact, and this microcrystal-free preparation suppressed LPS- or MSU crystal-induced monocyte activation, a process depending on the intracellular uptake of sUA via the urate transporter SLC2A9/GLUT9. CD14+ monocytes isolated from hyperuricemic patients were less responsive to inflammatory stimuli compared with monocytes from healthy individuals. Treatment with plasma from hyperuricemic patients impaired the inflammatory function of CD14+ monocytes, an effect fully reversible by removing sUA from hyperuricemic plasma. Moreover, Alb-creERT2;Glut9 lox/lox mice with hyperuricemia (serum UA of 9-11 mg/dl) showed a suppressed inflammatory response to MSU crystals compared with Glut9 lox/lox controls without hyperuricemia. Taken together, we unravel a technical explanation for discrepancies in the published literature on immune effects of sUA and identify hyperuricemia as an intrinsic suppressor of innate immunity, in which sUA modulates the capacity of monocytes to respond to danger signals. Thus, sUA is not only a substrate for the formation of MSU crystals but also an intrinsic inhibitor of MSU crystal-induced tissue inflammation.
Collapse
Affiliation(s)
- Qiuyue Ma
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig Maximilian University of Munich, 80336 Munich, Bavaria, Germany
| | - Mohsen Honarpisheh
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig Maximilian University of Munich, 80336 Munich, Bavaria, Germany
| | - Chenyu Li
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig Maximilian University of Munich, 80336 Munich, Bavaria, Germany
| | - Markus Sellmayr
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig Maximilian University of Munich, 80336 Munich, Bavaria, Germany
| | - Maja Lindenmeyer
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig Maximilian University of Munich, 80336 Munich, Bavaria, Germany.,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Claudia Böhland
- Department of Radiation Oncology, Hospital of the Ludwig Maximilian University of Munich, 80336 Munich, Germany; and
| | - Paola Romagnani
- Department of Biomedical Experimental and Clinical Sciences "Maria Serio," University of Florence, 50139 Florence, Italy
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig Maximilian University of Munich, 80336 Munich, Bavaria, Germany
| | - Stefanie Steiger
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig Maximilian University of Munich, 80336 Munich, Bavaria, Germany;
| |
Collapse
|
44
|
Murao A, Arif A, Brenner M, Denning NL, Jin H, Takizawa S, Nicastro B, Wang P, Aziz M. Extracellular CIRP and TREM-1 axis promotes ICAM-1-Rho-mediated NETosis in sepsis. FASEB J 2020; 34:9771-9786. [PMID: 32506691 DOI: 10.1096/fj.202000482r] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/09/2020] [Accepted: 05/17/2020] [Indexed: 12/15/2022]
Abstract
Extracellular cold-inducible RNA-binding protein (eCIRP) is a damage-associated molecular pattern (DAMP). Intercellular adhesion molecule-1 (ICAM-1) expressing neutrophils produce excessive amounts of neutrophil extracellular traps (NETs). We reveal that eCIRP generates ICAM-1+ neutrophils through triggering receptor expressed on myeloid cells-1 (TREM-1) and the ICAM-1+ neutrophils involve Rho GTPase to promote NETosis. Treatment of BMDN with rmCIRP increased the frequency of ICAM-1+ BMDN, while rmCIRP-treated TREM-1-/- BMDN or pretreatment of BMDN with TREM-1 inhibitor LP17 significantly decreased the frequency of ICAM-1+ neutrophils. The frequencies of ICAM-1+ neutrophils in blood and lungs were markedly decreased in rmCIRP-injected mice or septic mice treated with LP17. Coculture of ICAM-1-/- neutrophils or wild-type (WT) neutrophils with WT macrophages in the presence of a peptidylarginine deiminase 4 (PAD4) inhibitor reduced TNF-α and IL-6 compared to WT neutrophils treated with rmCIRP. Treatment of ICAM-1-/- neutrophils with rmCIRP resulted in reduced quantities of NETs compared to WT rmCIRP-treated neutrophils. Treatment of BMDN with rmCIRP-induced Rho activation, while blockade of ICAM-1 significantly decreased Rho activation. Inhibition of Rho significantly decreased rmCIRP-induced NET formation in BMDN. TREM-1 plays a critical role in the eCIRP-mediated increase of ICAM-1 expression in neutrophils, leading to the increased NET formation via Rho activation to exaggerate inflammation.
Collapse
Affiliation(s)
- Atsushi Murao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Adnan Arif
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Max Brenner
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA.,Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| | - Naomi-Liza Denning
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA.,Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| | - Hui Jin
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Satoshi Takizawa
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Benjamin Nicastro
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA.,Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA.,Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA
| |
Collapse
|
45
|
Raggi F, Bosco MC. Targeting Mononuclear Phagocyte Receptors in Cancer Immunotherapy: New Perspectives of the Triggering Receptor Expressed on Myeloid Cells (TREM-1). Cancers (Basel) 2020; 12:cancers12051337. [PMID: 32456204 PMCID: PMC7281211 DOI: 10.3390/cancers12051337] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/18/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammatory cells are major players in the onset of cancer. The degree of inflammation and type of inflammatory cells in the tumor microenvironment (TME) are responsible for tilting the balance between tumor progression and regression. Cancer-related inflammation has also been shown to influence the efficacy of conventional therapy. Mononuclear phagocytes (MPs) represent a major component of the inflammatory circuit that promotes tumor progression. Despite their potential to activate immunosurveillance and exert anti-tumor responses, MPs are subverted by the tumor to support its growth, immune evasion, and spread. MP responses in the TME are dictated by a network of stimuli integrated through the cross-talk between activatory and inhibitory receptors. Alterations in receptor expression/signaling can create excessive inflammation and, when chronic, promote tumorigenesis. Research advances have led to the development of new therapeutic strategies aimed at receptor targeting to induce a tumor-infiltrating MP switch from a cancer-supportive toward an anti-tumor phenotype, demonstrating efficacy in different human cancers. This review provides an overview of the role of MP receptors in inflammation-mediated carcinogenesis and discusses the most recent updates regarding their targeting for immunotherapeutic purposes. We focus in particular on the TREM-1 receptor, a major amplifier of MP inflammatory responses, highlighting its relevance in the development and progression of several types of inflammation-associated malignancies and the promises of its inhibition for cancer immunotherapy.
Collapse
|
46
|
Denning NL, Aziz M, Ochani M, Prince JM, Wang P. Inhibition of a triggering receptor expressed on myeloid cells-1 (TREM-1) with an extracellular cold-inducible RNA-binding protein (eCIRP)-derived peptide protects mice from intestinal ischemia-reperfusion injury. Surgery 2020; 168:478-485. [PMID: 32439208 DOI: 10.1016/j.surg.2020.04.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Intestinal ischemia-reperfusion injury results in morbidity and mortality from both local injury and systemic inflammation and acute lung injury. Extracellular cold-inducible RNA-binding protein is a damage associated molecular pattern that fuels systemic inflammation and potentiates acute lung injury. We recently discovered a triggering receptor expressed on myeloid cells-1 serves as a novel receptor for extracellular cold-inducible RNA-binding protein. We developed a 7-aa peptide, named M3, derived from the cold-inducible RNA-binding protein, which interferes with cold-inducible RNA-binding protein's binding to a triggering receptor expressed on myeloid cells-1. Here, we hypothesized that M3 protects mice against intestinal ischemia-reperfusion injury. METHODS Intestinal ischemia was induced in C57BL/6 mice via clamping of the superior mesenteric artery for 60 minutes. At reperfusion, mice were treated intraperitoneally with M3 (10 mg/kg body weight) or normal saline vehicle. Mice were killed 4 hours after reperfusion and blood and lungs were collected for various analysis. A 24-hours survival after intestinal ischemia-reperfusion was assessed. RESULTS Serum levels of organ injury markers aspartate aminotransferase, alanine aminotransferase, lactate dehydrogenase, and lactate were increased with intestinal ischemia-reperfusion, while treatment with M3 significantly decreased their levels. Serum, intestinal, and lung levels of proinflammatory cytokines and chemokines were also increased by intestinal ischemia-reperfusion, and treatment with M3 significantly reduced these values. Intestinal ischemia-reperfusion caused significant histological intestinal and lung injuries, which were mitigated by M3. Treatment with M3 improved the survival from 40% to 80% after intestinal ischemia-reperfusion. CONCLUSION Inhibition of triggering receptor expressed on myeloid cells-1 by an extracellular cold-inducible RNA-binding protein-derived small peptide (M3) decreased inflammation, reduced lung injury, and improved survival in intestinal ischemia-reperfusion injury. Thus, blocking the extracellular cold-inducible RNA-binding protein-triggering receptor expressed on myeloid cells-1 interaction is a promising therapeutic avenue for mitigating intestinal ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Naomi-Liza Denning
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY; Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY; Elmezzi Graduate School of Molecular Medicine, Manhasset, NY
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY; Elmezzi Graduate School of Molecular Medicine, Manhasset, NY
| | - Mahendar Ochani
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY
| | - Jose M Prince
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY; Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY; Cohen Children's Medical Center at Hofstra/Northwell Health, New Hyde Park, NY
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY; Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY; Elmezzi Graduate School of Molecular Medicine, Manhasset, NY.
| |
Collapse
|
47
|
TREM-1 and TREM-2 Expression on CD14 + Cells in Bronchoalveolar Lavage Fluid in Pulmonary Sarcoidosis and Hypersensitivity Pneumonitis in the Context of T Cell Immune Response. Mediators Inflamm 2020; 2020:9501617. [PMID: 32508528 PMCID: PMC7244974 DOI: 10.1155/2020/9501617] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/17/2020] [Accepted: 04/21/2020] [Indexed: 11/17/2022] Open
Abstract
Background Sarcoidosis and hypersensitivity pneumonitis (HP) are immunologically mediated processes caused by hypersensitivity reaction accompanied by similar features including lymphocytic alveolitis and granuloma formation. Recent studies describe the role of TREM receptors in T cell activation, differentiation, and granuloma formation. Alveolar macrophages activation via TREM receptors may be the key factor mediating subsequent immune response. The aim of the study was to analyse TREM-1 and TREM-2 expression to identify further molecular mechanisms participating in the immunopathogenesis of sarcoidosis and HP. Methods Flow cytometry was performed to analyse TREM-1 and TREM-2 expression on CD14+ cells in bronchoalveolar lavage fluid from patients having sarcoidosis or HP and a control group. Results The study proved increased TREM-1 expression on alveolar macrophages in pulmonary sarcoidosis and diminished TREM-1 expression in HP-Sarcoidosis: median: 76.7; HP: median: 29.9; control: median: 53.3, (sarcoidosis versus HP: p < 0.001; sarcoidosis versus control: p < 0.05). TREM-2 expression was increased in both, sarcoidosis and HP-sarcoidosis: median: 34.79; HP: median: 36.00; control: median: 12.98, (sarcoidosis versus control: p < 0.05; HP versus control: p < 0.05). Correlation analysis showed negative correlation between TREM-1 and total number of CD8+ cytotoxic T cells. In sarcoidosis TREM-1 expression decreased with changes of HRCT image, decrease in CD4/CD8 ratio and decrease in DLCO. Conclusions Differences in TREM receptor expression in sarcoidosis (increase in TREM-1 and TREM-2) and HP (increase in TREM-2) and correlation analysis suggests that activation via TREM may participate in typical immunological characteristics of sarcoidosis and HP.
Collapse
|
48
|
de Oliveira Matos A, Dos Santos Dantas PH, Figueira Marques Silva-Sales M, Sales-Campos H. The role of the triggering receptor expressed on myeloid cells-1 (TREM-1) in non-bacterial infections. Crit Rev Microbiol 2020; 46:237-252. [PMID: 32326783 DOI: 10.1080/1040841x.2020.1751060] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The triggering receptor expressed on myeloid cells 1 (TREM-1) is a receptor of the innate immune system, expressed mostly by myeloid cells and primarily associated with pro- inflammatory responses. Although the exact nature of its ligands has not yet been fully elucidated, many microorganisms or danger signals have been proposed as inducers of its activation or the secretion of sTREM-1, the soluble form with putative anti-inflammatory effects. In the course of the 20 years since its first description, several studies have investigated the involvement of TREM-1 in bacterial infections. However, the number of studies describing the role of TREM-1 in fungal, viral and parasite-associated infections has only increased in the last few years, showing a diverse contribution of the receptor in these scenarios, with beneficial or detrimental activities depending on the context. Therefore, this review aims to discuss how TREM-1 may influence viral, fungal and parasitic infection outcomes, highlighting its potential as a therapeutic target and biomarker for diagnosis and prognosis of non-bacterial infectious diseases.
Collapse
|
49
|
Koivisto O, Hanel A, Carlberg C. Key Vitamin D Target Genes with Functions in the Immune System. Nutrients 2020; 12:E1140. [PMID: 32325790 PMCID: PMC7230898 DOI: 10.3390/nu12041140] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/18/2022] Open
Abstract
The biologically active form of vitamin D3, 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3), modulates innate and adaptive immunity via genes regulated by the transcription factor vitamin D receptor (VDR). In order to identify the key vitamin D target genes involved in these processes, transcriptome-wide datasets were compared, which were obtained from a human monocytic cell line (THP-1) and peripheral blood mononuclear cells (PBMCs) treated in vitro by 1,25(OH)2D3, filtered using different approaches, as well as from PBMCs of individuals supplemented with a vitamin D3 bolus. The led to the genes ACVRL1, CAMP, CD14, CD93, CEBPB, FN1, MAPK13, NINJ1, LILRB4, LRRC25, SEMA6B, SRGN, THBD, THEMIS2 and TREM1. Public epigenome- and transcriptome-wide data from THP-1 cells were used to characterize these genes based on the level of their VDR-driven enhancers as well as the level of the dynamics of their mRNA production. Both types of datasets allowed the categorization of the vitamin D target genes into three groups according to their role in (i) acute response to infection, (ii) infection in general and (iii) autoimmunity. In conclusion, 15 genes were identified as major mediators of the action of vitamin D in innate and adaptive immunity and their individual functions are explained based on different gene regulatory scenarios.
Collapse
Affiliation(s)
| | | | - Carsten Carlberg
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland; (O.K.); (A.H.)
| |
Collapse
|
50
|
Denning NL, Aziz M, Murao A, Gurien SD, Ochani M, Prince JM, Wang P. Extracellular CIRP as an endogenous TREM-1 ligand to fuel inflammation in sepsis. JCI Insight 2020; 5:134172. [PMID: 32027618 DOI: 10.1172/jci.insight.134172] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/30/2020] [Indexed: 01/24/2023] Open
Abstract
Extracellular cold-inducible RNA-binding protein (eCIRP) is a recently discovered damage-associated molecular pattern. Understanding the precise mechanism by which it exacerbates inflammation is essential. Here we identified that eCIRP is a new biologically active endogenous ligand of triggering receptor expressed on myeloid cells-1 (TREM-1), fueling inflammation in sepsis. Surface plasmon resonance revealed a strong binding affinity between eCIRP and TREM-1, and fluorescence resonance energy transfer assay confirmed eCIRP's interaction with TREM-1 in macrophages. Targeting TREM-1 by its siRNA or a decoy peptide, LP17, or by using TREM-1-/- mice dramatically reduced eCIRP-induced inflammation. We developed a potentially novel 7-aa peptide derived from human eCIRP, M3, which blocked the interaction of TREM-1 and eCIRP. M3 suppressed inflammation induced by eCIRP or agonist TREM-1 antibody cross-linking in murine macrophages or human peripheral blood monocytes. M3 also inhibited eCIRP-induced systemic inflammation and tissue injury. Treatment with M3 further protected mice from sepsis, improved acute lung injury, and increased survival. Thus, we have discovered a potentially novel TREM-1 ligand and developed a new peptide, M3, to block eCIRP-TREM-1 interaction and improve outcomes in sepsis.
Collapse
Affiliation(s)
- Naomi-Liza Denning
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, New York, USA.,Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, New York, USA
| | - Atsushi Murao
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Steven D Gurien
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Mahendar Ochani
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Jose M Prince
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Ping Wang
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, New York, USA.,Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| |
Collapse
|