1
|
Fan Z, Wang L, Sun S, Ge Z. The properties of TREM1 and its emerging role in pain-related diseases. Mol Brain 2025; 18:15. [PMID: 40011963 PMCID: PMC11866596 DOI: 10.1186/s13041-025-01187-w] [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: 10/06/2024] [Accepted: 02/08/2025] [Indexed: 02/28/2025] Open
Abstract
The TREM1 receptor, a member of the TREMs family, is expressed by myeloid cells and functions as an initiator or enhancer of the inflammatory response, playing a pivotal role in the regulation of inflammation. In recent years, it has been found that TREM1-mediated inflammatory response is involved in the regulation of pain-related diseases. This article provides an extensive review on the structural characteristics and distribution patterns, ligand, signaling pathways, inhibitors, and pathophysiological roles of TREM1 in pain disorders aiming to further elucidate its biological function and offer novel insights for clinical interventions targeting pain-related diseases.
Collapse
Affiliation(s)
- Zhenzhen Fan
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, 730000, China
| | - Longde Wang
- Expert Workstation of Academician Wang Longde, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Songtang Sun
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, 450003, China.
| | - Zhaoming Ge
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, 730000, China.
- Expert Workstation of Academician Wang Longde, Lanzhou University Second Hospital, Lanzhou, 730000, China.
- Gansu Provincial Neurology Clinical Medical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China.
| |
Collapse
|
2
|
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
|
3
|
Bale S, Verma P, Yalavarthi B, Bajželj M, Hasan SA, Silverman JN, Broderick K, Shah KA, Hamill T, Khanna D, Sigalov AB, Bhattacharyya S, Varga J. Inhibiting triggering receptor expressed on myeloid cells 1 signaling to ameliorate skin fibrosis. JCI Insight 2024; 9:e176319. [PMID: 39418109 PMCID: PMC11623937 DOI: 10.1172/jci.insight.176319] [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: 10/25/2023] [Accepted: 10/11/2024] [Indexed: 10/19/2024] Open
Abstract
Systemic sclerosis (SSc) is characterized by immune system failure, vascular insult, autoimmunity, and tissue fibrosis. TGF-β is a crucial mediator of persistent myofibroblast activation and aberrant extracellular matrix production in SSc. The factors responsible for this are unknown. By amplifying pattern recognition receptor signaling, triggering receptor expressed on myeloid cells 1 (TREM-1) is implicated in multiple inflammatory conditions. In this study, we used potentially novel ligand-independent TREM-1 inhibitors in preclinical models of fibrosis and explanted SSc skin fibroblasts in order to investigate the pathogenic role of TREM-1 in SSc. Selective pharmacological TREM-1 blockade prevented and reversed skin fibrosis induced by bleomycin in mice and mitigated constitutive collagen synthesis and myofibroblast features in SSc fibroblasts in vitro. Our results implicate aberrantly activated TREM-1 signaling in SSc pathogenesis, identify a unique approach to TREM-1 blockade, and suggest a potential therapeutic benefit for TREM-1 inhibition.
Collapse
Affiliation(s)
- Swarna Bale
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Priyanka Verma
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Bharath Yalavarthi
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Matija Bajželj
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Syed A.M. Hasan
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jenna N. Silverman
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Katherine Broderick
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kris A. Shah
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Timothy Hamill
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Dinesh Khanna
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Swati Bhattacharyya
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - John Varga
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
Chen X, Yu L, Meng S, Zhao J, Huang X, Wang Z, Zhou Z, Huang Y, Hong T, Duan J, Su T, Cao Z, Chi Y, Huang T, Wang H. Inhibition of TREM-1 ameliorates angiotensin II-induced atrial fibrillation by attenuating macrophage infiltration and inflammation through the PI3K/AKT/FoxO3a signaling pathway. Cell Signal 2024; 124:111458. [PMID: 39384003 DOI: 10.1016/j.cellsig.2024.111458] [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: 07/31/2024] [Revised: 09/29/2024] [Accepted: 10/05/2024] [Indexed: 10/11/2024]
Abstract
Inflammation and infiltration of immune cells are intricately linked to the pathogenesis of atrial fibrillation (AF). Triggering receptor expressed on myeloid cells-1 (TREM-1), an enhancer of inflammation, is implicated in various cardiovascular disorders. However, the precise role and potential mechanisms of TREM-1 in the development of AF remain ambiguous. Atrial samples from patients with AF were used to assess the expression levels of TREM-1. An angiotensin II (Ang II)-induced AF mouse model was established to assess the functionality of TREM-1. Cardiac function and AF inducibility were assessed through echocardiography, programmed transvenous cardiac pacing, and atrial electrophysiological mapping. Peripheral blood and atrial inflammatory cells were assessed using flow cytometry. Using histology, bulk RNA sequencing, biochemical analyses, and cell cultures, the mechanistic role of TREM-1 in AF was elucidated. TREM-1 expression was upregulated and co-localized with macrophages in the atria of patients with AF. Pharmacological inhibition of TREM-1 decreased Ang II-induced atrial enlargement and electrical remodeling. TREM-1 inhibition also ameliorated Ang II-induced NLRP3 inflammasome activation, inflammatory factor release, atrial fibrosis, and macrophage infiltration. Transcriptomic analysis revealed that TREM-1 modulates Ang II-induced inflammation through the PI3K/AKT/FoxO3a signaling pathway. In vitro studies further supported these findings, demonstrating that TREM-1 activation exacerbates Ang II-induced inflammation, while overexpression of FoxO3a counteracts this effect. This study discovered the critical role of TREM-1 in the pathogenesis of AF and its underlying molecular mechanisms. Inhibition of TREM-1 provides a new therapeutic strategy for the treatment of AF.
Collapse
Affiliation(s)
- Xin Chen
- Postgraduate Training Base of General Hospital of Northern Theater Command, Jinzhou Medical University, Jinzhou, Liaoning 121001, PR China; State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Liming Yu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Shan Meng
- Postgraduate Training Base of General Hospital of Northern Theater Command, Jinzhou Medical University, Jinzhou, Liaoning 121001, PR China; State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Jikai Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Xinyi Huang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Zhishang Wang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Zijun Zhou
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Yuting Huang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Tao Hong
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Postgraduate College, Dalian Medical University, Dalian, Liaoning 116000, PR China; Pediatric Surgery Ward, Fuwai Hospital Chinese Academy of Medical Sciences, ShenZhen 518000, PR China
| | - Jinfeng Duan
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Postgraduate College, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Tong Su
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning 110167, PR China
| | - Zijun Cao
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Postgraduate College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110847, PR China
| | - Yanbang Chi
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Department of Obstetrics and Gynecology, General Hospital of Northern Theater Command, Shenyang 110016, PR China
| | - Tao Huang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China.
| | - Huishan Wang
- Postgraduate Training Base of General Hospital of Northern Theater Command, Jinzhou Medical University, Jinzhou, Liaoning 121001, PR China; State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
5
|
Theobald V, Schmitt FCF, Middel CS, Gaissmaier L, Brenner T, Weigand MA. Triggering receptor expressed on myeloid cells-1 in sepsis, and current insights into clinical studies. Crit Care 2024; 28:17. [PMID: 38191420 PMCID: PMC10775509 DOI: 10.1186/s13054-024-04798-2] [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: 11/01/2023] [Accepted: 01/04/2024] [Indexed: 01/10/2024] Open
Abstract
Triggering receptor expressed on myeloid cells-1 (TREM-1) is a pattern recognition receptor and plays a critical role in the immune response. TREM-1 activation leads to the production and release of proinflammatory cytokines, chemokines, as well as its own expression and circulating levels of the cleaved soluble extracellular portion of TREM-1 (sTREM-1). Because patients with sepsis and septic shock show elevated sTREM-1 levels, TREM-1 has attracted attention as an important contributor to the inadequate immune response in this often-deadly condition. Since 2001, when the first blockade of TREM-1 in sepsis was performed, many potential TREM-1 inhibitors have been established in animal models. However, only one of them, nangibotide, has entered clinical trials, which have yielded promising data for future treatment of sepsis, septic shock, and other inflammatory disease such as COVID-19. This review discusses the TREM-1 pathway and important ligands, and highlights the development of novel inhibitors as well as their clinical potential for targeted treatment of various inflammatory conditions.
Collapse
Affiliation(s)
- Vivienne Theobald
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Im Neuenheimer Feld 672, 69120, Heidelberg, Germany
| | - Felix Carl Fabian Schmitt
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Im Neuenheimer Feld 672, 69120, Heidelberg, Germany
| | - Chiara Simone Middel
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Im Neuenheimer Feld 672, 69120, Heidelberg, Germany
| | - Lena Gaissmaier
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Im Neuenheimer Feld 672, 69120, Heidelberg, Germany
| | - Thorsten Brenner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Markus Alexander Weigand
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Im Neuenheimer Feld 672, 69120, Heidelberg, Germany.
| |
Collapse
|
6
|
François B, Lambden S, Fivez T, Gibot S, Derive M, Grouin JM, Salcedo-Magguilli M, Lemarié J, De Schryver N, Jalkanen V, Hicheur T, Garaud JJ, Cuvier V, Ferrer R, Bestle M, Pettilä V, Mira JP, Bouisse C, Mercier E, Vermassen J, Huberlant V, Vinatier I, Anguel N, Levy M, Laterre PF. Prospective evaluation of the efficacy, safety, and optimal biomarker enrichment strategy for nangibotide, a TREM-1 inhibitor, in patients with septic shock (ASTONISH): a double-blind, randomised, controlled, phase 2b trial. THE LANCET. RESPIRATORY MEDICINE 2023; 11:894-904. [PMID: 37269870 DOI: 10.1016/s2213-2600(23)00158-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/19/2023] [Accepted: 04/19/2023] [Indexed: 06/05/2023]
Abstract
BACKGROUND Activation of the triggering receptor expressed on myeloid cells-1 (TREM-1) pathway is associated with septic shock outcomes. Data suggest that modulation of this pathway in patients with activated TREM-1 might improve survival. Soluble TREM-1 (sTREM-1), a potential mechanism-based biomarker, might facilitate enrichment of patient selection in clinical trials of nangibotide, a TREM-1 modulator. In this phase 2b trial, we aimed to confirm the hypothesis that TREM1 inhibition might improve outcomes in patients with septic shock. METHODS This double-blind, randomised, placebo-controlled, phase 2b trial assessed the efficacy and safety of two different doses of nangibotide compared with placebo, and aimed to identify the optimum treatment population, in patients across 42 hospitals with medical, surgical, or mixed intensive care units (ICUs) in seven countries. Non-COVID-19 patients (18-85 years) meeting the standard definition of septic shock, with documented or suspected infection (lung, abdominal, or urinary [in patients ≥65 years]), were eligible within 24 h of vasopressor initiation for the treatment of septic shock. Patients were randomly assigned in a 1:1:1 ratio to intravenous nangibotide 0·3 mg/kg per h (low-dose group), nangibotide 1·0 mg/kg per h (high-dose group), or matched placebo, using a computer-generated block randomisation scheme (block size 3). Patients and investigators were masked to treatment allocation. Patients were grouped according to sTREM-1 concentrations at baseline (established from sepsis observational studies and from phase 2a change to data) into high sTREM-1 (≥ 400 pg/mL). The primary outcome was the mean difference in total Sequential Organ Failure Assessment (SOFA) score from baseline to day 5 in the low-dose and high-dose groups compared with placebo, measured in the predefined high sTREM-1 (≥ 400 pg/mL) population and in the overall modified intention-to-treat population. Secondary endpoints included all-cause 28-day mortality, safety, pharmacokinetics, and evaluation of the relationship between TREM-1 activation and treatment response. This study is registered with EudraCT, 2018-004827-36, and Clinicaltrials.gov, NCT04055909. FINDINGS Between Nov 14, 2019, and April 11, 2022, of 402 patients screened, 355 were included in the main analysis (116 in the placebo group, 118 in the low-dose group, and 121 in the high-dose group). In the preliminary high sTREM-1 population (total 253 [71%] of 355; placebo 75 [65%] of 116; low-dose 90 [76%] of 118; high-dose 88 [73%] of 121), the mean difference in SOFA score from baseline to day 5 was 0·21 (95% CI -1·45 to 1·87, p=0·80) in the low-dose group and 1·39 (-0·28 to 3·06, p=0·104) in the high-dose group versus placebo. In the overall population, the difference in SOFA score from baseline to day 5 between the placebo group and low-dose group was 0·20 (-1·09 to 1·50; p=0·76),and between the placebo group and the high-dose group was 1·06 (-0·23 to 2·35, p=0·108). In the predefined high sTREM-1 cutoff population, 23 (31%) patients in the placebo group, 35 (39%) in the low-dose group, and 25 (28%) in the high-dose group had died by day 28. In the overall population, 29 (25%) patients in the placebo, 38 (32%) in the low-dose, and 30 (25%) in the high-dose group had died by day 28. The number of treatment-emergent adverse events (111 [96%] patients in the placebo group, 113 [96%] in the low-dose group, and 115 [95%] in the high-dose group) and serious treatment-emergent adverse events (28 [24%], 26 [22%], and 31 [26%]) was similar between all three groups. High-dose nangibotide led to a clinically relevant improvement in SOFA score (of two points or more) from baseline to day 5 over placebo in those with higher cutoff concentrations (≥532 pg/mL) of sTREM-1 at baseline. Low dose nangibotide displayed a similar pattern with lower magnitude of effect across all cutoff values. INTERPRETATION This trial did not achieve the primary outcome of improvement in SOFA score at the predefined sTREM-1 value. Future studies are needed to confirm the benefit of nangibotide at higher concentrations of TREM-1 activation. FUNDING Inotrem.
Collapse
Affiliation(s)
- Bruno François
- Medical-Surgical ICU Department and Inserm CIC1435 & UMR1092, CRICS-TRIGGERSEP Network, CHU Limoges, Limoges, France.
| | - Simon Lambden
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK; Inotrem, Paris, France
| | - Tom Fivez
- Ziekenhuis Oost-Limburg, Genk, Belgium
| | - Sebastien Gibot
- Intensive Care Unit, Centre Hospitalier Regional Universitaire, Nancy, France
| | | | - Jean-Marie Grouin
- Statistics Department, Université de Rouen, Mont Saint-Aignan, France
| | | | | | | | - Ville Jalkanen
- Tampere University Hospital, Intensive Care Unit, Tampere, Finland
| | | | | | | | - Ricard Ferrer
- Intensive Care Department, Hospital Universitari Vall d'Hebron, SODIR Research Group, Vall d'Hebron Institut de Recerca, Spain; Paseig de la Vall d'Hebron, Barcelona, Spain
| | - Morten Bestle
- Department of Anaesthesia and Intensive Care, Copenhagen University Hospital-North Zealand, Denmark; Department of Clinical Medicine, University of Copenhagen, Hilleroed, Denmark
| | - Ville Pettilä
- University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jean-Paul Mira
- Groupe Hospitalier Cochin St Vincent de Paul La Roche Guyon, Paris, France
| | - Camille Bouisse
- Centre Hospitalier de Bourg-en-Bresse, Bourg-en-Bresse, France
| | | | | | | | - Isabelle Vinatier
- Centre Hospitalier Départemental de Vendée, La Roche-sur-Yon, France
| | | | - Mitchell Levy
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Warren Alpert School of Medicine at Brown University, Providence, RI, USA
| | | |
Collapse
|
7
|
François B, Lambden S, Garaud JJ, Derive M, Grouin JM, Asfar P, Darreau C, Mira JP, Quenot JP, Lemarié J, Mercier E, Lacherade JC, Vinsonneau C, Fivez T, Helms J, Badie J, Levy M, Cuvier V, Salcedo-Magguilli M, Laszlo-Pouvreau AL, Laterre PF, Gibot S, ESSENTIAL investigators. Evaluation of the efficacy and safety of TREM-1 inhibition with nangibotide in patients with COVID-19 receiving respiratory support: the ESSENTIAL randomised, double-blind trial. EClinicalMedicine 2023; 60:102013. [PMID: 37350989 PMCID: PMC10231876 DOI: 10.1016/j.eclinm.2023.102013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/03/2023] [Accepted: 05/03/2023] [Indexed: 06/05/2023] Open
Abstract
Background Activation of the TREM-1 pathway is associated with outcome in life threatening COVID-19. Data suggest that modulation of this pathway with nangibotide, a TREM-1 modulator may improve survival in TREM-1 activated patients (identified using the biomarker sTREM-1). Methods Phase 2 double-blind randomized controlled trial assessing efficacy, safety, and optimum treatment population of nangibotide (1.0 mg/kg/h) compared to placebo. Patients aged 18-75 years were eligible within 7 days of SARS-CoV-2 documentation and within 48 h of the onset of invasive or non-invasive respiratory support because of COVID-19-related ARDS. Patients were included from September 2020 to April 2022, with a pause in recruitment between January and August 2021. Primary outcome was the improvement in clinical status defined by a seven-point ordinal scale in the overall population with a planned sensitivity analysis in the subgroup of patients with a sTREM-1 level above the median value at baseline (high sTREM-1 group). Secondary endpoints included safety and all-cause 28-day and day 60 mortality. The study was registered in EudraCT (2020-001504-42) and ClinicalTrials.gov (NCT04429334). Findings The study was stopped after 220 patients had been recruited. Of them, 219 were included in the mITT analysis. Nangibotide therapy was associated with an improved clinical status at day 28. Fifty-two (52.0%) of patients had improved in the placebo group compared to 77 (64.7%) of the nangibotide treated population, an odds ratio (95% CI) for improvement of 1.79 (1.02-3.14), p = 0.043. In the high sTREM-1 population, 18 (32.7%) of placebo patients had improved by day 28 compared to 26 (48.1%) of treated patients, an odds ratio (95% CI) of 2.17 (0.96-4.90), p = 0.063 was observed. In the overall population, 28 (28.0%) of placebo treated patients were not alive at the day 28 visit compared to 19 (16.0%) of nangibotide treated patients, an absolute improvement (95% CI) in all-cause mortality at day 28, adjusted for baseline clinical status of 12.1% (1.18-23.05). In the high sTREM-1 population (n = 109), 23 (41.8%) of patients in the placebo group and 12 (22.2%) of patients in the nangibotide group were not alive at day 28, an adjusted absolute reduction in mortality of 19.9% (2.78-36.98). The rate of treatment emergent adverse events was similar in both placebo and nangibotide treated patients. Interpretation Whilst the study was stopped early due to low recruitment rate, the ESSENTIAL study demonstrated that TREM-1 modulation with nangibotide is safe in COVID-19, and results in a consistent pattern of improved clinical status and mortality compared to placebo. The relationship between sTREM-1 and both risk of death and treatment response merits further evaluation of nangibotide using precision medicine approaches in life threatening viral pneumonitis. Funding The study was sponsored by Inotrem SA.
Collapse
Affiliation(s)
- Bruno François
- Medical-Surgical ICU Department and Inserm CIC1435 & UMR1092, CHU Dupuytren, Limoges, France
| | - Simon Lambden
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Inotrem SA, Paris, France
| | | | | | | | - Pierre Asfar
- Department of Intensive Care, CHU d’Angers, France
| | | | - Jean-Paul Mira
- Department of Intensive Care, Groupe Hospitalier Cochin, Paris, France
| | - Jean-Pierre Quenot
- Department of Intensive Care, Burgundy University Hospital, Dijon, France
| | | | - Emmanuelle Mercier
- Department of Intensive Care, CHRU Tours Hôpital Bretonneau, Tours, France
| | - Jean-Claude Lacherade
- Department of Intensive Care, Centre Hospitalier Départemental de Vendée, La Roche-Sur-Yon, France
| | | | - Tom Fivez
- Department of Intensive Care, Ziekenhuis Oost-Limburg, Genk, Belgium
| | - Julie Helms
- Department of Intensive Care, Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, Université de Strasbourg (UNISTRA), Faculté de Médecine and Inserm UMR 1260, RNM, FMTS, Strasbourg, France
| | - Julio Badie
- Department of Intensive Care, Hôpital Nord Franche-Comté, Trevenans, France
| | - Mitchell Levy
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Warren Alpert School of Medicine at Brown University, Providence, RI, USA
| | | | | | | | | | - Sébastien Gibot
- Intensive Care Unit, Centre Hospitalier Regional Universitaire (CHRU), 54000 Nancy, France
| | - ESSENTIAL investigators
- Medical-Surgical ICU Department and Inserm CIC1435 & UMR1092, CHU Dupuytren, Limoges, France
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Inotrem SA, Paris, France
- Université de Rouen, 76821 Mont Saint-Aignan, France
- Department of Intensive Care, CHU d’Angers, France
- Department of Intensive Care, CHU Le Mans, France
- Department of Intensive Care, Groupe Hospitalier Cochin, Paris, France
- Department of Intensive Care, Burgundy University Hospital, Dijon, France
- Department of Intensive Care, Hôtel Dieu, Nantes, France
- Department of Intensive Care, CHRU Tours Hôpital Bretonneau, Tours, France
- Department of Intensive Care, Centre Hospitalier Départemental de Vendée, La Roche-Sur-Yon, France
- Department of Intensive Care, Centre Hospitalier de Béthune, France
- Department of Intensive Care, Ziekenhuis Oost-Limburg, Genk, Belgium
- Department of Intensive Care, Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, Université de Strasbourg (UNISTRA), Faculté de Médecine and Inserm UMR 1260, RNM, FMTS, Strasbourg, France
- Department of Intensive Care, Hôpital Nord Franche-Comté, Trevenans, France
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Warren Alpert School of Medicine at Brown University, Providence, RI, USA
- Department of Critical Care Medicine, CHR Mons-Hainaut, Mons, Belgium
- Intensive Care Unit, Centre Hospitalier Regional Universitaire (CHRU), 54000 Nancy, France
| |
Collapse
|
8
|
Yamaga S, Murao A, Ma G, Brenner M, Aziz M, Wang P. Radiation upregulates macrophage TREM-1 expression to exacerbate injury in mice. Front Immunol 2023; 14:1151250. [PMID: 37168858 PMCID: PMC10164953 DOI: 10.3389/fimmu.2023.1151250] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 04/12/2023] [Indexed: 05/13/2023] Open
Abstract
Introduction Exposure to high-dose ionizing radiation causes tissue injury, infections and even death due to immune dysfunction. The triggering receptor expressed on myeloid cells-1 (TREM-1) has been demonstrated to critically amplify and dysregulate immune responses. However, the role of TREM-1 in radiation injury remains unknown. Extracellular cold-inducible RNA-binding protein (eCIRP), a new damage-associated molecular pattern, is released from activated or stressed cells during inflammation. We hypothesized that ionizing radiation upregulates TREM-1 expression via eCIRP release to worsen survival. Methods RAW264.7 cells and peritoneal macrophages collected from C57BL/6 wild-type (WT) mice were exposed to 5- and 10-Gray (Gy) radiation. C57BL/6 WT and CIRP-/- mice underwent 10-Gy total body irradiation (TBI). TREM-1 expression on RAW264.7 cells and peritoneal macrophages in vitro and in vivo were evaluated by flow cytometry. eCIRP levels in cell culture supernatants and in peritoneal lavage isolated from irradiated mice were evaluated by Western blotting. We also evaluated 30-day survival in C57BL/6 WT, CIRP-/- and TREM-1-/- mice after 6.5-Gy TBI. Results The surface protein and mRNA levels of TREM-1 in RAW264.7 cells were significantly increased at 24 h after 5- and 10-Gy radiation exposure. TREM-1 expression on peritoneal macrophages was significantly increased after radiation exposure in vitro and in vivo. eCIRP levels were significantly increased after radiation exposure in cell culture supernatants of peritoneal macrophages in vitro and in peritoneal lavage in vivo. Moreover, CIRP-/- mice exhibited increased survival after 6.5-Gy TBI compared to WT mice. Interestingly, TREM-1 expression on peritoneal macrophages in CIRP-/- mice was significantly decreased compared to that in WT mice at 24 h after 10-Gy TBI. Furthermore, 30-day survival in TREM-1-/- mice was significantly increased to 64% compared to 20% in WT mice after 6.5-Gy TBI. Conclusion Our data indicate that ionizing radiation increases TREM-1 expression in macrophages via the release of eCIRP, and TREM-1 contributes to worse survival after total body irradiation. Thus, targeting TREM-1 could have the potential to be developed as a novel medical countermeasure for radiation injury.
Collapse
Affiliation(s)
- Satoshi Yamaga
- 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
| | - Gaifeng Ma
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Max Brenner
- 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
| | - 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
|
9
|
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
|
10
|
Zhong Y, Xu Y, Xue S, Zhu L, Lu H, Wang C, Chen H, Sang W, Ma J. Nangibotide attenuates osteoarthritis by inhibiting osteoblast apoptosis and TGF-β activity in subchondral bone. Inflammopharmacology 2022; 30:1107-1117. [PMID: 35391646 DOI: 10.1007/s10787-022-00984-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 03/25/2022] [Indexed: 11/26/2022]
Abstract
Osteoarthritis (OA) is a chronic joint disorder that causes cartilage degradation and subchondral bone abnormalities. Nangibotide, also known as LR12, is a dodecapeptide with considerable anti-inflammatory properties, but its significance in OA is uncertain. The aim of the study was to determine whether nangibotide could attenuate the progression of OA, and elucidate the underlying mechanism. In vitro experiments showed that nangibotide strongly inhibited TNF-α-induced osteogenic reduction, significantly enhanced osteoblast proliferation and prevented apoptosis in MC3T3-E1 cells. Male C57BL/6 J mice aged 2 months were randomly allocated to three groups: sham, ACLT, and ACLT with nangibotide therapy. Nangibotide suppressed ACLT-induced cartilage degradation and MMP-13 expression. MicroCT analysis revealed that nangibotide attenuated in vivo subchondral bone loss induced by ACLT. Histomorphometry results showed that nangibotide attenuated ACLT-induced osteoblast inhibition; TUNEL assays and immunohistochemical staining of cleaved-caspase3 further confirmed the in vivo anti-apoptotic effect of nangibotide on osteoblasts. Furthermore, we found that nangibotide exerted protective effects by suppressing TGF-β signaling mediated by Smad2/3 to restore coupled bone remodeling in the subchondral bone. In conclusion, the findings suggest that nangibotide might exert a protective effect on the bone-cartilage unit and maybe an alternative treatment option for OA.
Collapse
Affiliation(s)
- Yiming Zhong
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Rd, Shanghai, 200080, China
| | - Yiming Xu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Rd, Shanghai, 200080, China
| | - Song Xue
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Libo Zhu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Rd, Shanghai, 200080, China
| | - Haiming Lu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Rd, Shanghai, 200080, China
| | - Cong Wang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Rd, Shanghai, 200080, China
| | - Hongjie Chen
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Rd, Shanghai, 200080, China
| | - Weilin Sang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Rd, Shanghai, 200080, China.
| | - Jinzhong Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100, Haining Rd, Shanghai, 200080, China.
| |
Collapse
|
11
|
Liu T, Chen S, Xie X, Liu H, Wang Y, Qi S, Shi L, Zhou X, Zhang J, Wang S, Wang Y, Chen S, Dou S, Jiang X, Cui R, Jiang H. Soluble TREM-1, as a new ligand for the membrane receptor Robo2, promotes hepatic stellate cells activation and liver fibrosis. J Cell Mol Med 2021; 25:11113-11127. [PMID: 34750987 PMCID: PMC8650037 DOI: 10.1111/jcmm.17033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/22/2021] [Accepted: 10/09/2021] [Indexed: 12/12/2022] Open
Abstract
Triggering receptor expressed on myeloid cells‐1 (TREM‐1) exists in two forms: a transmembrane form and a soluble form (sTREM‐1). The levels of sTREM‐1 are elevated in supernatants of activated HSCs. However, the role of sTREM‐1 in HSC activation and liver fibrosis remains undefined. Previous studies have primarily focused on the transmembrane form of TREM‐1; we innovatively observed the function of sTREM‐1 as a ligand in liver fibrosis and screened its receptor. Here, recombinant sTREM‐1 was used as a stimulator which induced HSC activation and further aggravated liver fibrosis. Then, screening for sTREM‐1 interacting membrane receptors was performed using pull‐down assay followed by mass spectrometry, and the membrane receptor roundabout guidance receptor 2 (Robo2) was identified as a candidate receptor for sTREM‐1. The interaction between sTREM‐1 and Robo2 was verified by pull‐down and immunofluorescence. The role of Robo2 on sTREM‐1‐induced HSC activation and its downstream signal pathways was assessed by knockdown of Robo2 in LX‐2 cells. Furthermore, HSC‐specific knockdown of Robo2 was achieved in a mouse model of liver fibrosis by using a recombinant adeno‐associated virus (AAV) vector to confirm the role of the receptor, and we proved that Robo2 knockdown inhibited the activation of HSC and liver fibrosis, which also led to the inactivation of Smad2/3 and PI3K/Akt pathways in sTREM‐1‐induced HSC activation and liver fibrosis. In conclusion, sTREM‐1 acts as a new ligand of Robo2; the binding of sTREM‐1 to Robo2 initiates the activation of the downstream Smad2/3 and PI3K/Akt signalling pathways, thereby promoting HSC activation and liver fibrosis.
Collapse
Affiliation(s)
- Ting Liu
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, China
| | - Shujia Chen
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, China.,Department of Gastroenterology, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, China
| | - Xiaoli Xie
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, China
| | - Hongqun Liu
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, China
| | - Yongjuan Wang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, China
| | - Shengbin Qi
- Department of General Surgery, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, China
| | - Linping Shi
- Department of Gastroenterology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Xue Zhou
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, China
| | - Jiuna Zhang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, China
| | - Shuling Wang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, China
| | - Yijun Wang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, China
| | - Shengxiong Chen
- Department of Hepatobiliary Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shiying Dou
- Department of infectious diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoyu Jiang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, China
| | - Ruolin Cui
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, China
| | - Huiqing Jiang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Shijiazhuang, Hebei, China
| |
Collapse
|
12
|
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
|
13
|
Abstract
Sepsis is a syndrome which is defined as a dysregulated host response to infection leading to organ failure. Since it remains one of the leading causes of mortality worldwide, numerous drug candidates have already been tested, and continue to be developed, as potential adjunct therapies. Despite convincing mechanisms of action and robust pre-clinical data, almost all drug candidates in the field of sepsis have failed to demonstrate clinical efficacy in the past two decades. Accordingly, the development of new sepsis drugs has markedly decreased in the past few years. Nevertheless, thanks to a better understanding of sepsis pathophysiology and pathways, new promising drug candidates are currently being developed. Instead of a unique sepsis profile as initially suspected, various phenotypes have been characterised. This has resulted in the identification of multiple targets for new drugs together with relevant biomarkers, and a better understanding of the most appropriate time to intervention. Within the entire sepsis drugs portfolio, those targeting the immune response are probably the most promising. Monoclonal antibodies targeting either cytokines or infectious agents are undoubtedly part of the potential successful therapeutic classes to come.
Collapse
Affiliation(s)
- Philippe Vignon
- Medical-Surgical Intensive Care Unit, Dupuytren Teaching Hospital, 87000, Limoges, France. .,Inserm CIC 1435, Dupuytren Teaching Hospital, 87000, Limoges, France. .,Inserm UMR 1092, Dupuytren Teaching Hospital, 87000, Limoges, France. .,Réanimation Polyvalente, CHU Dupuytren, 2 Avenue Martin Luther king, 87042, Limoges, France.
| | - Pierre-François Laterre
- St Luc University Hospital, Université Catholique de Louvain, Avenue Hippocrate 12, 1200, Brussels, Belgium
| | - Thomas Daix
- Medical-Surgical Intensive Care Unit, Dupuytren Teaching Hospital, 87000, Limoges, France.,Inserm CIC 1435, Dupuytren Teaching Hospital, 87000, Limoges, France.,Inserm UMR 1092, Dupuytren Teaching Hospital, 87000, Limoges, France
| | - Bruno François
- Medical-Surgical Intensive Care Unit, Dupuytren Teaching Hospital, 87000, Limoges, France.,Inserm CIC 1435, Dupuytren Teaching Hospital, 87000, Limoges, France.,Inserm UMR 1092, Dupuytren Teaching Hospital, 87000, Limoges, France
| |
Collapse
|
14
|
Francois B, Lambden S, Gibot S, Derive M, Olivier A, Cuvier V, Witte S, Grouin JM, Garaud JJ, Salcedo-Magguilli M, Levy M, Laterre PF. Rationale and protocol for the efficacy, safety and tolerability of nangibotide in patients with septic shock (ASTONISH) phase IIb randomised controlled trial. BMJ Open 2021; 11:e042921. [PMID: 34233965 PMCID: PMC8264912 DOI: 10.1136/bmjopen-2020-042921] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Septic shock is the subgroup of patients with sepsis, which presents as vasopressor dependence, an elevated blood lactate concentration and is associated with a mortality of at least 30%. Expression of the triggering receptor expressed on myeloid cells 1 (TREM-1) pathway, measured using a serum biomarker of pathway activation (soluble TREM-1, sTREM-1) has been associated with outcome in septic shock. Preclinical and early phase patient data suggest that therapeutic modulation of this pathway may improve survival. METHODS AND ANALYSIS Efficacy, Safety and Tolerability of Nangibotide in Patients with Septic Shock is a phase IIb randomised controlled trial that will take place in up to 50 centres in seven countries and recruit 450 patients with septic shock to receive either placebo or one of two doses of nangibotide, a novel regulator of the TREM-1 pathway. The primary outcome will be the impact of nangibotide therapy on the change in Sequential Organ Failure Assessment score from a baseline determined before initiation of study drug therapy. This will be assessed first in the patients with an elevated sTREM-1 level and then in the study population as a whole. In addition to safety, secondary outcomes of the study will include efficacy of nangibotide in relation to sTREM-1 levels in terms of organ function, mortality and long-term morbidity. This study will also facilitate the development of a novel platform for the measurement of sTREM-1 at the point of care. ETHICS AND DISSEMINATION The study has been approved by the responsible ethics committees/institutional review boards in all study countries: Belgium: Universitair Ziekenhuis Antwerpen, France: CPP Ile de France II, Denmark: Region Hovedstaden, Spain: ethics committee from Valld'Hebron Hospital, Barcelona, Finland: Tukija, Ireland: St. James' Hospital (SJH) / Tallaght University Hospital (TUH) Joint Research Ethics Committee, USA: Lifespan, Providence TRIAL REGISTRATION NUMBERS: EudraCT Number: 2018-004827-36 and NCT04055909.
Collapse
Affiliation(s)
- Bruno Francois
- Medical‑Surgical ICU Department and Inserm CIC1435 & UMR1092, CRICS‑TRIGGERSEP Network, University of Limoges, Limoges, France
| | - Simon Lambden
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Sebastien Gibot
- Department of Intensive care medicine, CHRU de Nancy, Nancy, France
| | | | | | | | | | | | | | | | - Mitchell Levy
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brown University School of Medicine, Providence, Rhode Island, USA
| | | |
Collapse
|
15
|
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
|
16
|
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: 7] [Impact Index Per Article: 1.8] [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
|
17
|
Potentiation of NETs release is novel characteristic of TREM-1 activation and the pharmacological inhibition of TREM-1 could prevent from the deleterious consequences of NETs release in sepsis. Cell Mol Immunol 2021; 18:452-460. [PMID: 33420354 PMCID: PMC8026640 DOI: 10.1038/s41423-020-00591-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 11/08/2020] [Indexed: 12/27/2022] Open
Abstract
During sepsis, neutrophil activation induces endothelial cell (EC) dysfunction partly through neutrophil extracellular trap (NET) release. The triggering receptor expressed on myeloid cell-1 (TREM-1) is an orphan immune receptor that amplifies the inflammatory response mediated by Toll-like receptor-4 (TLR4) engagement. Although the key role of TLR4 signaling in NETosis is known, the role of TREM-1 in this process has not yet been investigated. Here, we report that TREM-1 potentiates NET release by human and murine neutrophils and is a component of the NET structure. In contrast, pharmacologic inhibition or genetic ablation of TREM-1 decreased NETosis in vitro and during experimental septic shock in vivo. Moreover, isolated NETs were able to activate ECs and impair vascular reactivity, and these deleterious effects were dampened by TREM-1 inhibition. TREM-1 may, therefore, constitute a new therapeutic target to prevent NETosis and associated endothelial dysfunction.
Collapse
|
18
|
Sigalov AB. SCHOOL of nature: ligand-independent immunomodulatory peptides. Drug Discov Today 2020; 25:1298-1306. [PMID: 32405248 PMCID: PMC7217646 DOI: 10.1016/j.drudis.2020.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/24/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022]
Abstract
Groundbreaking studies in protein biophysics have identified the mechanisms of transmembrane signaling at the level of druggable protein-protein interactions (PPIs). This resulted in the development of the signaling chain homooligomerization (SCHOOL) strategy to modulate cell responses using receptor-specific peptides. Inspired by nature, these short peptides use ligand-independent mechanisms of receptor inhibition and demonstrate potent efficacy in vitro and in vivo. The SCHOOL strategy is especially important when receptor ligands are unknown. An example is the triggering receptor expressed on myeloid cells-1 (TREM-1) receptor, an emerging therapeutic target involved in the pathogenesis of most inflammatory diseases. Here, I discuss advances in the field with a focus on TREM-1 inhibitory SCHOOL peptides that offer new hope for a 'magic bullet' cure for cancer, arthritis, sepsis, retinopathy, and other medical challenges.
Collapse
|
19
|
François B, Wittebole X, Ferrer R, Mira JP, Dugernier T, Gibot S, Derive M, Olivier A, Cuvier V, Witte S, Pickkers P, Vandenhende F, Garaud JJ, Sánchez M, Salcedo-Magguilli M, Laterre PF. Nangibotide in patients with septic shock: a Phase 2a randomized controlled clinical trial. Intensive Care Med 2020; 46:1425-1437. [PMID: 32468087 DOI: 10.1007/s00134-020-06109-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/11/2020] [Indexed: 01/03/2023]
Abstract
PURPOSE Nangibotide is a specific TREM-1 inhibitor that tempered deleterious host-pathogens interactions, restored vascular function, and improved survival, in animal septic shock models. This study evaluated the safety and pharmacokinetics of nangibotide and its effects on clinical and pharmacodynamic parameters in septic shock patients. METHODS This was a multicenter randomized, double-blind, two-stage study. Patients received either continuous infusion of nangibotide (0.3, 1.0, or 3.0 mg/kg/h) or placebo. Treatment began < 24 h after shock onset and continued for up to 5 days. Safety primary outcomes were adverse events (AEs), whether serious or not, and death. Exploratory endpoints evaluated nangibotide effects on pharmacodynamics, organ function, and mortality, and were analyzed according to baseline sTREM-1 concentrations. RESULTS Forty-nine patients were randomized. All treatment emergent AEs (TEAEs) were collected until Day 28. No significant differences were observed in TEAEs between treatment groups. No drug withdrawal linked to TEAE nor appearance of anti-drug antibodies were reported. Nangibotide pharmacokinetics appeared to be dose-proportional and clearance was dose-independent. Nangibotide did not significantly affect pharmacodynamic markers. Decrease in SOFA score LS mean change (± SE) from baseline to Day 5 in pooled nangibotide groups versus placebo was - 0.7 (± 0.85) in the randomized population and - 1.5 (± 1.12) in patients with high baseline plasma sTREM-1 concentrations (non-significant). This pattern was similar to organ support end points. CONCLUSION No significant increases in TEAEs were detected in nangibotide-treated patients versus placebo. These results encourage further evaluation of nangibotide and further exploration of plasma sTREM-1 concentrations as a predictive efficacy biomarker.
Collapse
Affiliation(s)
- Bruno François
- Medical-Surgical ICU Department and Inserm CIC1435 & UMR1092, CRICS-TRIGGERSEP Network, CHU Limoges, Limoges, France.
| | - Xavier Wittebole
- Department of Critical Care Medicine, St Luc University Hospital, Université Catholique de Louvain, Brussels, Belgium
| | - Ricard Ferrer
- ICU Department, Vall d'Hebron University Hospital, Barcelona, Spain
| | | | | | - Sébastien Gibot
- Medical ICU Department, Hospital Central, CHU Nancy, Nancy, France.,Inserm U1116, Nancy Medical Faculty, Lorraine University, Nancy , France
| | | | | | | | | | - Peter Pickkers
- ICU Department, Radboudumc Hospital, Nijmegen, The Netherlands
| | | | | | - Miguel Sánchez
- ICU Department, Hospital Clínico San Carlos, Madrid, Spain
| | | | - Pierre-François Laterre
- Department of Critical Care Medicine, St Luc University Hospital, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
20
|
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
|
21
|
Dantas PHDS, Matos ADO, da Silva Filho E, Silva-Sales M, Sales-Campos H. Triggering receptor expressed on myeloid cells-1 (TREM-1) as a therapeutic target in infectious and noninfectious disease: a critical review. Int Rev Immunol 2020; 39:188-202. [PMID: 32379561 DOI: 10.1080/08830185.2020.1762597] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The triggering receptor expressed on myeloid cells-1 (TREM-1) is an innate immune receptor found in the surface of several immune and non-immune cells. Since its first description in 2000, this molecule and its soluble form (sTREM-1) have been implicated in many diseases with infectious and noninfectious origins. As an amplifier of inflammation, the membrane-associated TREM-1 (mTREM-1) isoform induces the production of pro-inflammatory mediators, thus contributing to the pathogenesis of diseases such as sepsis, arthritis, colitis and infections. In this context, many studies have used molecules capable of inhibiting TREM-1 activity as anti-inflammatory drugs. In this regard, a few peptides have been showing promising results in the amelioration of detrimental immune responses. Some commercially available drugs, including corticosteroids and antibiotics, with known anti-inflammatory effects, have also shown activity in TREM-1 signaling. Therefore, considering the potential of this receptor as a therapeutic target, the present review encompasses the main compounds explored so far in TREM-1 modulation, highlighting and critically discussing its effects and major drawbacks of such approaches.
Collapse
Affiliation(s)
| | - Amanda de Oliveira Matos
- Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiânia, Goiás, Brazil
| | - Ernandes da Silva Filho
- Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiânia, Goiás, Brazil
| | - Marcelle Silva-Sales
- Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiânia, Goiás, Brazil
| | - Helioswilton Sales-Campos
- Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiânia, Goiás, Brazil
| |
Collapse
|
22
|
Sigalov AB. Commentary: Triggering Receptor Expressed on Myeloid Cells-1 Inhibitor Targeted to Endothelium Decreases Cell Activation. Front Immunol 2020; 11:173. [PMID: 32117302 PMCID: PMC7026307 DOI: 10.3389/fimmu.2020.00173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 01/22/2020] [Indexed: 12/20/2022] Open
|
23
|
Di Pillo E, Carrasco K, Brustolin B, Boufenzer A, Jolly L, Derive M, Lacolley P, Regnault V, Gibot S. Inhibition of triggering receptor expressed on myeloid cells-1 impairs thrombin generation. J Thromb Haemost 2020; 18:454-462. [PMID: 31680426 DOI: 10.1111/jth.14677] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 10/30/2019] [Indexed: 01/24/2023]
Abstract
BACKGROUND New evidence has shown the link between inflammation and thrombosis. Triggering receptor expressed on myeloid cells-1 (TREM-1) is an immunoreceptor expressed mostly on neutrophils and monocytes/macrophages. TREM-1 acts as an amplifier of the inflammatory response, and its pharmacological inhibition displays protective effects in various models of inflammatory disorders, in particular by dampening coagulation abnormalities and thrombocytopenia observed during acute inflammation. OBJECTIVES We aimed to decipher the role of TREM-1 in fostering thrombin generation. METHODS We measured thrombin generation (TG) by the use of calibrated automated thrombography with whole blood, and isolated primary human neutrophils and monocytes upon stimulation with lipopolysaccharide (LPS). Tissue factor (TF) expression was measured by flow cytometry and its activity by ELISA. Phosphatidylserine (PtdSer) exposure was determined by flow cytometry. A dodecapeptide (LR12) was used as a specific inhibitor of TREM-1. RESULTS LPS increased TG, TF expression, and activity, as well as the exposure of PtdSer on the surface of monocytes. LR12 dampened TF activity through the decrease of PtdSer exposure, leading to a reduction of thrombin generation. CONCLUSIONS TREM-1 inhibition decreases thrombin generation and could be an interesting target for the development of new inhibitors of leukocyte-associated thrombotic activity.
Collapse
Affiliation(s)
- Elisa Di Pillo
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | | | - Benjamin Brustolin
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | | | | | | | - Patrick Lacolley
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Véronique Regnault
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Sébastien Gibot
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Vandœuvre-lès-Nancy, France
- Service de Médecine Intensive Réanimation, Hôpital Central, Nancy, France
| |
Collapse
|
24
|
|
25
|
Gibot S, Jolly L, Lemarié J, Carrasco K, Derive M, Boufenzer A. Triggering Receptor Expressed on Myeloid Cells-1 Inhibitor Targeted to Endothelium Decreases Cell Activation. Front Immunol 2019; 10:2314. [PMID: 31632399 PMCID: PMC6779727 DOI: 10.3389/fimmu.2019.02314] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/12/2019] [Indexed: 12/21/2022] Open
Abstract
TREM-1 (Triggering Receptor Expressed on Myeloid cells-1) is an immunoreceptor expressed on neutrophils, monocytes/macrophages, and endothelial cells. It amplifies the inflammatory response driven by Toll-Like Receptors (TLR) engagement. The pharmacological inhibition of TREM-1 confers protection in several pre-clinical models of acute inflammation. In this study, we aimed to investigate the role of TREM-1 in endothelial cells using a sneaking ligand construct (SLC) inhibiting TREM-1 in the endothelium. The SLC was made of 3 modules: an E-selectin targeting domain, a Pseudomonas aeruginosa exotoxin a translocation domain, and a 7 aa peptide (LSKSLVF) that contains the interaction site between TREM-1 and its adaptor protein DAP-12. SLC peptide was effectively picked up by endothelial cells following LPS stimulation. It decreased LPS induced TREM-1 up-regulation and cell activation, neutrophils extravasation, and improved median survival time during experimental peritonitis in mice. We reported that a targeted endothelial TREM-1 inhibition is able to dampen cell activation and to confer protection during septic shock in mice. The use of such cell-specific, ligand- independent TREM-1 inhibitors deserve further investigations during acute or chronic inflammatory disorders.
Collapse
Affiliation(s)
- Sébastien Gibot
- INSERM UMRS-1116, Faculté de Médecine Nancy, Université de Lorraine, Nancy, France.,CHRU Nancy, Hôpital Central, Service de Réanimation Médicale, Nancy, France
| | | | - Jérémie Lemarié
- INSERM UMRS-1116, Faculté de Médecine Nancy, Université de Lorraine, Nancy, France.,CHRU Nancy, Hôpital Central, Service de Réanimation Médicale, Nancy, France
| | | | | | | |
Collapse
|
26
|
Jolly L, Carrasco K, Derive M, Lemarié J, Boufenzer A, Gibot S. Targeted endothelial gene deletion of triggering receptor expressed on myeloid cells-1 protects mice during septic shock. Cardiovasc Res 2019; 114:907-918. [PMID: 29361046 DOI: 10.1093/cvr/cvy018] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 01/18/2018] [Indexed: 12/21/2022] Open
Abstract
Aims TREM-1 (Triggering Receptor Expressed on Myeloid cells-1) is an immunoreceptor expressed on neutrophils and monocytes/macrophages whose role is to amplify the inflammatory response driven by Toll-Like Receptors engagement. The pharmacological inhibition of TREM-1 confers protection in several pre-clinical models of acute inflammation. In this study, we aimed to decipher the role of TREM-1 on the endothelium. Methods and results We first showed by qRT-PCR, flow cytometry and confocal microscopy that TREM-1 was expressed in human pulmonary microvascular endothelial cells as well as in mouse vasculature (aorta, mesenteric artery, and pulmonary vessels). TREM-1 expression was upregulated following septic insult. We next observed that TREM-1 engagement impaired mouse vascular reactivity and promoted vascular inflammation. The pharmacological inhibition of TREM-1 (using the synthetic inhibitory peptide LR12) prevented these disorders both in vitro and in vivo. We generated endothelium-conditional Trem-1 ko mice (EndoTREM-1-/-) and submitted them to a caecal ligation and puncture-induced septic shock. As compared with wild-type littermates, targeted endothelial Trem-1 deletion conferred protection during septic shock in modulating inflammatory cells mobilization and activation, in restoring vasoreactivity, and in improving the survival. Conclusion We reported that TREM-1 is expressed and inducible in endothelial cells and plays a direct role in vascular inflammation and dysfunction. The targeted deletion of endothelial Trem-1 conferred protection during septic shock in modulating inflammatory cells mobilization and activation, restoring vasoreactivity, and improving survival. The effect of TREM-1 on vascular tone, while impressive, deserves further investigations including the design of endothelium-specific TREM-1 inhibitors.
Collapse
Affiliation(s)
- Lucie Jolly
- INSERM UMRS-1116, Faculté de Médecine Nancy, Université de Lorraine, Nancy, France.,INOTREM SA, Nancy, France
| | - Kevin Carrasco
- INSERM UMRS-1116, Faculté de Médecine Nancy, Université de Lorraine, Nancy, France
| | | | - Jérémie Lemarié
- INSERM UMRS-1116, Faculté de Médecine Nancy, Université de Lorraine, Nancy, France.,CHRU Nancy, Service de Réanimation Médicale, Hôpital Central, 29 Avenue de Lattre de Tassigny, Nancy Cedex 54035, France
| | | | - Sébastien Gibot
- INSERM UMRS-1116, Faculté de Médecine Nancy, Université de Lorraine, Nancy, France.,CHRU Nancy, Service de Réanimation Médicale, Hôpital Central, 29 Avenue de Lattre de Tassigny, Nancy Cedex 54035, France
| |
Collapse
|
27
|
Gao S, Yi Y, Xia G, Yu C, Ye C, Tu F, Shen L, Wang W, Hua C. The characteristics and pivotal roles of triggering receptor expressed on myeloid cells-1 in autoimmune diseases. Autoimmun Rev 2018; 18:25-35. [PMID: 30408584 DOI: 10.1016/j.autrev.2018.07.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 07/09/2018] [Indexed: 01/13/2023]
Abstract
Triggering receptor expressed on myeloid cells-1 (TREM-1) engagement can directly trigger inflammation or amplify an inflammatory response by synergizing with TLRs or NLRs. Autoimmune diseases are a family of chronic systemic inflammatory disorders. The pivotal role of TREM-1 in inflammation makes it important to explore its immunological effects in autoimmune diseases. In this review, we summarize the structural and functional characteristics of TREM-1. Particularly, we discuss recent findings on TREM-1 pathway regulation in various autoimmune diseases, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), inflammatory bowel disease (IBD), type 1 diabetes (T1D), and psoriasis. This receptor may potentially be manipulated to alter the inflammatory response to chronic inflammation and possible therapies are explored in this review.
Collapse
Affiliation(s)
- Sheng Gao
- Laboratory Animal Center, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Yongdong Yi
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Guojun Xia
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Chengyang Yu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Chenmin Ye
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Fuyang Tu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Leibin Shen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Wenqian Wang
- Department of Breast Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China.
| | - Chunyan Hua
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China.
| |
Collapse
|
28
|
Cuvier V, Lorch U, Witte S, Olivier A, Gibot S, Delor I, Garaud JJ, Derive M, Salcedo-Magguilli M. A first-in-man safety and pharmacokinetics study of nangibotide, a new modulator of innate immune response through TREM-1 receptor inhibition. Br J Clin Pharmacol 2018; 84:2270-2279. [PMID: 29885068 DOI: 10.1111/bcp.13668] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 05/25/2018] [Accepted: 05/29/2018] [Indexed: 12/19/2022] Open
Abstract
AIMS The peptide nangibotide is the first clinical-stage agent targeting the immunoreceptor TREM-1 (triggering receptor expressed on myeloid cells-1) and is being investigated as a novel therapy for acute inflammatory disorders such as septic shock. This first-in-man, randomized, double-blind, ascending dose, placebo-controlled Phase I study evaluated the safety, tolerability and pharmacokinetics of nangibotide. METHODS Twenty-seven healthy subjects (aged 18-45 years) were randomized into eight groups. Nangibotide was administered as a single continuous intravenous infusion. The first two groups received a single i.v. dose of 1 and 10 mg, respectively, over 15 min. Subsequent groups were randomized in a product : placebo ratio of 3:1 at doses ranging from 0.03 to 6 mg kg-1 h-1 over 7 h 45 min, preceded by a 15-minute loading dose of up to 5 mg kg-1 . RESULTS Nangibotide was safe and well tolerated up to the highest dose tested. There were only few adverse events and they were mild in severity and considered unrelated to treatment. Nangibotide displayed dose-proportional PK properties, with a clearance of 6.6 l kg-1 h-1 for a subject of 70 kg and a 3 min effective half-life, which are compatible with extensive enzymatic metabolism in blood. Central and peripheral volumes of distribution were 16.7 l and 15.9 l respectively, indicating limited distribution of the drug mainly in blood and interstitial fluid. No circulating anti-drug antibodies were detectable up to 28 days after administration. CONCLUSIONS The novel immunomodulator nangibotide displayed favourable safety and PK profiles at all doses, including expected pharmacologically active doses, and warrants further clinical development.
Collapse
Affiliation(s)
| | - Ulrike Lorch
- Richmond Pharmacology, St George's University of London, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Carrasco K, Boufenzer A, Jolly L, Le Cordier H, Wang G, Heck AJ, Cerwenka A, Vinolo E, Nazabal A, Kriznik A, Launay P, Gibot S, Derive M. TREM-1 multimerization is essential for its activation on monocytes and neutrophils. Cell Mol Immunol 2018; 16:460-472. [PMID: 29568119 DOI: 10.1038/s41423-018-0003-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 01/01/2018] [Accepted: 01/09/2018] [Indexed: 12/18/2022] Open
Abstract
The triggering receptor expressed on myeloid cells-1 (TREM-1) is a receptor expressed on innate immune cells. By promoting the amplification of inflammatory signals that are initially triggered by Toll-like receptors (TLRs), TREM-1 has been characterized as a major player in the pathophysiology of acute and chronic inflammatory diseases, such as septic shock, myocardial infarction, atherosclerosis, and inflammatory bowel diseases. However, the molecular events leading to the activation of TREM-1 in innate immune cells remain unknown. Here, we show that TREM-1 is activated by multimerization and that the levels of intracellular Ca2+ release, reactive oxygen species, and cytokine production correlate with the degree of TREM-1 aggregation. TREM-1 activation on primary human monocytes by LPS required a two-step process consisting of upregulation followed by clustering of TREM-1 at the cell surface, in contrast to primary human neutrophils, where LPS induced a rapid cell membrane reorganization of TREM-1, which confirmed that TREM-1 is regulated differently in primary human neutrophils and monocytes. In addition, we show that the ectodomain of TREM-1 is able to homooligomerize in a concentration-dependent manner, which suggests that the clustering of TREM-1 on the membrane promotes its oligomerization. We further show that the adapter protein DAP12 stabilizes TREM-1 surface expression and multimerization. TREM-1 multimerization at the cell surface is also mediated by its endogenous ligand, a conclusion supported by the ability of the TREM-1 inhibitor LR12 to limit TREM-1 multimerization. These results provide evidence for ligand-induced, receptor-mediated dimerization of TREM-1. Collectively, our findings uncover the mechanisms necessary for TREM-1 activation in monocytes and neutrophils.
Collapse
Affiliation(s)
- Kevin Carrasco
- INOTREM, Vandœuvre-les-Nancy, France.,UMR-S 1116, Defaillance cardiovasculaire aigue et chronique, Vandœuvre-les-Nancy, France
| | | | - Lucie Jolly
- INOTREM, Vandœuvre-les-Nancy, France.,UMR-S 1116, Defaillance cardiovasculaire aigue et chronique, Vandœuvre-les-Nancy, France
| | - Helene Le Cordier
- UMR7365, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), CNRS-Université de Lorraine, Vandœuvre-les-Nancy, France
| | - Guanbo Wang
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences and Netherlands Proteomics Center, Utrecht University, Utrecht, The Netherlands
| | - Albert Jr Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences and Netherlands Proteomics Center, Utrecht University, Utrecht, The Netherlands
| | - Adelheid Cerwenka
- Innate Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | - Alexandre Kriznik
- Service Commun de Biophysique Interactions Moléculaires (SCBIM), FR3209, Biopôle de l'Université de Lorraine, Vandœuvre-les-Nancy, France
| | | | - Sebastien Gibot
- UMR-S 1116, Defaillance cardiovasculaire aigue et chronique, Vandœuvre-les-Nancy, France
| | | |
Collapse
|
30
|
Shi R, Zhang J, Peng Z, Yuan S, Gao S, Chen L, Yuan Y. Expression level of 12-amino acid triggering receptor on myeloid cells-like transcript 1 derived peptide alleviates lipopolysaccharide-induced acute lung injury in mice. Int J Mol Med 2018; 41:2159-2168. [PMID: 29393375 DOI: 10.3892/ijmm.2018.3443] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 11/23/2017] [Indexed: 11/06/2022] Open
Abstract
Acute lung injury (ALI) is a critical illness with a high morbidity and mortality rate due to severe inflammation in the lungs. The effects and underlying mechanism of the triggering receptor expressed on myeloid cells‑1 (TREM‑1)‑like transcript‑1‑derived peptide (LR12) on ALI remain unclear. The aim of the present study was to determine whether LR12 attenuates lipopolysaccharide (LPS)‑induced ALI and elucidate the mechanism underlying it. Male C57BL/6 mice were randomly assigned to three groups as follows: Sham group, LPS + scramble group and LPS + LR12 group. Normal saline (NS) or LPS was administrated by intratracheal instillation, and NS, LR12 or LR12 scramble was administered intraperitoneally 30 min later. The treatment was repeated every 3 h three times. Mice were sacrificed 24 h later. Pulmonary pathological changes, the lung wet/dry weight ratio, the macrophage and neutrophil counts in bronchoalveolar lavage fluid and myeloperoxidase (MPO) activity in the lung tissues were observed. The inflammatory cytokines were evaluated by enzyme‑linked immunosorbent assay and lung neutrophil infiltration was detected by immunohistochemistry. Nuclear factor (NF)‑κB p65 and TREM‑1 were analyzed by western blotting, and the activation of NF‑κB was detected by electrophoretic mobility shift assay. LPS‑induced pathohistological injury, edema and neutrophil infiltration were significantly alleviated by TREM‑1 inhibitor, LR12. The proinflammatory cytokines [interleukin (IL)‑6, IL‑1β, tumor necrosis factor‑α] and chemokines (keratinocyte chemokine and monocyte chemoattractant protein‑1) were significantly reduced, whereas the anti‑inflammatory cytokines, IL‑10 were significantly increased by LR12. LR12 was identified to significantly decrease p65 expression levels in the nucleus and inhibit the activity of NF‑κB. Furthermore, LR12 alleviated LPS‑induced ALI by reducing the expression of TREM‑1, increasing the release of soluble TREM‑1 and inhibiting activation of the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Ruili Shi
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jiancheng Zhang
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zhang Peng
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Shiying Yuan
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Sumin Gao
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Lin Chen
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yin Yuan
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
31
|
Kökten T, Gibot S, Lepage P, D'Alessio S, Hablot J, Ndiaye NC, Busby-Venner H, Monot C, Garnier B, Moulin D, Jouzeau JY, Hansmannel F, Danese S, Guéant JL, Muller S, Peyrin-Biroulet L. TREM-1 Inhibition Restores Impaired Autophagy Activity and Reduces Colitis in Mice. J Crohns Colitis 2018; 12:230-244. [PMID: 28961797 DOI: 10.1093/ecco-jcc/jjx129] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 09/15/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Triggering receptor expressed on myeloid cells-1 [TREM-1] is known to amplify inflammation in several diseases. Autophagy and endoplasmic reticulum [ER] stress, which activate the unfolded protein response [UPR], are closely linked and defects in these pathways contribute to the pathogenesis of inflammatory bowel disease [IBD]. Both autophagy and UPR are deeply involved in host-microbiota interactions for the clearance of intracellular pathogens, thus contributing to dysbiosis. We investigated whether inhibition of TREM-1 would prevent aberrant inflammation by modulating autophagy and ER stress and preventing dysbiosis. METHODS An experimental mouse model of colitis was established by dextran sulphate sodium treatment. TREM-1 was inhibited, either pharmacologically by LR12 peptide or genetically with Trem-1 knock-out [KO] mice. Colon tissues and faecal pellets of control and colitic mice were used. Levels of macroautophagy, chaperone-mediated autophagy [CMA], and UPR proteins were evaluated by western blotting. The composition of the intestinal microbiota was assessed by MiSeq sequencing in both LR12-treated and KO animals. RESULTS We confirmed that inhibition of TREM-1 attenuates the severity of colitis clinically, endoscopically and histologically. We observed an increase in macroautophagy [ATG1/ULK-1, ATG13, ATG5, ATG16L1, and MAP1LC3-I/II] and in CMA [HSPA8 and HSP90AA1], whereas there was a decrease in the UPR [PERK, IRE-1α, and ATF-6α] protein expression levels in TREM-1 inhibited colitic mice. TREM-1 inhibition prevented dysbiosis. CONCLUSIONS TREM-1 may represent a novel drug target for the treatment of IBD, by modulating autophagy activity and ER stress.
Collapse
Affiliation(s)
- Tunay Kökten
- Faculté de Médecine, Université de Lorraine, Vandœuvre-Lès-Nancy, France
| | - Sébastien Gibot
- Faculté de Médecine, Université de Lorraine, Vandœuvre-Lès-Nancy, France.,Service de Réanimation Médicale, Hôpital Central, Nancy, France
| | - Patricia Lepage
- Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | - Silvia D'Alessio
- Department of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center and Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Julie Hablot
- Ingénierie Moléculaire et Physiopathologie Articulaire [IMoPA], Université de Lorraine, Vandœuvre-Lès-Nancy, France
| | | | - Hélène Busby-Venner
- Département d'Anatomie et Cytologie Pathologiques, Centre Hospitalier Universitaire Nancy-Brabois, Vandœuvre-Lès-Nancy, France
| | - Céline Monot
- Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | - Benjamin Garnier
- Immunopathologie et Chimie Thérapeutique, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - David Moulin
- Ingénierie Moléculaire et Physiopathologie Articulaire [IMoPA], Université de Lorraine, Vandœuvre-Lès-Nancy, France
| | - Jean-Yves Jouzeau
- Ingénierie Moléculaire et Physiopathologie Articulaire [IMoPA], Université de Lorraine, Vandœuvre-Lès-Nancy, France
| | - Franck Hansmannel
- Faculté de Médecine, Université de Lorraine, Vandœuvre-Lès-Nancy, France
| | - Silvio Danese
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Humanitas Clinical and Research Center, Milan, Italy
| | - Jean-Louis Guéant
- Faculté de Médecine, Université de Lorraine, Vandœuvre-Lès-Nancy, France
| | - Sylviane Muller
- Immunopathologie et Chimie Thérapeutique, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Laurent Peyrin-Biroulet
- Faculté de Médecine, Université de Lorraine, Vandœuvre-Lès-Nancy, France.,Département d'Hépatogastroentérologie, Centre Hospitalier Universitaire Nancy-Brabois, Vandœuvre-Lès-Nancy, France
| |
Collapse
|
32
|
Targeting the immune response in atherosclerosis: It's time for clinical trials! Arch Cardiovasc Dis 2017; 110:643-645. [DOI: 10.1016/j.acvd.2017.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 08/28/2017] [Accepted: 08/29/2017] [Indexed: 11/19/2022]
|
33
|
Tammaro A, Derive M, Gibot S, Leemans JC, Florquin S, Dessing MC. TREM-1 and its potential ligands in non-infectious diseases: from biology to clinical perspectives. Pharmacol Ther 2017; 177:81-95. [PMID: 28245991 DOI: 10.1016/j.pharmthera.2017.02.043] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Triggering receptor expressed on myeloid cells-1 (TREM-1) is expressed on the majority of innate immune cells and to a lesser extent on parenchymal cells. Upon activation, TREM-1 can directly amplify an inflammatory response. Although it was initially demonstrated that TREM-1 was predominantly associated with infectious diseases, recent evidences shed new light into its role in sterile inflammatory diseases. Indeed, TREM-1 receptor and its signaling pathways contribute to the pathology of several non-infectious acute and chronic inflammatory diseases, including atherosclerosis, ischemia reperfusion-induced tissue injury, colitis, fibrosis and cancer. This review, aims to give an extensive overview of TREM-1 in non-infectious diseases, with the focus on the therapeutic potential of TREM-1 intervention strategies herein. In addition, we provide the reader with a functional enrichment analysis of TREM-1 signaling pathway and potential TREM-1 ligands in these diseases, obtained via in silico approach. We discuss pre-clinical studies which show that TREM-1 inhibition, via synthetic soluble TREM-1 protein mimickers, is effective in treating (preventing) specific inflammatory disorders, without significant effects on antibacterial response. Further research aimed at identifying specific TREM-1 ligands, in different inflammatory disorders, is required to further unravel the role of this receptor, and explore new avenues to modulate its function.
Collapse
Affiliation(s)
- Alessandra Tammaro
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | - Sebastien Gibot
- Medical Intensive Care Unit, Hôpital Central, CHU Nancy, Nancy, France; Inserm UMR_S1116, Faculté de Médecine, Université de Lorraine, Nancy, France
| | - Jaklien C Leemans
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sandrine Florquin
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Pathology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Mark C Dessing
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
34
|
Aldasoro Arguinano AA, Dadé S, Stathopoulou M, Derive M, Coumba Ndiaye N, Xie T, Masson C, Gibot S, Visvikis-Siest S. TREM-1 SNP rs2234246 regulates TREM-1 protein and mRNA levels and is associated with plasma levels of L-selectin. PLoS One 2017; 12:e0182226. [PMID: 28771614 PMCID: PMC5542552 DOI: 10.1371/journal.pone.0182226] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/15/2017] [Indexed: 01/01/2023] Open
Abstract
High levels of TREM-1 are associated with cardiovascular and inflammatory diseases risks and the most recent studies have showed that TREM-1 deletion or blockade is associated with up to 60% reduction of the development of atherosclerosis. So far, it is unknown whether the levels of TREM-1 protein are genetically regulated. Moreover, TREM family receptors have been suggested to regulate the cellular adhesion process. The goal of this study was to investigate whether polymorphisms within TREM-1 are regulating the variants of serum TREM-1 levels and the expression levels of their mRNA. Furthermore, we aimed to point out associations between polymorphisms on TREM-1 and blood levels of selectins. Among the 10 SNPs studied, the minor allele T of rs2234246, was associated with increased sTREM-1 in the discovery population (p-value = 0.003), explaining 33% of its variance, and with increased levels of mRNA (p-value = 0.007). The same allele was associated with increased soluble L-selectin levels (p-value = 0.011). The higher levels of sTREM-1 and L-selectin were confirmed in the replication population (p-value = 0.0007 and p-value = 0.018 respectively). We demonstrated for the first time one SNP on TREM-1, affecting its expression levels. These novel results, support the hypothesis that TREM-1 affects monocytes extravasation and accumulation processes leading to atherogenesis and atherosclerotic plaque progression, possibly through increased inflammation and subsequent higher expression of sL-selectin.
Collapse
Affiliation(s)
- Alex-Ander Aldasoro Arguinano
- UMR INSERM U1122; IGE-PCV ‘Interactions Gène-Environnement en Physiopathologie Cardiovasculaire’, Faculté de Pharmacie–Université de Lorraine, Nancy, France
| | - Sébastien Dadé
- UMR INSERM U1122; IGE-PCV ‘Interactions Gène-Environnement en Physiopathologie Cardiovasculaire’, Faculté de Pharmacie–Université de Lorraine, Nancy, France
| | - Maria Stathopoulou
- UMR INSERM U1122; IGE-PCV ‘Interactions Gène-Environnement en Physiopathologie Cardiovasculaire’, Faculté de Pharmacie–Université de Lorraine, Nancy, France
| | | | - Ndeye Coumba Ndiaye
- UMR INSERM U1122; IGE-PCV ‘Interactions Gène-Environnement en Physiopathologie Cardiovasculaire’, Faculté de Pharmacie–Université de Lorraine, Nancy, France
| | - Ting Xie
- UMR INSERM U1122; IGE-PCV ‘Interactions Gène-Environnement en Physiopathologie Cardiovasculaire’, Faculté de Pharmacie–Université de Lorraine, Nancy, France
| | - Christine Masson
- UMR INSERM U1122; IGE-PCV ‘Interactions Gène-Environnement en Physiopathologie Cardiovasculaire’, Faculté de Pharmacie–Université de Lorraine, Nancy, France
| | | | - Sophie Visvikis-Siest
- UMR INSERM U1122; IGE-PCV ‘Interactions Gène-Environnement en Physiopathologie Cardiovasculaire’, Faculté de Pharmacie–Université de Lorraine, Nancy, France
- Department of Internal Medicine and Geriatrics, CHU Technopôle Nancy-Brabois, Rue du Morvan, Vandoeuvre-lès-Nancy, France
- * E-mail:
| |
Collapse
|
35
|
Joffre J, Potteaux S, Zeboudj L, Loyer X, Boufenzer A, Laurans L, Esposito B, Vandestienne M, de Jager SCA, Hénique C, Zlatanova I, Taleb S, Bruneval P, Tedgui A, Mallat Z, Gibot S, Ait-Oufella H. Genetic and Pharmacological Inhibition of TREM-1 Limits the Development of Experimental Atherosclerosis. J Am Coll Cardiol 2017; 68:2776-2793. [PMID: 28007141 DOI: 10.1016/j.jacc.2016.10.015] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/12/2016] [Accepted: 10/04/2016] [Indexed: 01/19/2023]
Abstract
BACKGROUND Innate immune responses activated through myeloid cells contribute to the initiation, progression, and complications of atherosclerosis in experimental models. However, the critical upstream pathways that link innate immune activation to foam cell formation are still poorly identified. OBJECTIVES This study sought to investigate the hypothesis that activation of the triggering receptor expressed on myeloid cells (TREM-1) plays a determinant role in macrophage atherogenic responses. METHODS After genetically invalidating Trem-1 in chimeric Ldlr-/-Trem-1-/- mice and double knockout ApoE-/-Trem-1-/- mice, we pharmacologically inhibited Trem-1 using LR12 peptide. RESULTS Ldlr-/- mice reconstituted with bone marrow deficient for Trem-1 (Trem-1-/-) showed a strong reduction of atherosclerotic plaque size in both the aortic sinus and the thoracoabdominal aorta, and were less inflammatory compared to plaques of Trem-1+/+ chimeric mice. Genetic invalidation of Trem-1 led to alteration of monocyte recruitment into atherosclerotic lesions and inhibited toll-like receptor 4 (TLR 4)-initiated proinflammatory macrophage responses. We identified a critical role for Trem-1 in the upregulation of cluster of differentiation 36 (CD36), thereby promoting the formation of inflammatory foam cells. Genetic invalidation of Trem-1 in ApoE-/-/Trem-1-/- mice or pharmacological blockade of Trem-1 in ApoE-/- mice using LR-12 peptide also significantly reduced the development of atherosclerosis throughout the vascular tree, and lessened plaque inflammation. TREM-1 was expressed in human atherosclerotic lesions, mainly in lipid-rich areas with significantly higher levels of expression in atheromatous than in fibrous plaques. CONCLUSIONS We identified TREM-1 as a major upstream proatherogenic receptor. We propose that TREM-1 activation orchestrates monocyte/macrophage proinflammatory responses and foam cell formation through coordinated and combined activation of CD36 and TLR4. Blockade of TREM-1 signaling may constitute an attractive novel and double-hit approach for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jeremie Joffre
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Stephane Potteaux
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Lynda Zeboudj
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Xavier Loyer
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | - Ludivine Laurans
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bruno Esposito
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marie Vandestienne
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Saskia C A de Jager
- Laboratory for Experimental Cardiology, University Medical Center, Utrecht, the Netherlands
| | - Carole Hénique
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Ivana Zlatanova
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Soraya Taleb
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Patrick Bruneval
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Department of Anatomopathology, Hôpital Européen Georges Pompidou, Assistance Publique-Hopitaux de Paris, Paris, France
| | - Alain Tedgui
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Ziad Mallat
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Sebastien Gibot
- INSERM Unité mixte de Recherche-S1116, Faculté de Médecine, Université de Lorraine, Medical Intensive Care Unit, Hôpital Central, Nancy, France
| | - Hafid Ait-Oufella
- INSERM U970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Medical Intensive Care Unit, Hôpital Saint-Antoine, Assistance Publique-Hopitaux de Paris, Université Pierre-et-Marie Curie, Paris, France.
| |
Collapse
|
36
|
LR12-peptide quantitation in whole blood by RP-HPLC and intrinsic fluorescence detection: Validation and pharmacokinetic study. Biomed Chromatogr 2017; 31. [DOI: 10.1002/bmc.3877] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 09/05/2016] [Accepted: 10/24/2016] [Indexed: 11/07/2022]
|
37
|
Liu T, Zhou Y, Li P, Duan JX, Liu YP, Sun GY, Wan L, Dong L, Fang X, Jiang JX, Guan CX. Blocking triggering receptor expressed on myeloid cells-1 attenuates lipopolysaccharide-induced acute lung injury via inhibiting NLRP3 inflammasome activation. Sci Rep 2016; 6:39473. [PMID: 28004759 PMCID: PMC5177963 DOI: 10.1038/srep39473] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 11/23/2016] [Indexed: 12/12/2022] Open
Abstract
Acute lung injury (ALI) is associated with high mortality and uncontrolled inflammation plays a critical role in ALI. TREM-1 is an amplifier of inflammatory response, and is involved in the pathogenesis of many infectious diseases. NLRP3 inflammasome is a member of NLRs family that contributes to ALI. However, the effect of TREM-1 on NLRP3 inflammasome and ALI is still unknown. This study aimed to determine the effect of TREM-1 modulation on LPS-induced ALI and activation of the NLRP3 inflammasome. We showed that LR12, a TREM-1 antagonist peptide, significantly improved survival of mice after lethal doses of LPS. LR12 also attenuated inflammation and lung tissue damage by reducing histopathologic changes, infiltration of the macrophage and neutrophil into the lung, and production of the pro-inflammatory cytokine, and oxidative stress. LR12 decreased expression of the NLRP3, pro-caspase-1 and pro-IL-1β, and inhibited priming of the NLRP3 inflammasome by inhibiting NF-κB. LR12 also reduced the expression of NLRP3 and caspase-1 p10 protein, and secretion of the IL-1β, inhibited activation of the NLRP3 inflammasome by decreasing ROS. For the first time, these data show that TREM-1 aggravates inflammation in ALI by activating NLRP3 inflammasome, and blocking TREM-1 may be a potential therapeutic approach for ALI.
Collapse
Affiliation(s)
- Tian Liu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Ping Li
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Jia-Xi Duan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yong-Ping Liu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Guo-Ying Sun
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Li Wan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Liang Dong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China.,Department of Anesthesiology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 56300, China
| | - Xiang Fang
- Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jian-Xin Jiang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| |
Collapse
|
38
|
Effect of TREM-1 blockade and single nucleotide variants in experimental renal injury and kidney transplantation. Sci Rep 2016; 6:38275. [PMID: 27928159 PMCID: PMC5143803 DOI: 10.1038/srep38275] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 11/07/2016] [Indexed: 12/18/2022] Open
Abstract
Renal ischemia reperfusion (IR)-injury induces activation of innate immune response which sustains renal injury and contributes to the development of delayed graft function (DGF). Triggering receptor expressed on myeloid cells-1 (TREM-1) is a pro-inflammatory evolutionary conserved pattern recognition receptor expressed on a variety of innate immune cells. TREM-1 expression increases following acute and chronic renal injury. However, the function of TREM-1 in renal IR is still unclear. Here, we investigated expression and function of TREM-1 in a murine model of renal IR using different TREM-1 inhibitors: LP17, LR12 and TREM-1 fusion protein. In a human study, we analyzed the association of non-synonymous single nucleotide variants in the TREM1 gene in a cohort comprising 1263 matching donors and recipients with post-transplant outcomes, including DGF. Our findings demonstrated that, following murine IR, renal TREM-1 expression increased due to the influx of Trem1 mRNA expressing cells detected by in situ hybridization. However, TREM-1 interventions by means of LP17, LR12 and TREM-1 fusion protein did not ameliorate IR-induced injury. In the human renal transplant cohort, donor and recipient TREM1 gene variant p.Thr25Ser was not associated with DGF, nor with biopsy-proven rejection or death-censored graft failure. We conclude that TREM-1 does not play a major role during experimental renal IR and after kidney transplantation.
Collapse
|
39
|
Jérémie L, Amir B, Marc D, Sébastien G. The Triggering Receptor Expressed on Myeloid cells-1: A new player during acute myocardial infarction. Pharmacol Res 2015; 100:261-5. [PMID: 26318764 DOI: 10.1016/j.phrs.2015.07.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 07/28/2015] [Indexed: 01/08/2023]
Abstract
Following myocardial ischemia, an intense activation of the immune system occurs that leads to inflammatory cytokines and chemokines production and to the recruitment of neutrophils and mononuclear cells in the infarcted area. Although pro-inflammatory signals initiate the cellular events necessary for scar formation, excessive and prolonged inflammation promotes deleterious cardiac remodeling and dysfunction. The triggering receptor expressed on myeloid cells-1 (TREM-1) is a highly conserved immune-receptor expressed by neutrophils and monocytes that acts as an amplifier of the innate immune response. Blockade of TREM-1 activation protects from hyper-responsiveness and death during severe infections. Here we review the role of TREM-1 in orchestrating the inflammatory response that follows MI. TREM-1 deletion (Trem-1-/-) or modulation by the use of a short inhibitory peptide (LR12) dampens myocardial inflammation, limits leukocyte recruitment, and improves heart function and survival in mice or pigs. Moreover, the soluble form of TREM-1 (sTREM-1) is found in the plasma of patients suffering from an acute MI and its concentration is an independent predictor of death. This suggests that TREM-1 may constitute a new therapeutic target during acute MI.
Collapse
Affiliation(s)
- Lemarié Jérémie
- Service de Réanimation Médicale, Hôpital Central, CHU Nancy, Université de Lorraine, Nancy, France; Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Vandoeuvre-les-Nancy, France
| | | | | | - Gibot Sébastien
- Service de Réanimation Médicale, Hôpital Central, CHU Nancy, Université de Lorraine, Nancy, France; Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Vandoeuvre-les-Nancy, France.
| |
Collapse
|
40
|
Lemarié J, Boufenzer A, Popovic B, Tran N, Groubatch F, Derive M, Labroca P, Barraud D, Gibot S. Pharmacological inhibition of the triggering receptor expressed on myeloid cells-1 limits reperfusion injury in a porcine model of myocardial infarction. ESC Heart Fail 2015; 2:90-99. [PMID: 28834656 PMCID: PMC6410538 DOI: 10.1002/ehf2.12029] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 03/16/2015] [Accepted: 03/17/2015] [Indexed: 01/01/2023] Open
Abstract
Aims Limitation of ischemia/reperfusion injury is a major therapeutic target after acute myocardial infarction (AMI). Toll‐like receptors are implicated in the inflammatory response that occurs during reperfusion. The triggering receptor expressed on myeloid cells (TREM)‐1 acts as an amplifier of the immune response triggered by toll‐like receptor engagement. We hypothesized that administration of a TREM‐1 inhibitory peptide (LR12) could limit reperfusion injury in a porcine model of AMI. Methods and results AMI was induced in 15 adult minipigs by a closed‐chest coronary artery occlusion‐reperfusion technique. Animals were randomized to receive LR12 or vehicle before reperfusion (LR12 n = 7, vehicle n = 8), and were monitored during 18 h. AMI altered hemodynamics and cardiac function, as illustrated by a drop of mean arterial pressure, cardiac index, cardiac power index, ejection fraction, and real‐time pressure–volume loop‐derived parameters. TREM‐1 inhibition by LR12 significantly improved these dysfunctions (P < 0.03) and limited infarct size, as assessed by lower creatine phosphokinase and troponin I concentrations (P < 0.005). Pulmonary, renal, and hepatic impairments occurred after AMI and were attenuated by LR12 administration as assessed by a better PaO2 to FiO2 ratio, a less positive fluid balance, and lower liver enzymes levels (P < 0.05). Conclusion Inhibition of the TREM‐1 pathway by a synthetic peptide limited myocardial reperfusion injury in a clinically relevant porcine model of AMI.
Collapse
Affiliation(s)
- Jérémie Lemarié
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France.,Medical Intensive Care Unit, Hôpital Central, CHU Nancy, Nancy, France
| | - Amir Boufenzer
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France
| | - Batric Popovic
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France.,Department of Cardiology, Hôpital Brabois, CHU Nancy, Nancy, France
| | - Nguyen Tran
- School of Surgery, Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France
| | - Frederique Groubatch
- School of Surgery, Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France
| | | | | | - Damien Barraud
- Medical Intensive Care Unit, Hôpital Central, CHU Nancy, Nancy, France
| | - Sébastien Gibot
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France.,Medical Intensive Care Unit, Hôpital Central, CHU Nancy, Nancy, France
| |
Collapse
|
41
|
Boufenzer A, Lemarié J, Simon T, Derive M, Bouazza Y, Tran N, Maskali F, Groubatch F, Bonnin P, Bastien C, Bruneval P, Marie PY, Cohen R, Danchin N, Silvestre JS, Ait-Oufella H, Gibot S. TREM-1 Mediates Inflammatory Injury and Cardiac Remodeling Following Myocardial Infarction. Circ Res 2015; 116:1772-82. [PMID: 25840803 DOI: 10.1161/circresaha.116.305628] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 04/03/2015] [Indexed: 11/16/2022]
Abstract
RATIONALE Optimal outcome after myocardial infarction (MI) depends on a coordinated healing response in which both debris removal and repair of the myocardial extracellular matrix play a major role. However, adverse remodeling and excessive inflammation can promote heart failure, positioning leucocytes as central protagonists and potential therapeutic targets in tissue repair and wound healing after MI. OBJECTIVE In this study, we examined the role of triggering receptor expressed on myeloid cells-1(TREM-1) in orchestrating the inflammatory response that follows MI. TREM-1, expressed by neutrophils and mature monocytes, is an amplifier of the innate immune response. METHODS AND RESULTS After infarction, TREM-1 expression is upregulated in ischemic myocardium in mice and humans. Trem-1 genetic invalidation or pharmacological inhibition using a synthetic peptide (LR12) dampens myocardial inflammation, limits neutrophils recruitment and monocyte chemoattractant protein-1 production, thus reducing classical monocytes mobilization to the heart. It also improves left ventricular function and survival in mice (n=20-22 per group). During both permanent and transient myocardial ischemia, Trem-1 blockade also ameliorates cardiac function and limits ventricular remodeling as assessed by fluorodeoxyglucose-positron emission tomographic imaging and conductance catheter studies (n=9-18 per group). The soluble form of TREM-1 (sTREM-1), a marker of TREM-1 activation, is detectable in the plasma of patients having an acute MI (n=1015), and its concentration is an independent predictor of death. CONCLUSIONS These data suggest that TREM-1 could constitute a new therapeutic target during acute MI.
Collapse
Affiliation(s)
- Amir Boufenzer
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.)
| | - Jérémie Lemarié
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.)
| | - Tabassome Simon
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.)
| | - Marc Derive
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.)
| | - Youcef Bouazza
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.)
| | - Nguyen Tran
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.)
| | - Fatiha Maskali
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.)
| | - Frédérique Groubatch
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.)
| | - Philippe Bonnin
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.)
| | - Claire Bastien
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.)
| | - Patrick Bruneval
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.)
| | - Pierre-Yves Marie
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.)
| | - Raphael Cohen
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.)
| | - Nicolas Danchin
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.)
| | - Jean-Sébastien Silvestre
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.)
| | - Hafid Ait-Oufella
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.)
| | - Sébastien Gibot
- From the Inserm UMR_S1116 (A.B., J.L., M.D., Y.B., S.G.) and School of Surgery (N. T., F. G.), Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France; Medical Intensive Care Unit, Hôpital Central (J.L., S.G.), Nancyclotep, Hôpital Brabois (F.M., P.-Y.M.), and Department of Pathology, Hôpital Brabois (C.B.), CHU Nancy, Nancy, France; Assistance Publique Hôpitaux de Paris (APHP), Department of Clinical Pharmacology, URC-EST, Hôpital Saint-Antoine, Paris, France (T.S.); UPMC University Paris 06, Paris, France (T.S.); INOTREM SA, Nancy, France (M.D.); Inserm U965, Paris, France (P.B.); Paris Cardiovascular Research Center, Inserm U970, Paris, France (P. B., R.C., J.-S.S., H.A.-O.); APHP, Department of Cardiology, Hôpital Européen Georges Pompidou, Paris, France (N.D.); and Université Paris-Descartes, Paris, France (N.D.).
| |
Collapse
|
42
|
Roe K, Gibot S, Verma S. Triggering receptor expressed on myeloid cells-1 (TREM-1): a new player in antiviral immunity? Front Microbiol 2014; 5:627. [PMID: 25505454 PMCID: PMC4244588 DOI: 10.3389/fmicb.2014.00627] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 11/03/2014] [Indexed: 11/25/2022] Open
Abstract
The triggering receptor expressed on myeloid cells (TREM) family of protein receptors is rapidly emerging as a critical regulator of a diverse array of cellular functions, including amplification of inflammation. Although the ligand(s) for TREM have not yet been fully identified, circumstantial evidence indicates that danger- and pathogen-associated molecular patterns (DAMPs and PAMPs) can induce cytokine production via TREM-1 activation. The discovery of novel functions of TREM, such as regulation of T-cell proliferation and activation of antigen-presenting cells, suggests a larger role of TREM proteins in modulation of host immune responses to microbial pathogens, such as bacteria and fungi. However, the significance of TREM signaling in innate immunity to virus infections and the underlying mechanisms remain largely unclear. The nature and intensity of innate immune responses, specifically production of type I interferon and inflammatory cytokines is a crucial event in dictating recovery vs. adverse outcomes from virus infections. In this review, we highlight the emerging roles of TREM-1, including synergy with classical pathogen recognition receptors. Based on the literature using viral PAMPs and other infectious disease models, we further discuss how TREM-1 may influence host-virus interactions and viral pathogenesis. A deeper conceptual understanding of the mechanisms associated with pathogenic and/or protective functions of TREM-1 in antiviral immunity is essential to develop novel therapeutic strategies for the control of virus infection by modulating innate immune signaling.
Collapse
Affiliation(s)
- Kelsey Roe
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa Honolulu, HI, USA
| | - Sébastien Gibot
- Service de Réanimation Médicale, University Hospital of Nancy Nancy, France
| | - Saguna Verma
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa Honolulu, HI, USA
| |
Collapse
|
43
|
Pelham CJ, Pandya AN, Agrawal DK. Triggering receptor expressed on myeloid cells receptor family modulators: a patent review. Expert Opin Ther Pat 2014; 24:1383-95. [PMID: 25363248 DOI: 10.1517/13543776.2014.977865] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Triggering receptor expressed on myeloid cells (TREM) receptors and TREM-like transcript (TLT; or TREML) receptors of the immunoglobulin superfamily are known as key modulators of host immune responses. TREM-1 (CD354) and TREM-2 share the transmembrane adaptor DNAX-activation protein of 12 kDa (DAP12), but they possess separate stimulatory and inhibitory functional roles, especially in myeloid cells. AREAS COVERED This review covers findings related to TREMs and TLTs published in patent applications from their discovery in 2000 to the present. New roles for TREM-1, TREM-2, TLT-1 and TLT-2 in maladies ranging from acute and chronic inflammatory disorders to cardiovascular diseases and cancers are discussed. Putative endogenous ligands and novel synthetic peptide blockers are also discussed. EXPERT OPINION So far, therapeutic use of activators/blockers specific for TREMs and TLTs has been limited to preclinical animal models. TREM-1 is an immediate therapeutic target for acute and chronic inflammatory conditions, especially sepsis. Certain mutations in DAP12 and TREM-2 manifest into a disorder named polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy, and newly identified TREM-2 variants confer a significant increase in risk of developing Alzheimer's disease. This makes TREM-2 an attractive therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Christopher J Pelham
- Creighton University School of Medicine, Department of Biomedical Sciences and Center for Clinical & Translational Science , Omaha, NE 68178 , USA
| | | | | |
Collapse
|
44
|
Ferrer-Acosta Y, González M, Fernández M, Valance WA. Emerging Roles for Platelets in Inflammation and Disease. ACTA ACUST UNITED AC 2014; 2. [PMID: 28758142 PMCID: PMC5531291 DOI: 10.4172/2332-0877.1000149] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Platelets and their interaction with cells of the immune system contribute through a variety of molecular mechanisms to support hemostasis and inflammation. These simple yet essential cells exert their effects in lymphocytes, monocytes, and neutrophils, both recruiting and modulating their function after activation. Emerging evidence is starting to define the mechanisms that allow platelets to also play pivotal roles in host defense. For example, platelet cell-surface expression of toll-like receptors allows platelets to direct neutrophil activation toward extracellular trap formation and facilitate the elimination of blood pathogens. In addition to these well-known receptors, two of the most recently discovered platelet receptors, C-type lectin receptor 2 (CLEC-2), and TREM-like transcript-1 (TLT-1), have been shown to modulate hemostatic and inflammation-related roles in platelets. This review will discuss the evolution of our understanding of platelet functions from hemostasis to inflammation, and highlight novel mechanisms that platelets use to mediate hemostasis under inflammatory pressure.
Collapse
Affiliation(s)
| | | | - Mónica Fernández
- University of Puerto Rico, Mayagüez Campus, Mayagüez, Puerto Rico, USA
| | - Washington A Valance
- University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico, USA.,Universidad Central del Caribe, Bayamón, Puerto Rico, USA
| |
Collapse
|
45
|
Attenuation of Responses to Endotoxin by the Triggering Receptor Expressed on Myeloid Cells-1 Inhibitor LR12 in Nonhuman Primate. Anesthesiology 2014; 120:935-42. [DOI: 10.1097/aln.0000000000000078] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Abstract
Background:
The triggering receptor expressed on myeloid cells-1 is an immunoreceptor that amplifies the inflammatory response mediated by toll-like receptors engagement. Triggering receptor expressed on myeloid cells-1 inhibitory peptides such LR12 have been shown to prevent hyperresponsiveness and death in several experimental models of septic shock.
Methods:
Twelve adult male Cynomolgus (Macaca fascicularis) monkeys exposed to an intravenous bolus of endotoxin (10 μg/kg) were randomized to receive LR12 or placebo (n = 6 per group) as an initial intravenous bolus followed by an 8-h continuous intravenous infusion. An additional group of four only received vehicle infusion. Vital signs were monitored for 8 h. Blood was sampled at H0, 1, 2, 4, and 8 for analysis of clinical chemistries, leukocyte count, coagulation parameters, and cytokine plasma concentration.
Results:
LR12 showed no effect on heart rate and body temperature. By contrast to the placebo group, which experienced a 25 to 40% blood pressure decrease after endotoxin administration, LR12-treated monkeys remained normotensive. Endotoxin induced leukopenia at 2 h (mean leukocyte count, 7.62 g/l vs. 21.1 at H0), which was attenuated by LR12. LR12 also attenuated cytokine production.
Conclusions:
The triggering receptor expressed on myeloid cells-1 inhibitor LR12 is able to mitigate endotoxin-associated clinical and biological alterations, with no obvious side effects. This study paves the way for future phases Ia and Ib trials in humans.
Collapse
|
46
|
Pelham CJ, Agrawal DK. Emerging roles for triggering receptor expressed on myeloid cells receptor family signaling in inflammatory diseases. Expert Rev Clin Immunol 2013; 10:243-56. [PMID: 24325404 DOI: 10.1586/1744666x.2014.866519] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Innate immune receptors represent important therapeutic targets for inflammatory disorders. In particular, the Toll-like receptor (TLR) family has emerged as a promoter of chronic inflammation that contributes to obesity, insulin resistance and atherosclerosis. Importantly, triggering receptor expressed on myeloid cells-1 (TREM-1) has been characterized as an 'amplifier' of TLR2 and TLR4 signaling. TREM-1- and TREM-2-dependent signaling, as opposed to TREM-like transcript-1 (TLT-1 or TREML1), are mediated through association with the transmembrane adaptor DNAX activation protein of 12 kDa (DAP12). Recessive inheritance of rare mutations in DAP12 or TREM-2 results in a disorder called polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy, and surprisingly these subjects are not immunocompromised. Recent progress into the roles of TREM/DAP12 signaling is critically reviewed here with a focus on metabolic, cardiovascular and inflammatory diseases. The expanding repertoire of putative ligands for TREM receptors is also discussed.
Collapse
Affiliation(s)
- Christopher J Pelham
- Department of Biomedical Sciences and Center for Clinical & Translational Science, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE 68178, USA
| | | |
Collapse
|
47
|
|