1
|
Ueland T, Nkele I, Hoel H, Lockman S, Michelsen AE, Moshomo T, Aukrust P, Mohammed T, Trøseid M, Mosepele M. Markers of extracellular matrix degradation and inflammasome activation are associated with carotid plaques in virally suppressed people with HIV in Botswana. AIDS 2025; 39:22-30. [PMID: 39283741 DOI: 10.1097/qad.0000000000004014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/06/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND HIV is associated with increased risk of cardiovascular disease. We investigated soluble markers of extracellular matrix (ECM) remodeling and inflammation in relation to presence of carotid plaques in a well characterized adult cross-sectional study of people with HIV (PWH) and matched people without HIV in Botswana. METHODS Using enzyme immunoassays we analyzed plasma ECM remodeling mediators including galectin-3 (GAL-3), cystatin B (CysB), and growth/differentiation factor 15 (GDF-15) and the inflammatory marker interleukin (IL)-18 in 196 without HIV and 197 PWH of which 36 were ART-naïve. RESULTS We found (i) PWH had higher plasma levels of the ECM markers GAL-3 and CysB and the NLRP3 inflammasome activation marker IL-18, mainly in ART naïve participants, (ii) PWH on ART had markedly higher GDF-15, associated with use of first generation nucleoside analogs; iii) high levels of CysB and IL-18 correlated with presence of carotid plaques. CONCLUSION In PWH, high levels of CysB and IL-18 were associated with the presence of carotid plaques. For IL-18, this was observed in the study population as a whole, whereas the association for CysB was restricted to PWH.
Collapse
Affiliation(s)
- Thor Ueland
- Research Institute for Internal Medicine
- Institute of Clinical Medicine, University of Oslo
- Thrombosis Research Center (TREC), Division of internal medicine, University hospital of North Norway, Tromsø Norway
| | - Isaac Nkele
- Department of Medicine, Faculty of Medicine, University of Botswana
- Botswana Harvard Health Partnership, Gaborone, Botswana
| | - Hedda Hoel
- Research Institute for Internal Medicine
- Lovisenberg Diakonal Hospital, Nydalen, Oslo, Norway
| | - Shahin Lockman
- Botswana Harvard Health Partnership, Gaborone, Botswana
- Department of Immunology & Infectious Diseases Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Annika E Michelsen
- Research Institute for Internal Medicine
- Institute of Clinical Medicine, University of Oslo
| | - Thato Moshomo
- Department of Medicine, Faculty of Medicine, University of Botswana
| | - Pål Aukrust
- Research Institute for Internal Medicine
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet
- Institute of Clinical Medicine, University of Oslo
| | | | - Marius Trøseid
- Research Institute for Internal Medicine
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet
- Institute of Clinical Medicine, University of Oslo
| | - Mosepele Mosepele
- Department of Medicine, Faculty of Medicine, University of Botswana
- Botswana Harvard Health Partnership, Gaborone, Botswana
- Department of Immunology & Infectious Diseases Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
2
|
Vanpouille C, Wells A, DeGruttola V, Lynch M, Zhang X, Fitzgerald W, Tu X, Chaillon A, Landay A, Weber K, Scully E, Karn J, Gianella S. Cytokine trajectory over time in men and women with HIV on long-term antiretroviral therapy. AIDS 2025; 39:1-10. [PMID: 39639719 PMCID: PMC11631044 DOI: 10.1097/qad.0000000000004033] [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: 06/28/2024] [Accepted: 10/01/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE Although antiretroviral therapy (ART) suppresses viral replication and reduces inflammation, it does not lead to the normalization of cytokines. The long-term effects of ART beyond viral suppression have not been studied and are mostly limited to cross-sectional research. DESIGN The impact of long-term ART on the trajectory of 40 cytokines/chemokines in 31 men and 59 women who maintained viral suppression over a median period of 6 years (317 visits ranging from 24 to 384 weeks post ART initiation) were measured by Luminex. METHODS We used a generalized additive model with a Gaussian distribution and identity link function to model concentrations over time and investigate sex and race differences. RESULTS While most cytokine/chemokine trajectories remained stable, the trajectory of nine markers of monocyte/macrophage activation (IP-10, I-TAC, MIG, sCD163, sCD14, MCP-1, MIP-3β, CXCL13, TNF-α) decreased over time (adj. P < 0.05). Despite continuous viral suppression, M-CSF, IL-15, and LBP increased over time (adj. P < 0.05). sCD14 was the only cytokine whose trajectory differed by sex (adj. P = 0.033). Overall, women had lower mean levels of IL-18 but higher levels of sCD14 than did men (adj. P < 0.05). GROα, LBP, and sCD14 showed significant differences between races (adj. P < 0.05). No association between cytokines and cellular HIV DNA/RNA was found. CONCLUSION Our study reveals a continuous decline in markers of monocyte/macrophage activation over 6 years of suppressive ART, indicating that long-term treatment may mitigate inflammaging and cardiovascular-related outcomes. The higher levels of sCD14 observed in women are consistent with them having greater innate immune activation than men do.
Collapse
Affiliation(s)
- Christophe Vanpouille
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Alan Wells
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Victor DeGruttola
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Miranda Lynch
- Department of Biostatistics and Bioinformatics, Hauptman-Woodward Medical Research Institute, Buffalo, NY, USA
| | - Xinlian Zhang
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Wendy Fitzgerald
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Xin Tu
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Antoine Chaillon
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Alan Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Kathleen Weber
- Hektoen Institute of Medicine/Cook County Health, Chicago, IL, USA
| | - Eileen Scully
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jonathan Karn
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH, USA
| | - Sara Gianella
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
3
|
Naushad W, Premadasa LS, Okeoma BC, Mohan M, Okeoma CM. Extracellular condensates (ECs) are endogenous modulators of HIV transcription and latency reactivation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.14.613037. [PMID: 39345617 PMCID: PMC11429871 DOI: 10.1101/2024.09.14.613037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Persistence of human immunodeficiency virus (HIV) latent reservoir is the major challenge to HIV cure because the latent reservoir is not eliminated by antiretroviral therapy (ART), and they serve as sources for viral rebound upon cessation of ART. Mechanisms regulating viral persistence are not well understood. This study used model systems of post-integration latency to explore the role of basal ganglia (BG) isolated extracellular condensates (ECs) in reprogramming HIV latent cells. We found that BG ECs from uninfected macaques (VEH) and SIV infected macaques (VEH|SIV) activate latent HIV transcription in various model systems. VEH and VEH|SIV ECs significantly increased expression of viral antigen in latently infected cells. Activation of viral transcription, antigen expression, and latency reactivation was inhibited by ECs from the brain of macaques treated with Delta-9-tetrahydrocannabinol (THC) and infected with SIV (THC|SIV). Virus produced by latently infected cells treated with VEH|SIV ECs potentiated cell-cell and cell-free HIV transmission. VEH|SIV ECs also reversed dexamethasone-mediated inhibition of HIV transcription while TNFα-mediated reactivation of latency was reversed by THC|SIV ECs. Transcriptome and secretome analyses of total RNA and supernatants from latently infected cells treated with ECs revealed significant alteration in gene expression and cytokine secretion. THC|SIV ECs increased secretion of Th2 and decreased secretion of proinflammatory cytokines. Most strikingly, while VEH/SIV ECs robustly induced HIV RNA in latently HIV-infected cells, long-term low-dose THC administration enriched ECs for anti-inflammatory cargo that significantly diminished their ability to reactivate latent HIV, an indication that ECs are endogenous host factors that may regulate HIV persistence. Highlights ECs isolated from SIV infected macaques (VEH|SIV ECs) is a positive regulator of LTR-dependent HIV transcription and production of infectious viral particles in vitro.ECs isolated from THC treated SIV infected macaques (THC|SIV ECs) prevents the transcription and reactivation of HIV in latently infected cells and prevents production of viral particles in vitro.ECs reprogram host transcriptome and secretome in manners that or suppress promote reactivation of latent HIV reservoir.The above highlights led to the conclusion that while VEH/SIV ECs robustly induced HIV RNA in latently HIV-infected cells, long-term low-dose THC administration enriched ECs for anti-inflammatory cargo that significantly diminished their ability to reactivate latent HIV.
Collapse
|
4
|
De Clercq J, De Scheerder MA, Mortier V, Verhofstede C, Vandecasteele SJ, Allard SD, Necsoi C, De Wit S, Gerlo S, Vandekerckhove L. Longitudinal patterns of inflammatory mediators after acute HIV infection correlate to intact and total reservoir. Front Immunol 2024; 14:1337316. [PMID: 38250083 PMCID: PMC10796502 DOI: 10.3389/fimmu.2023.1337316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 12/11/2023] [Indexed: 01/23/2024] Open
Abstract
Background Despite the beneficial effects of antiretroviral therapy (ART) initiation during acute HIV infection (AHI), residual immune activation remains a hallmark of treated HIV infection. Methods Plasma concentrations of 40 mediators were measured longitudinally in 39 early treated participants of a Belgian AHI cohort (HIV+) and in 21 HIV-negative controls (HIV-). We investigated the association of the inflammatory profile with clinical presentation, plasma viral load, immunological parameters, and in-depth characterization of the HIV reservoir. Results While levels of most soluble mediators normalized with suppressive ART, we demonstrated the persistence of a pro-inflammatory signature in early treated HIV+ participants in comparison to HIV- controls. Examination of these mediators demonstrated a correlation with their levels during AHI, which seemed to be viremia-driven, and suggested involvement of an activated myeloid compartment, IFN-γ-signaling, and inflammasome-related pathways. Interestingly, some of these pro-inflammatory mediators correlated with a larger reservoir size and slower reservoir decay. In contrast, we also identified soluble mediators which were associated with favorable effects on immunovirological outcomes and reservoir, both during and after AHI. Conclusion These data highlight how the persistent pro-inflammatory profile observed in early ART treated individuals is shaped during AHI and is intertwined with viral dynamics.
Collapse
Affiliation(s)
- Jozefien De Clercq
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of General Internal Medicine, Ghent University Hospital, Ghent, Belgium
| | | | - Virginie Mortier
- Department of Diagnostic Sciences, Aids Reference Laboratory, Ghent University, Ghent, Belgium
| | - Chris Verhofstede
- Department of Diagnostic Sciences, Aids Reference Laboratory, Ghent University, Ghent, Belgium
| | | | - Sabine D Allard
- Department of Internal Medicine, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Coca Necsoi
- Department of Infectious Diseases, Saint-Pierre University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Stéphane De Wit
- Department of Infectious Diseases, Saint-Pierre University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Sarah Gerlo
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Linos Vandekerckhove
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of General Internal Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
5
|
Boby N, Williams KM, Das A, Pahar B. Toll-like Receptor 2 Mediated Immune Regulation in Simian Immunodeficiency Virus-Infected Rhesus Macaques. Vaccines (Basel) 2023; 11:1861. [PMID: 38140264 PMCID: PMC10747659 DOI: 10.3390/vaccines11121861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Toll-like receptors (TLRs) are crucial to the innate immune response. They regulate inflammatory reactions by initiating the production of pro-inflammatory cytokines and chemokines. TLRs also play a role in shaping the adaptive immune responses. While this protective response is important for eliminating infectious pathogens, persistent activation of TLRs may result in chronic immune activation, leading to detrimental effects. The role of TLR2 in regulating HIV-1 infection in vivo has yet to be well described. In this study, we used an SIV-infected rhesus macaque model to simulate HIV infection in humans. We evaluated the plasma of the macaques longitudinally and found a significant increase in the soluble TLR2 (sTLR2) level after SIV infection. We also observed an increase in membrane-bound TLR2 (mb-TLR2) in cytotoxic T cells, B cells, and NK cells in PBMC and NK cells in the gut after infection. Our results suggest that sTLR2 regulates the production of various cytokines and chemokines, including IL-18, IL-1RA, IL-15, IL-13, IL-9, TPO, FLT3L, and IL-17F, as well as chemokines, including IP-10, MCP-1, MCP-2, ENA-78, GRO-α, I-TAC, Fractalkine, SDF-1α, and MIP-3α. Interestingly, these cytokines and chemokines were also upregulated after the infection. The positive correlation between SIV copy number and sTLR2 in the plasma indicated the involvement of TLR2 in the regulation of viral replication. These cytokines and chemokines could directly or indirectly regulate viral replication through the TLR2 signaling pathways. When we stimulated PBMC with the TLR2 agonist in vitro, we observed a direct induction of various cytokines and chemokines. Some of these cytokines and chemokines, such as IL-1RA, IL-9, IL-15, GRO-α, and ENA-78, were positively correlated with sTLR2 in vivo, highlighting the direct involvement of TLR2 in the regulation of the production of these factors. Our findings suggest that TLR2 expression may be a target for developing new therapeutic strategies to combat HIV infection.
Collapse
Affiliation(s)
- Nongthombam Boby
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA; (N.B.); (K.M.W.)
| | - Kelsey M. Williams
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA; (N.B.); (K.M.W.)
| | - Arpita Das
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA;
| | - Bapi Pahar
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA; (N.B.); (K.M.W.)
- School of Medicine, Tulane University, New Orleans, LA 70118, USA
| |
Collapse
|
6
|
Torices S, Daire L, Simon S, Mendoza L, Daniels D, Joseph JA, Fattakhov N, Naranjo O, Teglas T, Toborek M. The NLRP3 inflammasome and gut dysbiosis as a putative link between HIV-1 infection and ischemic stroke. Trends Neurosci 2023; 46:682-693. [PMID: 37330380 PMCID: PMC10554647 DOI: 10.1016/j.tins.2023.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/26/2023] [Accepted: 05/18/2023] [Indexed: 06/19/2023]
Abstract
HIV-associated comorbidities, such as ischemic stroke, are prevalent in people with HIV (PWH). Several studies both in animal models and humans have revealed an association between activation of the inflammasome in HIV-1 infection and stroke. The gut microbiota is an important component in controlling neuroinflammation in the CNS. It has also been proposed to be involved in the pathobiology of HIV-1 infection, and has been associated with an increase in activation of the inflammasome. In this review, we provide an overview of the microbiota-gut-inflammasome-brain axis, focusing on the NLRP3 inflammasome and dysregulation of the microbiome as risk factors that may contribute to the outcome of ischemic stroke and recovery in PWH. We also focus on the potential of targeting the NLRP3 inflammasome as a novel therapeutic approach for PWH who are at risk of developing cerebrovascular diseases.
Collapse
Affiliation(s)
- Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA.
| | - Leah Daire
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA
| | - Sierra Simon
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA
| | - Luisa Mendoza
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA
| | - Destiny Daniels
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA
| | - Joelle-Ann Joseph
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA
| | - Nikolai Fattakhov
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA
| | - Oandy Naranjo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA
| | - Timea Teglas
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA.
| |
Collapse
|
7
|
Guerville F, Vialemaringe M, Cognet C, Duffau P, Lazaro E, Cazanave C, Bonnet F, Leleux O, Rossignol R, Pinson B, Tumiotto C, Gabriel F, Appay V, Déchanet-Merville J, Wittkop L, Faustin B, Pellegrin I. Mechanisms of systemic low-grade inflammation in HIV patients on long-term suppressive antiretroviral therapy: the inflammasome hypothesis. AIDS 2023; 37:1035-1046. [PMID: 36928274 DOI: 10.1097/qad.0000000000003546] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
OBJECTIVE We aimed to determine the contribution of inflammasome activation in chronic low-grade systemic inflammation observed in patients with HIV (PWH) on long-term suppressive antiretroviral therapy (ART) and to explore mechanisms of such activation. DESIGN Forty-two PWH on long-term suppressive ART (HIV-RNA < 40 copies/ml) were compared with 10 HIV-negative healthy controls (HC). METHODS Inflammasome activation was measured by dosing mature interleukin (IL)-1β and IL-18 cytokines in patient serum. We explored inflammasome pathways through ex vivo stimulation of PWH primary monocytes with inflammasome activators; expression of inflammasome components by transcriptomic analysis; and metabolomics analysis of patient sera. RESULTS Median (Q1; Q3) age, ART and viral suppression duration in PWH were 54 (48; 60), 15 (9; 20) and 7.5 (5; 12) years, respectively. Higher serum IL-18 was measured in PWH than in HC (61 (42; 77) vs. 36 (27-48 pg/ml), P = 0.009); IL-1β was detected in 10/42 PWH (0.5 (0.34; 0.80) pg/ml) but not in HC. Monocytes from PWH did not produce more inflammatory cytokines in vitro , but secretion of IL-1β in response to NOD like receptor family, pyrin domain containing 3 (NLRP3) inflammasome stimulation was higher than in HC. This was not explained at the transcriptional level. We found an oxidative stress molecular profile in PWH sera. CONCLUSION HIV infection with long-term effective ART is associated with a serum inflammatory signature, including markers of inflammasome activation, and an increased activation of monocytes upon inflammasome stimulation. Other cells should be investigated as sources of inflammatory cytokines in PWH. Oxidative stress might contribute to this chronic low-grade inflammation.
Collapse
Affiliation(s)
| | | | - Celine Cognet
- CHU Bordeaux, Laboratory of Immunology and Immunogenetics
| | - Pierre Duffau
- University Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, INSERM ERL 1303
- CHU Bordeaux, Service de Médecine Interne et Immunologie Clinique
| | - Estibaliz Lazaro
- University Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, INSERM ERL 1303
- CHU Bordeaux, Service de Médecine Interne
| | | | - Fabrice Bonnet
- University Bordeaux, INSERM, Institut Bergonié, BPH, U1219, CIC-EC 1401
- CHU Bordeaux, Hôpital Saint-André, Service de Médecine Interne et Maladies Infectieuses
| | - Olivier Leleux
- University Bordeaux, INSERM, Institut Bergonié, BPH, U1219, CIC-EC 1401
| | - Rodrigue Rossignol
- INSERM U1211, 33000 Bordeaux, France; Bordeaux University; CELLOMET, Functional Genomics Center (CGFB), 146 rue Léo Saignat
| | - Benoît Pinson
- Service Analyses Métaboliques TBMcore CNRS UAR 3427 INSERM US005 Université de Bordeaux, 1 rue Camille Saint-Saëns
| | | | | | - Victor Appay
- University Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, INSERM ERL 1303
| | | | - Linda Wittkop
- University Bordeaux, INSERM, Institut Bergonié, BPH, U1219, CIC-EC 1401
- INRIA SISTM team, Talence
- CHU de Bordeaux, Service d'information médicale, INSERM, Institut Bergonié, CIC-EC 1401, Bordeaux, France
| | - Benjamin Faustin
- University Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, INSERM ERL 1303
- Immunology Discovery, Janssen Research & Development, San Diego, California, USA
| | - Isabelle Pellegrin
- University Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, INSERM ERL 1303
- CHU Bordeaux, Laboratory of Immunology and Immunogenetics
| |
Collapse
|
8
|
Liao Y, Fu Z, Huang Y, Wu S, Wang Z, Ye S, Zeng W, Zeng G, Li D, Yang Y, Pei K, Yang J, Hu Z, Liang X, Hu J, Liu M, Jin J, Cai C. Interleukin-18-primed human umbilical cord-mesenchymal stem cells achieve superior therapeutic efficacy for severe viral pneumonia via enhancing T-cell immunosuppression. Cell Death Dis 2023; 14:66. [PMID: 36707501 PMCID: PMC9883134 DOI: 10.1038/s41419-023-05597-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/29/2023]
Abstract
Coronavirus disease 2019 (COVID-19) treatments are still urgently needed for critically and severely ill patients. Human umbilical cord-mesenchymal stem cells (hUC-MSCs) infusion has therapeutic benefits in COVID-19 patients; however, uncertain therapeutic efficacy has been reported in severe patients. In this study, we selected an appropriate cytokine, IL-18, based on the special cytokine expression profile in severe pneumonia of mice induced by H1N1virus to prime hUC-MSCs in vitro and improve the therapeutic effect of hUC-MSCs in vivo. In vitro, we demonstrated that IL-18-primed hUC-MSCs (IL18-hUCMSC) have higher proliferative ability than non-primed hUC-MSCs (hUCMSCcon). In addition, VCAM-1, MMP-1, TGF-β1, and some chemokines (CCL2 and CXCL12 cytokines) are more highly expressed in IL18-hUCMSCs. We found that IL18-hUCMSC significantly enhanced the immunosuppressive effect on CD3+ T-cells. In vivo, we demonstrated that IL18-hUCMSC infusion could reduce the body weight loss caused by a viral infection and significantly improve the survival rate. Of note, IL18-hUCMSC can also significantly attenuate certain clinical symptoms, including reduced activity, ruffled fur, hunched backs, and lung injuries. Pathologically, IL18-hUCMSC transplantation significantly enhanced the inhibition of inflammation, viral load, fibrosis, and cell apoptosis in acute lung injuries. Notably, IL18-hUCMSC treatment has a superior inhibitory effect on T-cell exudation and proinflammatory cytokine secretion in bronchoalveolar lavage fluid (BALF). Altogether, IL-18 is a promising cytokine that can prime hUC-MSCs to improve the efficacy of precision therapy against viral-induced pneumonia, such as COVID-19.
Collapse
Affiliation(s)
- Yan Liao
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Zeqin Fu
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Yinfu Huang
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Shiduo Wu
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Zhen Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Shaotang Ye
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Weijie Zeng
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Guifang Zeng
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Duanduan Li
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Yulin Yang
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Ke Pei
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Jian Yang
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Zhiwei Hu
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Xiao Liang
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Junyuan Hu
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China.
| | - Muyun Liu
- National-Local Associated Engineering Laboratory for Personalized Cell Therapy, Shenzhen, 518054, China.
| | - Juan Jin
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, 310000, China.
| | - Cheguo Cai
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
9
|
Divergent Cytokine and Chemokine Responses at Early Acute Simian Immunodeficiency Virus Infection Correlated with Virus Replication and CD4 T Cell Loss in a Rhesus Macaque Model. Vaccines (Basel) 2023; 11:vaccines11020264. [PMID: 36851142 PMCID: PMC9963901 DOI: 10.3390/vaccines11020264] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
Cytokine and chemokine levels remain one of the significant predictive factors of HIV pathogenesis and disease outcome. Understanding the impact of cytokines and chemokines during early acute infection will help to recognize critical changes during HIV pathogenesis and might assist in establishing improved HIV treatment and prevention methods. Sixty-one cytokines and chemokines were evaluated in the plasma of an SIV-infected rhesus macaque model. A substantial change in 11 cytokines/growth factors and 9 chemokines were observed during acute infection. Almost all the cytokines/chemokines were below the baseline values for an initial couple of days of infection. We detected six important cytokines/chemokines, such as IL-18, IP-10, FLT3L, MCP-1, MCP-2, and MIP-3β, that can be used as biomarkers to predict the peripheral CD4+ T cell loss and increased viral replication during the acute SIV/HIV infection. Hence, regulating IL-18, IP-10, FLT3L, MCP-1, MCP-2, and MIP-3β expression might provide an antiviral response to combat acute SIV/HIV infection.
Collapse
|
10
|
Otterdal K, Berg A, Michelsen AE, Yndestad A, Patel S, Gregersen I, Halvorsen B, Ueland T, Langeland N, Aukrust P. IL-18 and IL-18 binding protein are related to disease severity and parasitemia during falciparum malaria. BMC Infect Dis 2021; 21:1073. [PMID: 34663245 PMCID: PMC8524870 DOI: 10.1186/s12879-021-06751-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/29/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Several inflammatory molecules participate in the immune response to malaria. Interleukin (IL)-18 is an inflammatory cytokine activated by NLRP3 inflammasomes. In clinical falciparum malaria, with and without HIV co-infection, data on IL-18 and in particular on its binding protein, IL-18bp, is scarce. METHODS Clinical data and blood samples were collected from adults in Mozambique with P. falciparum infection, with (n = 70) and without (n = 61) HIV co-infection, from HIV-infected patients with similar symptoms without malaria (n = 58) and from healthy controls (n = 52). In vitro studies were performed in endothelial cells using hemozoin crystals. RESULTS (i) IL-18 and IL-18bp were markedly up-regulated during falciparum malaria with particular high levels in malaria patients co-infected with HIV and severe malaria disease. (ii) In the malaria group as a whole, both IL-18 and IL-18bp were positively correlated with disease severity, parasitemia, and endothelial cell activation as assessed by vWF in plasma. (iii) Whereas there was no change in IL-18 levels in malaria patients co-infected with HIV during follow-up, the patients with malaria only had slightly increased IL-18 levels. Further, the IL-18pb levels declined and thereby contributed to an increase in IL-18/IL-18bp ratio in all subgroups of malaria patients. (iv) IL-27, previously shown to be up-regulated in this malaria cohort, markedly induced a release of IL-18bp from endothelial cells in vitro, and notably, this presumably anti-inflammatory effect was counteracted by hemozoin. CONCLUSIONS Our findings suggest that the IL-18 system could be an important mediator in the immune pathogenesis during falciparum malaria, potentially also representing a target for therapy.
Collapse
Affiliation(s)
- Kari Otterdal
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Nydalen, PO Box 4950, 0424, Oslo, Norway.
| | - Aase Berg
- Department of Medicine, Stavanger University Hospital, PO Box 8100, 4068, Stavanger, Norway.,Department of Medicine, Central Hospital of Maputo, Maputo, Mozambique
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Nydalen, PO Box 4950, 0424, Oslo, Norway.,Faculty of Medicine, University of Oslo, 0316, Oslo, Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Nydalen, PO Box 4950, 0424, Oslo, Norway.,Faculty of Medicine, University of Oslo, 0316, Oslo, Norway
| | - Sam Patel
- Department of Medicine, Central Hospital of Maputo, Maputo, Mozambique
| | - Ida Gregersen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Nydalen, PO Box 4950, 0424, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Nydalen, PO Box 4950, 0424, Oslo, Norway.,Faculty of Medicine, University of Oslo, 0316, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Nydalen, PO Box 4950, 0424, Oslo, Norway.,Faculty of Medicine, University of Oslo, 0316, Oslo, Norway.,K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, 9019, Tromsø, Norway
| | - Nina Langeland
- Department of Clinical Science, University of Bergen, 5021, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, 5021, Bergen, Norway.,Department of Medicine, Haraldsplass Deaconess Hospital, 5009, Bergen, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Nydalen, PO Box 4950, 0424, Oslo, Norway.,Faculty of Medicine, University of Oslo, 0316, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, 0372, Oslo, Norway
| |
Collapse
|
11
|
Madeen EP, Maldarelli F, Groopman JD. Environmental Pollutants, Mucosal Barriers, and Pathogen Susceptibility; The Case for Aflatoxin B 1 as a Risk Factor for HIV Transmission and Pathogenesis. Pathogens 2021; 10:1229. [PMID: 34684180 PMCID: PMC8537633 DOI: 10.3390/pathogens10101229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/02/2022] Open
Abstract
HIV transmission risk is dependent on the infectivity of the HIV+ partner and personal susceptibility risk factors of the HIV- partner. The mucosal barrier, as the internal gatekeeper between environment and self, concentrates and modulates the internalization of ingested pathogens and pollutants. In this review, we summarize the localized effects of HIV and dietary toxin aflatoxin B1 (AFB1), a common pollutant in high HIV burden regions, e.g., at the mucosal barrier, and evidence for pollutant-viral interactions. We compiled literature on HIV and AFB1 geographic occurrences, mechanisms of action, related co-exposures, personal risk factors, and HIV key determinants of health. AFB1 exposure and HIV sexual transmission hotspots geographically co-localize in many low-income countries. AFB1 distributes to sexual mucosal tissues generating inflammation, microbiome changes and a reduction of mucosal barrier integrity, effects that are risk factors for increasing HIV susceptibility. AFB1 exposure has a positive correlation to HIV viral load, a risk factor for increasing the infectivity of the HIV+ partner. The AFB1 exposure and metabolism generates inflammation that recruits HIV susceptible cells and generates chemokine/cytokine activation in tissues exposed to HIV. Although circumstantial, the available evidence makes a compelling case for studies of AFB1 exposure as a risk factor for HIV transmission, and a modifiable new component for combination HIV prevention efforts.
Collapse
Affiliation(s)
- Erin P. Madeen
- Department of Cancer Prevention, National Institute of Health, Shady Grove, MD 21773, USA
- HIV Dynamics and Replication Program, NCI-Frederick, Frederick, MD 21703, USA;
| | - Frank Maldarelli
- HIV Dynamics and Replication Program, NCI-Frederick, Frederick, MD 21703, USA;
| | - John D. Groopman
- Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA;
| |
Collapse
|
12
|
Association of cell free mitochondrial DNA and caspase-1 expression with disease severity and ARTs efficacy in HIV infection. Mol Biol Rep 2021; 48:3327-3336. [PMID: 33886057 DOI: 10.1007/s11033-021-06313-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/24/2021] [Indexed: 10/21/2022]
Abstract
HIV infection is a global health concern. Current HIV-diagnostics provide information about the disease progression and efficacy of anti-retroviral therapies (ARVs), but this information is very limited and sometimes imprecise. Present study assessed the potential role of mononuclear cell (MNC) death, expression of caspases (1&3) and cell free mitochondrial DNA (CF mt-DNA) in HIV infected individuals. Apoptosis, cell-count, expression of caspases and CF mt-DNA were measured through flow cytometry and qPCR, respectively, in HIV infected individuals (n = 120) divided in two groups i.e. ARVs-receiving (treated, n = 87), ART-naïve (untreated, n = 37) and healthy individuals (n = 47). Data showed significant (p < 0.0001) cell death in untreated individuals than treated and healthy individuals. CD4-positive T-cell percentage declined (p < 0.0001) in untreated as compared to treated individuals. Caspase-1, an indicator of pyroptosis, and CF mt-DNA were also elevated in untreated HIV infected individuals. Untreated individuals when administered with ARVs showed improved CD4-positive T-cell percentage, lower caspase-1, CF mt-DNA and cell death. Data elucidated positive co-relation between cell death and CF mt-DNA in treated and untreated HIV infected individuals. While CD4-positive T-cell percentage was negatively correlated with caspase-1 expression and CF mt-DNA. Elevated levels of CF mt-DNA and caspase-1 in HIV infected individuals, positive correlation between cell death and CF mt-DNA, negative correlation of CD4-positive T-cell percentage with CF mt-DNA and caspase-1 expression clearly indicated the potential of CF mt-DNA and caspase-1 as a novel disease progression and ARTs effectiveness biomarkers in HIV.
Collapse
|
13
|
Differential recognition of HIV-stimulated IL-1β and IL-18 secretion through NLR and NAIP signalling in monocyte-derived macrophages. PLoS Pathog 2021; 17:e1009417. [PMID: 33861800 PMCID: PMC8109768 DOI: 10.1371/journal.ppat.1009417] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 05/10/2021] [Accepted: 02/22/2021] [Indexed: 12/29/2022] Open
Abstract
Macrophages are important drivers of pathogenesis and progression to AIDS in HIV infection. The virus in the later phases of the infection is often predominantly macrophage-tropic and this tropism contributes to a chronic inflammatory and immune activation state that is observed in HIV patients. Pattern recognition receptors of the innate immune system are the key molecules that recognise HIV and mount the inflammatory responses in macrophages. The innate immune response against HIV-1 is potent and elicits caspase-1-dependent pro-inflammatory cytokine production of IL-1β and IL-18. Although, NLRP3 has been reported as an inflammasome sensor dictating this response little is known about the pattern recognition receptors that trigger the “priming” signal for inflammasome activation, the NLRs involved or the HIV components that trigger the response. Using a combination of siRNA knockdowns in monocyte derived macrophages (MDMs) of different TLRs and NLRs as well as chemical inhibition, it was demonstrated that HIV Vpu could trigger inflammasome activation via TLR4/NLRP3 leading to IL-1β/IL-18 secretion. The priming signal is triggered via TLR4, whereas the activation signal is triggered by direct effects on Kv1.3 channels, causing K+ efflux. In contrast, HIV gp41 could trigger IL-18 production via NAIP/NLRC4, independently of priming, as a one-step inflammasome activation. NAIP binds directly to the cytoplasmic tail of HIV envelope protein gp41 and represents the first non-bacterial ligand for the NAIP/NLRC4 inflammasome. These divergent pathways represent novel targets to resolve specific inflammatory pathologies associated with HIV-1 infection in macrophages. It has been previously shown that inflammasome activation can be triggered during viral infection to produce the active cytokines IL-1β and IL-18. Our study represents a significant advance, as we now show that in fact there are distinct NLR inflammasome complexes and viral ligands for IL-1β secretion (Vpu) compared to IL-18 secretion (gp41) in response to HIV-1. Most importantly, we show that the HIV envelope protein gp41 represents the first non-bacterial ligand for the assembly of the NAIP/NLRC4 inflammasome. HIV gp41 is a viroporin, and thus our data demonstrates for the first time that the NAIP/NLRC4 inflammasome assembles for all pore-forming proteins, irrespective of whether they have a viral or bacterial origin. This is critical for the host antiviral response and has broad implications for innate immunity in general.
Collapse
|
14
|
Okay G, Koc MM, Guler EM, Yabaci A, Kocyigit A, Akkoyunlu Y. The Effect of Antiretroviral Therapy on IL-6, IL-1β, TNF-α, IFN-γ Levels and their Relationship with HIV-RNA and CD4+ T Cells in HIV Patients. Curr HIV Res 2020; 18:354-361. [PMID: 32652911 DOI: 10.2174/1570162x18666200712174642] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/10/2020] [Accepted: 07/15/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND Serum cytokine levels over the course of HIV infection usually increase with immunosuppression and decrease after antiretroviral treatment (ART). OBJECTIVES The aim of the study is to compare cytokine levels between HIV-infected patients (HIP) and controls and investigate the relationship between CD4+T cell count, HIV-RNA levels, and cytokine levels. METHODS The study subjects comprised ART-naive HIP (n=30) with no comorbidities and age-and sex-matched healthy controls. We measured levels of IL-6, IL-1β, TNF-α, and IFN-γ in serum samples of HIP at the beginning and at month 6 of ART and in controls. RESULTS The mean age of the study subjects was 38.7 ±10.3 years, with men making up 86.7% of the study subjects (n=26). IL-6, IL-1β, and TNF-α levels were significantly higher in both ART-naive (p<0.001, p=0.002, p=0.001) and ART-experienced HIP (p<0.001) than controls. The IFN-γ level was lower in both ART-naive and ART-experienced HIP compared to controls (p=0.082 and p=0.002). There was a positive correlation between the CD4+T cell count and serum concentration of IFN- γ(r=0.320, p<0.05). While the serum IFN-γ concentration showed a negative correlation with the HIVRNA level(r=-0.412, p<0.001), the serum IL-1β, IL-6, and TNF-α concentrations showed a positive correlation with the HIV-RNA level (r=0.349, p<0.001; r:0.54, p<0.001; r:0.438, p<0.00). CONCLUSION Although serum concentrations of IL-6, IL-1β and TNF-α showed a significant decrease after ART, they were still significantly higher than the controls. IFN-γ responded differently to ART compared to the other cytokines, indicating that it may play a distinct and important role in the pathogenesis of HIV infection.
Collapse
Affiliation(s)
- Gülay Okay
- Bezmialem Vakif University, Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Istanbul, Turkey
| | - Meliha Meric Koc
- Bezmialem Vakif University, Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Istanbul, Turkey
| | - Eray Metin Guler
- Bezmialem Vakif University, Faculty of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
| | - Ayşegül Yabaci
- Bezmialem Vakif University, Department of Biostatistics, Istanbul, Turkey
| | - Abdürrahim Kocyigit
- Bezmialem Vakif University, Faculty of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
| | - Yasemin Akkoyunlu
- Bezmialem Vakif University, Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Istanbul, Turkey
| |
Collapse
|
15
|
Vecchié A, Bonaventura A, Toldo S, Dagna L, Dinarello CA, Abbate A. IL-18 and infections: Is there a role for targeted therapies? J Cell Physiol 2020; 236:1638-1657. [PMID: 32794180 DOI: 10.1002/jcp.30008] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/24/2020] [Accepted: 08/01/2020] [Indexed: 01/08/2023]
Abstract
Interleukin (IL)-18 is a pro-inflammatory cytokine belonging to the IL-1 family, first identified for its interferon-γ-inducing properties. IL-18 regulates both T helper (Th) 1 and Th2 responses. It acts synergistically with IL-12 in the Th1 paradigm, whereas with IL-2 and without IL-12 it can induce Th2 cytokine production from cluster of differentation (CD)4+ T cells, natural killer (NK cells, NKT cells, as well as from Th1 cells. IL-18 also plays a role in the hemophagocytic lymphohistiocytosis, a life-threatening condition characterized by a cytokine storm that can be secondary to infections. IL-18-mediated inflammation was largely studied in animal models of bacterial, viral, parasitic, and fungal infections. These studies highlight the contribution of either IL-18 overproduction by the host or overresponsiveness of the host to IL-18 causing an exaggerated inflammatory burden and leading to tissue injury. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the coronavirus disease 2019 (COVID-19). The damage in the later phase of the disease appears to be driven by a cytokine storm, including interleukin IL-1 family members and secondary cytokines like IL-6. IL-18 may participate in this hyperinflammation, as it was previously found to be able to cause injury in the lung tissue of infected animals. IL-18 blockade has become an appealing therapeutic target and has been tested in some IL-18-mediated rheumatic diseases and infantile-onset macrophage activation syndrome. Given its role in regulating the immune response to infections, IL-18 blockade might represent a therapeutic option for COVID-19, although further studies are warranted to investigate more in detail the exact role of IL-18 in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Alessandra Vecchié
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | - Aldo Bonaventura
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia.,Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
| | - Stefano Toldo
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Charles A Dinarello
- Department of Medicine and Immunology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Antonio Abbate
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
16
|
The IL-18, IL-12, and IFN-γ expression in HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) patients, HTLV-1 carriers, and healthy subjects. J Neurovirol 2020; 26:338-346. [PMID: 32270468 DOI: 10.1007/s13365-020-00832-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 01/23/2020] [Accepted: 02/13/2020] [Indexed: 10/24/2022]
Abstract
Interleukin (IL)-12, IL-18, and interferon gamma (IFN-γ) can induce Th1-inflammatory responses in favor of HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) manifestation. In this study, the gene expression and plasma levels of these cytokines were evaluated. The peripheral blood mononuclear cells (PBMCs) in 20 HAM/TSP patients, 21 asymptomatic carriers (ACs), and 21 healthy subjects (HSs) were assessed for the expression of IL-18, IL-12, and IFN-γ, using qRT-PCR. The plasma level of IL-18 and IFN-γ were measured by an ELISA method. The mean of HTLV-1 proviral load (PVL) in the HAM/TSPs was 1846.59 ± 273.25 and higher than ACs at 719.58 ± 150.72 (p = 0.001). The IL-12 was considerably expressed only in nine ACs, five HAM/TSPs, and all HSs. Furthermore, the gene expression and plasma levels of IL-18 were lower in the HTLV-1-positive group than the control group (p = 0.001 and 0.012, respectively); however, there was no significant difference between the ACs and HAM/TSPs. The IFN-γ level was higher in the HTLV-1-positive group (p < 0.001) than HSs. Although there were no correlation between plasma levels of IL-18 and IFN-γ with PVL in the ACs, a positive correlation was observed between plasma IL-18 levels and PVL (r = 0.654, p = 0.002). The highest levels of IFN-γ were observed in the HAM/TSPs which has a significant correlation with HTLV-1-HBZ (r = 0.387, p = 0.05) but not with Tax. However, no significant correlation was found between PVL and proinflammatory pattern. Apart from the IFN-γ as a lymphokine, as a host factor, and HTLV-1-HBZ, as a viral agent, the other proinflammatory monokines or HTLV-1 factors are among the less-effective agents in the maintenance of HAM/TSP.
Collapse
|
17
|
Leal VNC, Reis EC, Pontillo A. Inflammasome in HIV infection: Lights and shadows. Mol Immunol 2019; 118:9-18. [PMID: 31835091 DOI: 10.1016/j.molimm.2019.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/27/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023]
Abstract
The importance of inflammasome, and related cytokines IL-1ß and IL-18, in host defense against pathogens is well documented, however, at the same time, dysregulation of inflammasome has been associated to multifactorial diseases characterized by chronic inflammation (i.e.: metabolic disorders, cardiovascular diseases, neurodegenerative diseases, autoimmunity, cancer). Inflammasome activation has been described in response to HIV-1 and possibly contributes to the resistance against virus establishment, however, on the other hand, when viral infection becomes chronic, independently from antiretroviral therapy, the increase constitutive activation of inflammasome has been eventually associated to a worse prognosis, raising the question about the role played by inflammasome and/or some specific receptors in this context. Due to the chance to imply targeted therapies that inhibit inflammasome activation and/or cytokines release, it will be important to define the impact of the complex in the pathogenesis of HIV. The purpose of this review is to depict the double-faced inflammasome role in HIV-1 infection, trying to unveil whether besides its role in first line defense against the virus, it exerts a harmful effect during the chronic phase of infection.
Collapse
Affiliation(s)
- Vinicius Nunes Cordeiro Leal
- Laboratorio de Imunogenetica, Departamento de Imunologia, Instituto de Ciencias Biomedicas (ICB), Universidade de Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Edione Cristina Reis
- Laboratorio de Imunogenetica, Departamento de Imunologia, Instituto de Ciencias Biomedicas (ICB), Universidade de Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Alessandra Pontillo
- Laboratorio de Imunogenetica, Departamento de Imunologia, Instituto de Ciencias Biomedicas (ICB), Universidade de Sao Paulo (USP), Sao Paulo, SP, Brazil.
| |
Collapse
|
18
|
Matamoros JA, da Silva MIF, de Moura PMMF, Leitão MDCG, Coimbra EC. Reduced Expression of IL-1β and IL-18 Proinflammatory Interleukins Increases the Risk of Developing Cervical Cancer. Asian Pac J Cancer Prev 2019; 20:2715-2721. [PMID: 31554368 PMCID: PMC6976845 DOI: 10.31557/apjcp.2019.20.9.2715] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Indexed: 12/13/2022] Open
Abstract
Background: The objective of this study was to analyze the gene expression profile of the proinflammatory interleukins, (IL-1β and IL-18) in patients with premalignant lesions and cervical cancer. Methods: Total IL-1β and IL-18 mRNA was quantified by qPCR to obtain the expression data in cervical tissues. A total of 74 cervical biopsies were obtained from women undergoing a colposcopy. The samples were divided into: normal (19), low level lesions (LSIL) or NIC I (17), high level lesions (HSIL) or CIN II and CIN III (29) and cancer (9). The normal cervical tissue samples were included as controls. The OR and 95% CI were calculated for the determination of the risk of progression between each type of lesion and cancer using logistic regression. Results: The results showed that an increase in the risk of progression of pre-neoplastic lesions to cancer was between 2.5 and 2.08 times higher in women with lower IL-1β and IL-18 expression, respectively. Conclusions: This study provided evidence that IL-1β and IL-18 are potential biomarkers that can be explored in further studies for monitoring the evolution of pre-neoplastic lesions and avoiding overtreatment or undertreatment of the patients.
Collapse
Affiliation(s)
- Jose Anibal Matamoros
- Laboratory of Molecular Biology of Viruses, Biological Sciences Institute, University of Pernambuco, Brazil.
| | | | | | - Maria da Conceição Gomes Leitão
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Biological Sciences Center, Federal University of Pernambuco, Recife, Brazil
| | - Eliane Campos Coimbra
- Laboratory of Molecular Biology of Viruses, Biological Sciences Institute, University of Pernambuco, Brazil.
| |
Collapse
|
19
|
Feria MG, Taborda NA, Hernandez JC, Rugeles MT. HIV replication is associated to inflammasomes activation, IL-1β, IL-18 and caspase-1 expression in GALT and peripheral blood. PLoS One 2018; 13:e0192845. [PMID: 29672590 PMCID: PMC5909617 DOI: 10.1371/journal.pone.0192845] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 01/31/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus (HIV) promotes an inflammatory process, leading to the progressive loss of the functional capacity of the immune system. The HIV infection induces alterations in several tissues, but mainly in the gut-associated lymphoid tissue (GALT). However, the degree of GALT deterioration varies among infected individuals. In fact, it has been shown that HIV-controllers, who spontaneously control viral replication, exhibit a lower inflammatory response, and a relative normal frequency and function of most of the immune cells. Inflammasomes are molecular complexes involved in the inflammatory response, being NLRP1, NLRP3, NLRC4, AIM2 and Pyrin inflammasomes, the best characterized so far. These complexes regulate the maturation of cytokines of the IL-1 family, including IL-1β and IL-18. These cytokines have been associated with immune activation and expansion of HIV target cells, promoting viral replication. Interesting, some reports indicate that HIV induces the activation of the NLRP3 inflammasome, but the role of this, and other inflammasomes during HIV infection, especially in GALT, remains unclear. OBJECTIVE To compare the relative expression of inflammasome components and the proinflammatory response related to their activity, between HIV-progressors and HIV-controllers. METHODS GALT biopsies and peripheral blood mononuclear cells (PBMCs) from 15 HIV-controllers and 15 HIV-progressors were obtained. The relative expression of the following inflammasome components were evaluated by RT-PCR: NLRP3, NLRC4, NLRP1, AIM2, ASC, Caspase-1, IL-1β and IL-18. In addition, plasma concentration of IL-18 was evaluated as an indicator of baseline proinflammatory status. Finally, in supernatants of PBMCs in vitro stimulated with inflammasome agonists, the concentrations of IL-1β and IL-18 were quantified by ELISA. RESULTS HIV-progressors exhibited higher expression of IL-1β, IL-18 and caspase-1 genes in GALT and PBMCs compared with HIV-controllers. In addition, HIV-progressors had also increased expression of ASC in PBMCs. When plasma levels were evaluated, IL-18 was increased in HIV-progressors. Interesting, these patients also showed an increased production of IL-1β in supernatants of PBMCs stimulated in vitro with the agonists of AIM2, NLRP1 and NLRC4 inflammasomes. Finally, the expression of caspase-1, NLRP1, IL-1β and IL-18 in GALT or peripheral blood was correlated with CD4+ T-cell count and viral load. CONCLUSION Our results suggest that during HIV-infection, the required signals to induce the expression of different components of the inflammasomes are produced, both in GALT and in periphery. The activation of these molecular complexes could increase the number of target cells, favoring HIV replication and cell death, promoting the disease progression.
Collapse
Affiliation(s)
- Manuel Gerónimo Feria
- Grupo Inmunovirología, Facultad de medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Natalia Andrea Taborda
- Grupo Inmunovirología, Facultad de medicina, Universidad de Antioquia UdeA, Medellín, Colombia
- Grupo de Investigaciones Biomédicas Uniremington, Programa de Medicina, Facultad de Ciencias de la Salud, Corporación Universitaria Remington, Medellín, Colombia
| | - Juan C. Hernandez
- Grupo Inmunovirología, Facultad de medicina, Universidad de Antioquia UdeA, Medellín, Colombia
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
| | - Maria Teresa Rugeles
- Grupo Inmunovirología, Facultad de medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| |
Collapse
|
20
|
Allam O, Samarani S, Mehraj V, Jenabian MA, Tremblay C, Routy JP, Amre D, Ahmad A. HIV induces production of IL-18 from intestinal epithelial cells that increases intestinal permeability and microbial translocation. PLoS One 2018; 13:e0194185. [PMID: 29601578 PMCID: PMC5877838 DOI: 10.1371/journal.pone.0194185] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 02/26/2018] [Indexed: 12/24/2022] Open
Abstract
Interleukin-18 (IL-18) is a pleiotropic cytokine of the IL-1 family with multiple context dependent functions. We and others have shown that HIV infection is accompanied by increased circulating levels of IL-18 along with decreased levels of its antagonist, Interleukin-18 Binding Protein (IL-18BP). The infection is also accompanied by intestinal inflammation and decreased intestinal integrity as measured by intestinal permeability, regeneration and repair. However, little is known concerning the relation between high level of IL-18 associated with the viral infection and intestinal permeability. Here we demonstrate that HIV treatment increases production of IL-18 and decreases that of IL-18BP production in human intestinal epithelial cell (IEC) lines. IL-18 causes apoptosis of the IEC by activating caspase-1 and caspase-3. It induces epithelial barrier hyperpermeability by decreasing and disrupting both tight and adherens junction proteins, occludin, claudin 2 and beta-catenin. Disorganization of F-actin was also observed in the IEC that were exposed to the cytokine. Moreover IL-18 decreases transepithelial electrical resistance (TEER) in Caco-2 and increases permeability in HT29 monolayers. The cells' treatment with IL-18 causes an increase in the expression of phosphorylated myosin II regulatory light-chain (p-MLC) and myosin light-chain kinase (MLCK), and a decrease in phosphorylated Signal Transducer and Activator of Transcription (p-STAT)-5. This increase in p-MLC is suppressed by a Rho-kinase (ROCK)-specific inhibitor. Interestingly, the levels of the cytokine correlate with those of LPS in the circulation in three different categories of HIV infected patients (HAART-naïve and HAART-treated HIV-infected individuals, and Elite controls) as well as in healthy controls. Collectively, these results suggest that the HIV-induced IL-18 plays a role in increased intestinal permeability and microbial translocation observed in HIV-infected individuals.
Collapse
Affiliation(s)
- Ossama Allam
- Laboratory of Innate Immunity, CHU Ste-Justine Research Center/Department of Microbiology, Infectious Diseases & Immunology, University of Montreal, Montreal, Québec, Canada
| | - Suzanne Samarani
- Laboratory of Innate Immunity, CHU Ste-Justine Research Center/Department of Microbiology, Infectious Diseases & Immunology, University of Montreal, Montreal, Québec, Canada
| | - Vikram Mehraj
- Division of Hematology & Chronic Viral Illness Service, McGill University, Montreal, Québec, Canada
| | | | - Cecile Tremblay
- CHUM/ Department of Microbiology, Infectious Diseases & Immunology, University of Montreal, Montreal, Québec, Canada
| | - Jean-Pierre Routy
- Division of Hematology & Chronic Viral Illness Service, McGill University, Montreal, Québec, Canada
| | - Devendra Amre
- CHU Ste-Justine Research Center/Department of Pediatrics, University of Montreal, Montreal, Québec, Canada
| | - Ali Ahmad
- Laboratory of Innate Immunity, CHU Ste-Justine Research Center/Department of Microbiology, Infectious Diseases & Immunology, University of Montreal, Montreal, Québec, Canada
- * E-mail:
| |
Collapse
|
21
|
Wu X, Zhang LL, Yin LB, Fu YJ, Jiang YJ, Ding HB, Chu ZX, Shang H, Zhang ZN. Deregulated MicroRNA-21 Expression in Monocytes from HIV-Infected Patients Contributes to Elevated IP-10 Secretion in HIV Infection. Front Immunol 2017; 8:1122. [PMID: 28955339 PMCID: PMC5601991 DOI: 10.3389/fimmu.2017.01122] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/25/2017] [Indexed: 01/12/2023] Open
Abstract
Persistent activation and inflammation impair immune response and trigger disease progression in HIV infection. Emerging evidence supports the supposition that excessive production of interferon-inducible protein 10 (IP-10), a critical inflammatory cytokine, leads to immune dysfunction and disease progression in HIV infection. In this study, we sought to elucidate the cause of the upregulated production of IP-10 in HIV infection and explore the underlying mechanisms. Bolstering miR-21 levels using mimics resulted in the obvious suppression of lipopolysaccharide (LPS)-induced IP-10 in monocyte leukemia cells THP-1 and vice versa. The analysis of the primary monocytes of HIV patients revealed significantly less miR-21 than in healthy controls; this was opposite to the tendency of IP-10 levels in plasma. The secretion of IP-10 due to LPS stimulation was not affected by miR-21 modulation in the differentiated THP-1 macrophages (THP-1-MA). We found a novel switch, IFN-stimulated gene 15 (ISG15), which triggers the expression of IP-10 and is significantly upregulated during the differentiation of THP-1 into THP-1-MA. The inhibition of ISG15 can restore the regulation of IP-10 by miR-21. In summary, IP-10 expression in monocytes is regulated by miR-21, whereas in macrophages, this fine-tuning is attenuated by the enhanced expression of ISG15. This study paves the way to a comprehensive understanding of the molecular regulatory mechanism of IP-10, a key point in immune intervention strategy.
Collapse
Affiliation(s)
- Xian Wu
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Le-Le Zhang
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Lin-Bo Yin
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Ya-Jing Fu
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Yong-Jun Jiang
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Hai-Bo Ding
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Zhen-Xing Chu
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Hong Shang
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Zi-Ning Zhang
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| |
Collapse
|
22
|
Freeman ML, Morris SR, Lederman MM. CD161 Expression on Mucosa-Associated Invariant T Cells is Reduced in HIV-Infected Subjects Undergoing Antiretroviral Therapy Who Do Not Recover CD4 + T Cells. Pathog Immun 2017; 2:335-351. [PMID: 28868514 PMCID: PMC5578469 DOI: 10.20411/pai.v2i3.136] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background: Mucosa-associated invariant T (MAIT) cells are a recently identified class of innate-like T cells that are involved in the mucosal immune response. MAIT cells are characterized by expression of TCR Vα7.2 and CD161. In HIV infection, there is a profound early loss of MAIT cells from the circulation that never fully recovers, even after prolonged viral control with antiretroviral therapy (ART). Methods: We analyzed PBMCs from fresh whole blood from HIV-negative or ART-treated HIV-positive donors with full (Immune Success) or impaired (Immune Failure) CD4+ T- cell recovery by flow cytometry for T-cell markers, TCR Vα7.2, and CD161. The PBMCs were cultured with or without TCR-mediated stimulation, and CD161 expression was assessed on Vα7.2+ T cells. Interferon-γ (IFNγ) production was assessed by intracellular cytokine staining. Results: We found a decrease in the percentage of CD3+ T cells that expressed CD161 and the percentage of Vα7.2+ T cells that expressed CD161, in HIV-infected individuals. We also found a significant increase in the percentage of T cells that were Vα7.2+CD161- in immune failure compared to controls, accompanied by an increase in the percentage of Vα7.2+CD161- T cells that express CD8+ in donors with immune failure, but not immune success. After TCR stimulation in vitro, Vα7.2+ T cells reduced expression of CD161, yet Vα7.2+ CD161- cells from immune failure donors retained the ability to express IFNγ on stimulation. Conclusions: Our findings suggest that in immune failure patients, the reduction in peripheral MAIT cells is due, at least in part, to a loss in CD161 expression, and is not merely the result of trafficking into mucosal tissues or cell death. These CD161- cells retain their function.
Collapse
Affiliation(s)
- Michael L Freeman
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | - Michael M Lederman
- Center for AIDS Research, Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, Ohio
| |
Collapse
|
23
|
Slaats J, ten Oever J, van de Veerdonk FL, Netea MG. IL-1β/IL-6/CRP and IL-18/ferritin: Distinct Inflammatory Programs in Infections. PLoS Pathog 2016; 12:e1005973. [PMID: 27977798 PMCID: PMC5158075 DOI: 10.1371/journal.ppat.1005973] [Citation(s) in RCA: 217] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The host inflammatory response against infections is characterized by the release of pro-inflammatory cytokines and acute-phase proteins, driving both innate and adaptive arms of the immune response. Distinct patterns of circulating cytokines and acute-phase responses have proven indispensable for guiding the diagnosis and management of infectious diseases. This review discusses the profiles of acute-phase proteins and circulating cytokines encountered in viral and bacterial infections. We also propose a model in which the inflammatory response to viral (IL-18/ferritin) and bacterial (IL-6/CRP) infections presents with specific plasma patterns of immune biomarkers.
Collapse
Affiliation(s)
- Jeroen Slaats
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- * E-mail:
| | - Jaap ten Oever
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frank L. van de Veerdonk
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
24
|
Iannello A, Samarani S, Allam O, Jenabian MA, Mehraj V, Amre D, Routy JP, Tremblay C, Ahmad A. A potentially protective role of IL-18 Binding Protein in HIV-infected Long-Term Non-Progressors. Cytokine 2016; 90:96-99. [PMID: 27863336 DOI: 10.1016/j.cyto.2016.10.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/26/2016] [Accepted: 10/28/2016] [Indexed: 02/07/2023]
Abstract
An imbalance between IL-18 and its antagonist, IL-18 Binding Protein, occurs in the circulation of HIV-infected individuals. We show here for the first time that HIV-infected Long Term Non-Progressors (LTNPs) do not develop this imbalance, and maintain normal levels of IL-18BP in the circulation. Their circulating levels of the antagonist correlate negatively with viral loads and show a positive trend with CD4+ T cells counts. The maintenance of normal production of IL-18BP may contribute, at least in part, to the ability of LTNPs to delay AIDS progression.
Collapse
Affiliation(s)
- Alexandre Iannello
- Laboratory of Innate Immunity, Centre Hospitalier Universitaire (CHU) Ste-Justine Research Center/Department of Microbiology, Infectiology & Immunology, University of Montreal, Montreal, Quebec, Canada
| | - Suzanne Samarani
- Laboratory of Innate Immunity, Centre Hospitalier Universitaire (CHU) Ste-Justine Research Center/Department of Microbiology, Infectiology & Immunology, University of Montreal, Montreal, Quebec, Canada
| | - Ossama Allam
- Laboratory of Innate Immunity, Centre Hospitalier Universitaire (CHU) Ste-Justine Research Center/Department of Microbiology, Infectiology & Immunology, University of Montreal, Montreal, Quebec, Canada
| | | | - Vikram Mehraj
- Division of Hematology & Chronic Viral Service, McGill University, Montreal, Quebec, Canada
| | - Devendra Amre
- CHU Ste-Justine Research Center/Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada
| | - Jean-Pierre Routy
- Division of Hematology & Chronic Viral Service, McGill University, Montreal, Quebec, Canada
| | - Cécile Tremblay
- CHUM/Department of Microbiology, Infectiology & Immunology, University of Montreal, Montreal, Quebec, Canada
| | - Ali Ahmad
- Laboratory of Innate Immunity, Centre Hospitalier Universitaire (CHU) Ste-Justine Research Center/Department of Microbiology, Infectiology & Immunology, University of Montreal, Montreal, Quebec, Canada.
| |
Collapse
|
25
|
Balagopal A, Gupte N, Shivakoti R, Cox AL, Yang WT, Berendes S, Mwelase N, Kanyama C, Pillay S, Samaneka W, Santos B, Poongulali S, Tripathy S, Riviere C, Lama JR, Cardoso SW, Sugandhavesa P, Semba RD, Hakim J, Hosseinipour MC, Kumarasamy N, Sanne I, Asmuth D, Campbell T, Bollinger RC, Gupta A. Continued Elevation of Interleukin-18 and Interferon-γ After Initiation of Antiretroviral Therapy and Clinical Failure in a Diverse Multicountry Human Immunodeficiency Virus Cohort. Open Forum Infect Dis 2016; 3:ofw118. [PMID: 27800521 PMCID: PMC5084713 DOI: 10.1093/ofid/ofw118] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/19/2016] [Indexed: 01/04/2023] Open
Abstract
Background. We assessed immune activation after antiretroviral therapy (ART) initiation to understand clinical failure in diverse settings. Methods. We performed a case-control study in ACTG Prospective Evaluation of Antiretrovirals in Resource-Limited Settings (PEARLS). Cases were defined as incident World Health Organization Stage 3 or 4 human immunodeficiency virus (HIV) disease or death, analyzed from ART weeks 24 (ART24) to 96. Controls were randomly selected. Interleukin (IL)-6, interferon (IFN)-γ-inducible protein-10, IL-18, tumor necrosis factor-α, IFN-γ, and soluble CD14 (sCD14) were measured pre-ART and at ART24 in plasma. Continued elevation was defined by thresholds set by highest pre-ART quartiles (>Q3). Incident risk ratios (IRRs) for clinical progression were estimated by Poisson regression, adjusting for age, sex, treatment, country, time-updated CD4+ T-cell count, HIV ribonucleic acid (RNA), and prevalent tuberculosis. Results. Among 99 cases and 234 controls, median baseline CD4+ T-cell count was 181 cells/µL, and HIV RNA was 5.05 log10 cp/mL. Clinical failure was independently associated with continued elevations of IL-18 (IRR, 3.03; 95% confidence interval [CI], 1.27–7.20), sCD14 (IRR, 2.17; 95% CI, 1.02–4.62), and IFN-γ (IRR, 0.08; 95% CI, 0.01–0.61). Among 276 of 333 (83%) who were virologically suppressed at ART24, IFN-γ was associated with protection from failure, but the association with sCD14 was attenuated. Conclusions. Continued IL-18 and sCD14 elevations were associated with clinical ART failure. Interferon-γ levels may reflect preserved immune function.
Collapse
Affiliation(s)
- Ashwin Balagopal
- Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Nikhil Gupte
- Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Rupak Shivakoti
- Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Andrea L Cox
- Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Wei-Teng Yang
- Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Sima Berendes
- Malawi College of Medicine-Johns Hopkins University Research Project , Blantyre , Malawi
| | | | - Cecilia Kanyama
- University of North Carolina Project, Kamuzu Central Hospital , Lilongwe , Malawi
| | - Sandy Pillay
- Nelson Mandela School of Medicine , University of KwaZulu-Natal , Durban , South Africa
| | | | - Breno Santos
- Hospital Nossa Senhora de Conceição , Porto Alegre , Brazil
| | | | | | | | - Javier R Lama
- Asociación Civil Impacta Salud y Educación (IMPACTA) Peru Clinical Trials Unit , Lima
| | - Sandra W Cardoso
- Instituto de Pesquisa Clínica Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , Brazil
| | | | - Richard D Semba
- Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - James Hakim
- University of Zimbabwe, College of Health Sciences , Harare
| | - Mina C Hosseinipour
- University of North Carolina Project, Kamuzu Central Hospital, Lilongwe, Malawi; University of North Carolina, Chapel Hill
| | | | - Ian Sanne
- University of Witwatersrand , Johannesburg , South Africa
| | | | | | | | - Amita Gupta
- Johns Hopkins University School of Medicine , Baltimore, Maryland
| |
Collapse
|
26
|
Gama TGV, Pires FR, Armada L, Gonçalves LS. Cellular Profile and Expression of Immunologic Markers in Chronic Apical Periodontitis from HIV-infected Patients Undergoing Highly Active Antiretroviral Therapy. J Endod 2016; 42:921-7. [PMID: 27118467 DOI: 10.1016/j.joen.2016.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 03/07/2016] [Accepted: 03/13/2016] [Indexed: 02/08/2023]
Abstract
INTRODUCTION This study tested the hypothesis that the inflammatory cell profile (CD3-, CD4-, CD8-, CD20-, and CD68-positive cells) and the expression of immunologic markers (tumor necrosis factor α, interferon-γ, interleukin-6, and interleukin-18) in chronic apical periodontitis are the same between non-HIV-infected patients and HIV-infected patients undergoing highly active antiretroviral therapy (HAART). METHODS Thirty-four surgically excised chronic apical periodontitis lesions were sampled from 34 patients (17 HIV-infected and 17 non-HIV-infected). The lesions were extracted from teeth with no previous endodontic treatment. All HIV-infected patients were undergoing HAART. The specimens were submitted to histopathologic and immunohistochemical analyses by using an optical microscope. Immunoexpression was graded into 2 levels, focal to weak and moderate to strong. The χ(2), Fisher exact, and Mann-Whitney tests were used to analyze all significant differences between groups. RESULTS Periapical cysts represented 70.6% and 52.9% of the lesions in the HIV-infected and non-HIV-infected groups, respectively; however, no statistically significant difference was observed (P = .481). There were no statistically significant differences between groups for the inflammatory cell profile and for any of the immunologic markers (P > .05). CONCLUSIONS There are no statistically significant differences of the cellular profile and expression of immunologic markers in chronic apical periodontitis between non-HIV-infected patients and HIV-infected patients undergoing HAART.
Collapse
Affiliation(s)
- Túlio Gustavo Veiga Gama
- Post-graduation Program in Dentistry, Estácio de Sá University, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabio Ramoa Pires
- Post-graduation Program in Dentistry, Estácio de Sá University, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Armada
- Post-graduation Program in Dentistry, Estácio de Sá University, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucio Souza Gonçalves
- Post-graduation Program in Dentistry, Estácio de Sá University, Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
27
|
Samarani S, Allam O, Sagala P, Aldabah Z, Jenabian MA, Mehraj V, Tremblay C, Routy JP, Amre D, Ahmad A. Imbalanced production of IL-18 and its antagonist in human diseases, and its implications for HIV-1 infection. Cytokine 2016; 82:38-51. [PMID: 26898120 DOI: 10.1016/j.cyto.2016.01.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/12/2016] [Accepted: 01/15/2016] [Indexed: 12/16/2022]
Abstract
IL-18 is a pleiotropic and multifunctional cytokine that belongs to the IL-1 family. It is produced as a biologically inactive precursor, which is cleaved into its active mature form mainly by caspase-1. The caspase becomes active from its inactive precursor (procaspase-1) upon assembly of an inflammasome. Because of IL-18's potential pro-inflammatory and tissue destructive effects, its biological activities are tightly controlled in the body by its naturally occurring antagonist called IL-18BP. The antagonist is produced in the body both constitutively and in response to an increased production of IL-18 as a negative feedback mechanism. Under physiological conditions, most of IL-18 in the circulation is bound with IL-18BP and is inactive. However, an imbalance in the production of IL-18 and its antagonist (an increase in the production of IL-18 with a decrease, no increase or an insufficient increase in the production of IL-18BP) has been described in many chronic inflammatory diseases in humans. The imbalance results in an increase in the concentrations of free IL-18 (unbound with its antagonist) resulting in increased biological activities of the cytokine that contribute towards pathogenesis of the disease. In this article, we provide an overview of the current biology of IL-18 and its antagonist, discuss how the imbalance occurs in HIV infections and how it contributes towards development of AIDS and other non-AIDS-associated clinical conditions occurring in HIV-infected individuals undergoing combination anti-retroviral therapy (cART). Finally, we discuss challenges facing immunotherapeutic strategies aimed at restoring balance between IL-18 and its antagonist in these patients.
Collapse
Affiliation(s)
- Suzanne Samarani
- Laboratory of Innate Immunity, Canada; CHU-Sainte-Justine Research Center, Canada; Department of Microbiology, Infectiology & Immunology, Canada; University of Montreal, Montreal, Canada
| | - Ossama Allam
- Laboratory of Innate Immunity, Canada; CHU-Sainte-Justine Research Center, Canada; Department of Microbiology, Infectiology & Immunology, Canada; University of Montreal, Montreal, Canada
| | - Patrick Sagala
- Laboratory of Innate Immunity, Canada; CHU-Sainte-Justine Research Center, Canada; Department of Microbiology, Infectiology & Immunology, Canada; University of Montreal, Montreal, Canada
| | - Zainab Aldabah
- Laboratory of Innate Immunity, Canada; CHU-Sainte-Justine Research Center, Canada; Department of Microbiology, Infectiology & Immunology, Canada; University of Montreal, Montreal, Canada
| | | | - Vikram Mehraj
- McGill University Health Center, McGill University, Montreal, Canada
| | - Cécile Tremblay
- Department of Microbiology, Infectiology & Immunology, Canada; Division of Infectious Diseases, CHUM, Canada; University of Montreal, Montreal, Canada
| | - Jean-Pierre Routy
- McGill University Health Center, McGill University, Montreal, Canada
| | - Devendra Amre
- CHU-Sainte-Justine Research Center, Canada; Department of Pediatrics, Canada; University of Montreal, Montreal, Canada
| | - Ali Ahmad
- Laboratory of Innate Immunity, Canada; CHU-Sainte-Justine Research Center, Canada; Department of Microbiology, Infectiology & Immunology, Canada; University of Montreal, Montreal, Canada.
| |
Collapse
|
28
|
Fowler BJ, Gelfand BD, Kim Y, Kerur N, Tarallo V, Hirano Y, Amarnath S, Fowler DH, Radwan M, Young MT, Pittman K, Kubes P, Agarwal HK, Parang K, Hinton DR, Bastos-Carvalho A, Li S, Yasuma T, Mizutani T, Yasuma R, Wright C, Ambati J. Nucleoside reverse transcriptase inhibitors possess intrinsic anti-inflammatory activity. Science 2014; 346:1000-1003. [PMID: 25414314 PMCID: PMC4274127 DOI: 10.1126/science.1261754] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Nucleoside reverse transcriptase inhibitors (NRTIs) are mainstay therapeutics for HIV that block retrovirus replication. Alu (an endogenous retroelement that also requires reverse transcriptase for its life cycle)-derived RNAs activate P2X7 and the NLRP3 inflammasome to cause cell death of the retinal pigment epithelium in geographic atrophy, a type of age-related macular degeneration. We found that NRTIs inhibit P2X7-mediated NLRP3 inflammasome activation independent of reverse transcriptase inhibition. Multiple approved and clinically relevant NRTIs prevented caspase-1 activation, the effector of the NLRP3 inflammasome, induced by Alu RNA. NRTIs were efficacious in mouse models of geographic atrophy, choroidal neovascularization, graft-versus-host disease, and sterile liver inflammation. Our findings suggest that NRTIs are ripe for drug repurposing in P2X7-driven diseases.
Collapse
Affiliation(s)
- Benjamin J Fowler
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA. Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Bradley D Gelfand
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA. Department of Microbiology, Immunology, and Human Genetics, University of Kentucky, Lexington, KY 40536, USA. Department of Biomedical Engineering, University of Kentucky, Lexington, KY 40536, USA
| | - Younghee Kim
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Nagaraj Kerur
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Valeria Tarallo
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA. Angiogenesis Lab, Institute of Genetics and Biophysics, CNR, Naples, Italy
| | - Yoshio Hirano
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Shoba Amarnath
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniel H Fowler
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marta Radwan
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Mark T Young
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Keir Pittman
- Immunology Research Group, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Paul Kubes
- Immunology Research Group, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Hitesh K Agarwal
- Chapman University School of Pharmacy, 9401 Jeronimo Road, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
| | - Keykavous Parang
- Chapman University School of Pharmacy, 9401 Jeronimo Road, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
| | - David R Hinton
- Departments of Pathology and Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Ana Bastos-Carvalho
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Shengjian Li
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Tetsuhiro Yasuma
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Takeshi Mizutani
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Reo Yasuma
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Charles Wright
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Jayakrishna Ambati
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA. Department of Physiology, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
29
|
Fonville JM, Wilks SH, James SL, Fox A, Ventresca M, Aban M, Xue L, Jones TC, Le NMH, Pham QT, Tran ND, Wong Y, Mosterin A, Katzelnick LC, Labonte D, Le TT, van der Net G, Skepner E, Russell CA, Kaplan TD, Rimmelzwaan GF, Masurel N, de Jong JC, Palache A, Beyer WEP, Le QM, Nguyen TH, Wertheim HFL, Hurt AC, Osterhaus ADME, Barr IG, Fouchier RAM, Horby PW, Smith DJ. Antibody landscapes after influenza virus infection or vaccination. Science 2014; 346:996-1000. [PMID: 25414313 PMCID: PMC4246172 DOI: 10.1126/science.1256427] [Citation(s) in RCA: 358] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
We introduce the antibody landscape, a method for the quantitative analysis of antibody-mediated immunity to antigenically variable pathogens, achieved by accounting for antigenic variation among pathogen strains. We generated antibody landscapes to study immune profiles covering 43 years of influenza A/H3N2 virus evolution for 69 individuals monitored for infection over 6 years and for 225 individuals pre- and postvaccination. Upon infection and vaccination, titers increased broadly, including previously encountered viruses far beyond the extent of cross-reactivity observed after a primary infection. We explored implications for vaccination and found that the use of an antigenically advanced virus had the dual benefit of inducing antibodies against both advanced and previous antigenic clusters. These results indicate that preemptive vaccine updates may improve influenza vaccine efficacy in previously exposed individuals.
Collapse
Affiliation(s)
- J. M. Fonville
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - S. H. Wilks
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
| | - S. L. James
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
| | - A. Fox
- Oxford University Clinical Research Unit and Wellcome Trust Major Overseas Programme, Hanoi, Vietnam
| | - M. Ventresca
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - M. Aban
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL at the Peter Doherty Institute for Infection and Immunity, Melbourne VIC 3000, Australia
| | - L. Xue
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL at the Peter Doherty Institute for Infection and Immunity, Melbourne VIC 3000, Australia
| | - T. C. Jones
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
| | - N. M. H. Le
- Oxford University Clinical Research Unit and Wellcome Trust Major Overseas Programme, Hanoi, Vietnam
| | - Q. T. Pham
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - N. D. Tran
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - Y. Wong
- Oxford University Museum of Natural History, Oxford OX1 3PW, UK
| | - A. Mosterin
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
| | - L. C. Katzelnick
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
| | - D. Labonte
- Insect Biomechanics Group, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - T. T. Le
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - G. van der Net
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - E. Skepner
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
| | - C. A. Russell
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| | | | - G. F. Rimmelzwaan
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - N. Masurel
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - J. C. de Jong
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - A. Palache
- Abbott Laboratories, Weesp 1380 DA, the Netherlands
| | - W. E. P. Beyer
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - Q. M. Le
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - T. H. Nguyen
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - H. F. L. Wertheim
- Oxford University Clinical Research Unit and Wellcome Trust Major Overseas Programme, Hanoi, Vietnam
- Nuffield Department of Clinical Medicine, Centre for Tropical Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - A. C. Hurt
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL at the Peter Doherty Institute for Infection and Immunity, Melbourne VIC 3000, Australia
- Melbourne School of Population and Global Health, University of Melbourne, Parkville VIC 3010, Australia
| | - A. D. M. E. Osterhaus
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - I. G. Barr
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL at the Peter Doherty Institute for Infection and Immunity, Melbourne VIC 3000, Australia
| | - R. A. M. Fouchier
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - P. W. Horby
- Oxford University Clinical Research Unit and Wellcome Trust Major Overseas Programme, Hanoi, Vietnam
- Nuffield Department of Clinical Medicine, Centre for Tropical Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - D. J. Smith
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| |
Collapse
|
30
|
Roff SR, Noon-Song EN, Yamamoto JK. The Significance of Interferon-γ in HIV-1 Pathogenesis, Therapy, and Prophylaxis. Front Immunol 2014; 4:498. [PMID: 24454311 PMCID: PMC3888948 DOI: 10.3389/fimmu.2013.00498] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 12/17/2013] [Indexed: 12/24/2022] Open
Abstract
Interferon-γ (IFNγ) plays various roles in the pathogenesis of HIV/AIDS. In an HIV-1 infected individual, the production of IFNγ is detected as early as the acute phase and continually detected throughout the course of infection. Initially produced to clear the primary infection, IFNγ together with other inflammatory cytokines are involved in establishing a chronic immune activation that exacerbates clinical diseases associated with AIDS. Unlike Type 1 IFNs, IFNγ has no direct antiviral activity against HIV-1 in primary cultures, as supported by the in vivo findings of IFNγ therapy in infected subjects. Results from both in vitro and ex vivo studies show that IFNγ can instead enhance HIV-1 replication and its associated diseases, and therapies aimed at decreasing its production are under consideration. On the other hand, IFNγ has been shown to enhance cytotoxic T lymphocytes and NK cell activities against HIV-1 infected cells. These activities are important in controlling HIV-1 replication in an individual and will most likely play a role in the prophylaxis of an effective vaccine against HIV-1. Additionally, IFNγ has been used in combination with HIV-1 vaccine to augment antiviral immunity. Technological advancements have focused on using IFNγ as a biological marker to analyze the type(s) of immunity generated by candidate HIV vaccines and the levels of immunity restored by anti-retroviral drug therapies or novel immunotherapies. Hence, in addition to its valuable ancillary role as a biological marker for the development of effective HIV-1 prophylactic and therapeutic strategies, IFNγ has a vital role in promoting the pathogenesis of HIV.
Collapse
Affiliation(s)
- Shannon R. Roff
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Ezra N. Noon-Song
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Janet K. Yamamoto
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
31
|
Abstract
Several host genetic factors play an important role in susceptibility to human immunodeficiency virus type 1 (HIV-1) infection and in its progression to acquired immune deficiency syndrome (AIDS). The interleukin-18 (IL-18) is a multifunctional proinflammatory cytokine that regulates immune responses and plays a pathogenic role in HIV-1 infection by enhancing viral replication. Single nucleotide polymorphisms (SNPs) in the IL-18 gene promoter region may lead to altered transcriptional activity and IL-18 production, and may account for variation in the risk of HIV-1 infection. We have investigated the association between IL-18 promoter polymorphism −607C>A and HIV-1 infection through a case-control study of 500 patients with HIV-1/AIDS and an equal number of age and sex matched controls in a north Indian population. Genotyping using sequence specific primer-polymerase chain reaction (SSP-PCR) showed a statistically significant reduced risk of HIV-1 infection for the A>A genotype [odds ratio (OR) = 0.57, 95% confidence interval (95% CI) = 0.33–0.98, p = 0.040], but not for the C>A genotype (OR = 0.87, 95% CI = 0.66–1.14, p = 0.321). We concluded that the −607A allele of the IL-18 gene promoter polymorphism may play a protective role against the progression of HIV-1 infection in this population.
Collapse
|
32
|
Vandergeeten C, Fromentin R, Chomont N. The role of cytokines in the establishment, persistence and eradication of the HIV reservoir. Cytokine Growth Factor Rev 2012; 23:143-9. [PMID: 22743037 PMCID: PMC3767481 DOI: 10.1016/j.cytogfr.2012.05.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
HIV persists in cellular and anatomical reservoirs during Highly Active Antiretroviral Therapy (HAART). In vitro studies as well as in vivo observations have identified cytokines as important factors regulating the immunological and virological mechanisms involved in HIV persistence. Immunosuppressive cytokines might contribute to the establishment of viral latency by dampening T cell activation and HIV production, thereby creating the necessary immuno-virological condition for the establishment of a pool of latently infected cells. Other cytokines that are involved in the maintenance of memory CD4(+) T cells promote the persistence of these cells during HAART. Conversely, proinflammatory cytokines may favor HIV persistence by exacerbating low levels of ongoing viral replication in lymphoid tissues even after prolonged therapy. The ability of several cytokines to interfere with the molecular mechanisms responsible for HIV latency makes them attractive candidates for therapeutic strategies aimed at reducing the pool of latently infected cells. In this article, we review the role of cytokines in HIV persistence during HAART and discuss their role as potential eradicating agents.
Collapse
|
33
|
Watanabe D, Uehira T, Yonemoto H, Bando H, Ogawa Y, Yajima K, Taniguchi T, Kasai D, Nishida Y, Shirasaka T. Sustained high levels of serum interferon-γ during HIV-1 infection: a specific trend different from other cytokines. Viral Immunol 2011; 23:619-25. [PMID: 21142447 DOI: 10.1089/vim.2010.0065] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The expression levels of various cytokines increase with the progression of HIV-1 infection. However, the effects of antiretroviral therapy (ART) on serum cytokine levels have not been fully determined. In this study we measured serum cytokine levels of 35 HIV-1-infected Japanese adults. We first performed a cross-sectional study and observed that TNF-α, IL-6, IL-10, IL-18, and IL-7 levels all showed significant increases in those with advanced disease, and that this had a significant negative correlation with the CD4 cell count. However, IFN-γ levels did not show this relationship. A longitudinal study in 18 HIV-1-infected patients with a CD4 cell count <350/μL revealed that the introduction of ART reduced cytokine levels. Significant reductions of IL-7, IL-10, IFN-γ, and IL-18 levels were observed on days 30, 60, 90, and 90 after the initiation of ART, respectively. These results indicate a discrepancy between cross-sectional and longitudinal studies of serum levels of IFN-γ. To clarify this, we investigated serum IFN-γ levels in each patient. In 5 of the 15 patients IFN-γ levels did not decrease, even after ART initiation, and remained at 5 pg/mL or higher on day 120 after ART initiation. Higher IFN-γ levels (>5 pg/mL) were also observed in 2 of 7 asymptomatic patients, and 2 of 11 patients who underwent ART for 1 year or longer. These data demonstrate that IFN-γ levels in some patients increased and remained high even after the initiation of ART, which was a specific observation different from those of the other cytokines.
Collapse
Affiliation(s)
- Dai Watanabe
- AIDS Medical Center, Osaka National Hospital, National Hospital Organization, Osaka City, Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Yearley JH, Xia D, Pearson CB, Carville A, Shannon RP, Mansfield KG. Interleukin-18 predicts atherosclerosis progression in SIV-infected and uninfected rhesus monkeys (Macaca mulatta) on a high-fat/high-cholesterol diet. J Transl Med 2009; 89:657-67. [PMID: 19381133 PMCID: PMC2764242 DOI: 10.1038/labinvest.2009.29] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Interleukin (IL)-18 levels have been identified as important predictors of cardiovascular mortality and are often elevated in human immunodeficiency virus (HIV)-infected individuals. To investigate a possible function for IL-18 in atherogenesis in the context of early HIV infection, we used the simian immunodeficiency model of HIV infection. Acutely simian immunodeficiency virus-infected and uninfected rhesus monkeys (Macaca mulatta) on an atherogenic diet were evaluated prospectively for atherosclerotic lesion development relative to a panel of plasma markers including IL-18, IL-8, IL-1beta, IL-6, C-reactive protein, soluble vascular cell adhesion molecule-1, soluble E-selectin, and soluble intercellular adhesion molecule-1. Although no significant differences in lesion development were identified between groups after 35 days of infection, levels of plasma IL-18 measured 1 month before virus inoculation correlated significantly with atherosclerotic plaque cross-sectional area at the carotid bifurcation (P<0.001, R=0.946), common iliac bifurcation (P<0.01, R=0.789), and cranial abdominal aorta (P<0.01, R=0.747), as well as with extent of CD3+ and CD68+ cellular infiltration in vascular lesions (both P<0.001, R>or=0.835) in both groups. Atherosclerotic plaque area at the carotid and common iliac bifurcations also showed a weaker inverse correlation with baseline IL-8 levels, as did CD68+ signal area. Results implicate a strong role for IL-18 in early atherosclerosis progression and raise the possibility that the chronically elevated IL-18 levels seen in later stages of HIV infection may contribute significantly to accelerated atherogenesis in this population.
Collapse
Affiliation(s)
- Jennifer H Yearley
- Harvard Medical School, New England Primate Research Center, Southborough, MA 01772-9102, USA
| | | | | | | | | | | |
Collapse
|
35
|
Tornero C, Alberola J, Tamarit A, Navarro D. Effect of highly active anti-retroviral therapy and hepatitis C virus co-infection on serum levels of pro-inflammatory and immunoregulatory cytokines in human immunodeficiency virus-1-infected individuals. Clin Microbiol Infect 2007; 12:555-60. [PMID: 16700705 DOI: 10.1111/j.1469-0691.2006.01458.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This study aimed to determine the effect of highly active anti-retroviral therapy (HAART) and hepatitis C virus (HCV) co-infection on peripheral levels of interleukin (IL)-2, IL-10, IL-12 (p70), IL-18 and soluble tumour necrosis factor receptor type II (sTNFRII). Serum levels were monitored for a 1-year period in 25 patients infected with human immunodeficiency virus-1 (HIV-1) who were naive for HAART at the initiation of the study, and in four HIV-1-infected long-term non-progressors. Serum levels of both IL-18 and sTNFRII at baseline were significantly higher in HIV-1-infected patients than in controls. Baseline levels of IL-18 and sTNFRII were not significantly different in long-term non-progressors compared with the other patients. HCV co-infected patients had significantly higher levels of IL-18 and sTNFRII at each time-point compared with patients who were not co-infected with HCV. Irrespective of HCV status, response to HAART resulted in a significant decrease in the levels of both IL-18 and sTNFRII, particularly among patients who achieved HIV viral suppression, but the net decrease observed at the end of follow-up was lower in patients co-infected with HCV. No information was obtained from IL-2, IL-10 and IL-12 (p70) measurements. The data suggest that analysis of serum levels of IL-18 and sTNFRII may be a valuable tool for evaluating the response to HAART, and perhaps for assessing the degree of immune restoration achieved by HAART responders. The results also highlight the relevance of considering the HCV infection status of HIV-1-infected patients in order to avoid misinterpretation of IL-18 and sTNFRII measurements.
Collapse
Affiliation(s)
- C Tornero
- Department of Medicine, San Francisco de Borja Hospital, Gandía, Spain
| | | | | | | |
Collapse
|
36
|
Lama J, Planelles V. Host factors influencing susceptibility to HIV infection and AIDS progression. Retrovirology 2007; 4:52. [PMID: 17651505 PMCID: PMC1978541 DOI: 10.1186/1742-4690-4-52] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2007] [Accepted: 07/25/2007] [Indexed: 12/21/2022] Open
Abstract
Transmission of HIV first results in an acute infection, followed by an apparently asymptomatic period that averages ten years. In the absence of antiretroviral treatment, most patients progress into a generalized immune dysfunction that culminates in death. The length of the asymptomatic period varies, and in rare cases infected individuals never progress to AIDS. Other individuals whose behavioral traits put them at high-risk of HIV transmission, surprisingly appear resistant and never succumb to infection. These unique cases highlight the fact that susceptibility to HIV infection and progression to disease are complex traits modulated by environmental and genetic factors. Recent evidence has indicated that natural variations in host genes can influence the outcome of HIV infection and its transmission. In this review we summarize the available literature on the roles of cellular factors and their genetic variation in modulating HIV infection and disease progression.
Collapse
Affiliation(s)
- Juan Lama
- La Jolla Institute for Molecular Medicine, 4570 Executive Drive, Suite 100, San Diego, California 92121, USA
- RetroVirox, Inc. 4570 Executive Drive, Suite 100, San Diego, California 92121, USA
| | - Vicente Planelles
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East #2100 – Room 2520, Salt Lake City, Utah 84112, USA
| |
Collapse
|
37
|
Ahmad R, Iannello A, Samarani S, Morisset R, Toma E, Grosley M, Ahmad A. Contribution of platelet activation to plasma IL-18 concentrations in HIV-infected AIDS patients. AIDS 2006; 20:1907-9. [PMID: 16954739 DOI: 10.1097/01.aids.0000244217.46445.45] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
38
|
Segat L, Bevilacqua D, Boniotto M, Arraes LC, de Souza PR, de Lima Filho JL, Crovella S. IL-18 gene promoter polymorphism is involved in HIV-1 infection in a Brazilian pediatric population. Immunogenetics 2006; 58:471-3. [PMID: 16738942 DOI: 10.1007/s00251-006-0104-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2005] [Accepted: 02/14/2006] [Indexed: 12/31/2022]
Abstract
In our study, we identified a polymorphism (C-607A) in the promoter region of the IL-18 gene that shows different frequencies between human immunodeficiency virus (HIV)-1-infected children and healthy controls in a pediatric Brazilian population. The presence of the -607 C allele correlates to HIV-1 infection and confers an increased risk of infection in subjects carrying the single nucleotide polymorphism.
Collapse
Affiliation(s)
- Ludovica Segat
- Department of Reproductive and Developmental Sciences, University of Trieste, Via dell'Istria 65/1, Trieste, Italy.
| | | | | | | | | | | | | |
Collapse
|
39
|
Song W, Wilson CM, Allen S, Wang C, Li Y, Kaslow RA, Tang J. Interleukin 18 and human immunodeficiency virus type I infection in adolescents and adults. Clin Exp Immunol 2006; 144:117-24. [PMID: 16542373 PMCID: PMC1809636 DOI: 10.1111/j.1365-2249.2006.03050.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2006] [Indexed: 12/21/2022] Open
Abstract
Interleukin (IL)-18, a proinflammatory cytokine, has been recognized recently as an important factor in both treated and untreated patients with human immunodeficiency virus type 1 (HIV-1) infection. Consistent with all earlier reports, our quantification of serum IL-18 concentrations in 88 HIV-1 seropositive, North American adolescents (14-18 years old) revealed a positive correlation with cell-free HIV-1 viral load at two separate visits (Spearman's r = 0.31 and 0.50, respectively, P < 0.01 for both), along with a negative correlation with CD4+ T cell counts (r = -0.31 and -0.35, P < 0.01 for both). In additional analyses of 66 adults (21-58 years old) from Zambia, HIV-1 seroconversion was associated uniformly with elevated IL-18 production (P < 0.0001). These epidemiological relationships were independent of other population-related characteristics, including age, gender and ethnicity. In neither study population could serum IL-18 concentrations be associated with the IL-18 gene (IL18) promoter genotypes defined by five major single nucleotide polymorphisms. Collectively, these findings suggest that circulating IL-18 rather than the IL18 genotype may provide a useful biomarker for HIV-1-related events or outcomes.
Collapse
Affiliation(s)
- W Song
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35294-0022, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Couturier JP, Root-Bernstein RS. HIV may produce inhibitory microRNAs (miRNAs) that block production of CD28, CD4 and some interleukins. J Theor Biol 2005; 235:169-84. [PMID: 15862587 DOI: 10.1016/j.jtbi.2005.01.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2004] [Revised: 10/26/2004] [Accepted: 01/04/2005] [Indexed: 11/24/2022]
Abstract
It is well-known that HIV-1 infection results in a gradual decline of the CD4+ T-lymphocytes, but the underlying mechanism of this decline is not completely understood. Research has shown that HIV-1 infection of CD4+ T cells results in decreased CD28 expression, but the mechanism of this repression is unknown. There is also substantial evidence demonstrating regulatory involvement of microRNA (miRNA) during protein expression in plants and some animals, and reports have recently been published confirming the existence of viral-encoded miRNAs. Based on these findings, we hypothesize that viral-encoded miRNA from HIV-1 may directly alter T cell, macrophage and dendritic cell activity. To investigate a potential correlation between the genomic complementarity of HIV-1 and host cell protein expression, a local alignment search was performed to assess for regions of complementarity between the HIV-1 proviral genome and the mRNA coding sequence of various proteins expressed by CD+ T cells and macrophages. Regions of complementarity with strong correlations to the currently established criteria for miRNA:target mRNA activity were found between HIV-1 and CD28, CTLA-4 and some interleukins, suggesting that HIV-1 may produce translational repression in host cells.
Collapse
|
41
|
Wiercinska-Drapalo A, Jaroszewicz J, Flisiak R, Prokopowicz D. Plasma interleukin-18 is associated with viral load and disease progression in HIV-1-infected patients. Microbes Infect 2004; 6:1273-7. [PMID: 15555533 DOI: 10.1016/j.micinf.2004.07.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2004] [Accepted: 07/29/2004] [Indexed: 10/26/2022]
Abstract
Recent studies demonstrate persistent elevation of interleukin-18 (IL-18) concentration in human immunodeficiency virus type 1 (HIV-1)-infected patients. Due to pleiotropic action of IL-18 on the immune system, dysregulation of its synthesis may lead to inappropriate immune activation. The aim of this study was to determine possible correlation between IL-18 levels and the natural stages of HIV-1 infection. IL-18 plasma concentrations were determined in 42 patients in different stages of an HIV-1 infection and in 15 healthy controls. HIV infection resulted in a more than fourfold increase of plasma IL-18 concentration compared to healthy individuals (865 +/- 87 vs. 206 +/- 32 pg/ml, P < 0.001). Moreover, a positive correlation between plasma IL-18 concentration and HIV viral load was found (r = 0.44, P < 0.01). Further analysis showed marked elevation of IL-18 levels in late-stage symptomatic patients. Plasma IL-18 concentrations in patients receiving high-activity antiretroviral treatment (HAART) were significantly lower than in those not undergoing antiretroviral treatment. Individuals who did not reach viral suppression showed higher IL-18 plasma concentration than the group with achieved viral suppression. Excessive production of IL-18 observed in our study may promote viral replication and disease progression in advanced, especially late-stage HIV-infected patients. Furthermore, reduction of IL-18 concentration can be an important step in HAART-related immune restoration.
Collapse
Affiliation(s)
- Alicja Wiercinska-Drapalo
- Department of Infectious Diseases, Medical University of Bialystok, Zurawia 14 Street, 15-540 Bialystok, Poland.
| | | | | | | |
Collapse
|
42
|
Lindegaard B, Hansen ABE, Gerstoft J, Pedersen BK. High Plasma Level of Interleukin-18 in HIV-Infected Subjects With Lipodystrophy. J Acquir Immune Defic Syndr 2004; 36:588-93. [PMID: 15097301 DOI: 10.1097/00126334-200405010-00006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The level of interleukin-18 (IL-18) is elevated in patients with HIV infection as well as in people with insulin resistance (IR). As HIV-associated lipodystrophy (LD) shares metabolic characteristics with the metabolic syndrome, it was hypothesized that IL-18 would be elevated in patients with LD. Two groups of HIV-infected men with LD, one with fat accumulation (mixed group) (n = 12) and one without fat accumulation (lipoatrophic group) (n = 15) were included. Controls were HIV-positive men without LD (n = 15) and HIV-negative, age-matched men (n = 12). The levels of plasma IL-18 were elevated in all 3 HIV groups compared with HIV-negative controls (P <0.01). In the HIV groups the lipoatrophic group had the highest IL-18, followed by the mixed group and the HIV-positive controls. Only the differences between the lipoatrophic group and the HIV-positive controls were significant (P <0.01). Plasma IL-18 correlated with tumor necrosis factor-alpha (P <0.05), but not IL-6, adiponectin, or HOMA-IR (homeostasis model of insulin resistance). In contrast to the HIV-negative controls, IL-18 did not correlate with total or low-density cholesterol in either of the HIV groups. An inverse correlation was observed between IL-18 and limb fat (P <0.05). In conclusion, the level of IL-18 is elevated in patients with LD and closely linked to limb atrophy, whereas it is not associated with cholesterol or IR.
Collapse
Affiliation(s)
- Birgitte Lindegaard
- Department of Infectious Diseases, Rigshospitalet, University of Copenhagen, Denmark.
| | | | | | | |
Collapse
|
43
|
Alexandrakis MG, Passam FH, Sfiridaki K, Moschandrea J, Pappa C, Liapi D, Petreli E, Roussou P, Kyriakou DS. Interleukin-18 in multiple myeloma patients: serum levels in relation to response to treatment and survival. Leuk Res 2004; 28:259-66. [PMID: 14687621 DOI: 10.1016/s0145-2126(03)00261-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Interleukin-18 (IL-18) plays a role in the host's response to tumours and angiogenesis. We determined serum levels of IL-18, vascular endothelial growth factor (VEGF), angiogenin (ANG), tumor necrosis factor (TNF-alpha) and CRP in 65 newly diagnosed myeloma patients. IL-18, VEGF, angiogenin, TNF-alpha and CRP were significantly higher at stage III in comparison to stages II and I. These cytokines (measured in 27 patients) significantly decreased after treatment. In survival analysis, higher levels of IL-18 were associated with a poorer prognosis. We conclude that increased serum IL-18 in myeloma patients correlates with advanced disease, increased levels of angiogenic cytokines and worse survival.
Collapse
Affiliation(s)
- M G Alexandrakis
- Division of Medicine, University Hospital of Heraklion, Medical School of Crete, PO Box 1352, Crete, Greece.
| | | | | | | | | | | | | | | | | |
Collapse
|